Secreted proteins

Abstract
The invention provides human secreted proteins (SECP) and polynucleotides which identify and encode SECP. The invention also provides expression vectors, host cells, antibodies, agonists, and antagonists. The invention also provides methods for diagnosing, treating, or preventing disorders associated with aberrant expression of SECP.
Description
TECHNICAL FIELD

This invention relates to nucleic acid and amino acid sequences of secreted proteins and to the use of these sequences in the diagnosis, treatment, and prevention of cell proliferative, autoimmune/inflammatory, cardiovascular, neurological, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of secreted proteins.


BACKGROUND OF THE INVENTION

Protein transport and secretion are essential for cellular function. Protein transport is mediated by a signal peptide located at the amino terminus of the protein to be transported or secreted. The signal peptide is comprised of about ten to twenty hydrophobic amino acids which target the nascent protein from the ribosome to a particular membrane bound compartment such as the endoplasmic reticulum (ER). Proteins targeted to the ER may either proceed through the secretory pathway or remain in any of the secretory organelles such as the ER, Golgi apparatus, or lysosomes. Proteins that transit through the secretory pathway are either secreted into the extracellular space or retained in the plasma membrane. Proteins that are retained in the plasma membrane contain one or more transmembrane domains, each comprised of about 20 hydrophobic amino acid residues. Secreted proteins are generally synthesized as inactive precursors that are activated by post-translational processing events during transit through the secretory pathway. Such events include glycosylation, proteolysis, and removal of the signal peptide by a signal peptidase. Other events that may occur during protein transport include chaperone-dependent unfolding and folding of the nascent protein and interaction of the protein with a receptor or pore complex. Examples of secreted proteins with amino terminal signal peptides are discussed below and include proteins with important roles in cell-to-cell signaling. Such proteins include transmembrane receptors and cell surface markers, extracellular matrix molecules, cytokines, hormones, growth and differentiation factors, enzymes, neuropeptides, vasomediators, cell surface markers, and antigen recognition molecules. (Reviewed in Alberts, B. et al. (1994) Molecular Biology of The Cell, Garland Publishing, New York, N.Y., pp. 557-560, 582-592.)


Cell surface markers include cell surface antigens identified on leukocytic cells of the immune system. These antigens have been identified using systematic, monoclonal antibody (mAb)-based “shot gun” techniques. These techniques have resulted in the production of hundreds of mAbs directed against unknown cell surface leukocytic antigens. These antigens have been grouped into “clusters of differentiation” based on common immunocytochemical localization patterns in various differentiated and undifferentiated leukocytic cell types. Antigens in a given cluster are presumed to identify a single cell surface protein and are assigned a “cluster of differentiation” or “CD” designation. Some of the genes encoding proteins identified by CD antigens have been cloned and verified by standard molecular biology techniques. CD antigens have been characterized as both transmembrane proteins and cell surface proteins anchored to the plasma membrane via covalent attachment to fatty acid-containing glycolipids such as glycosyiphosphatidylinositol (GPI). (Reviewed in Barclay, A. N. et al. (1995) The Leucocyte Antigen Facts Book, Academic Press, San Diego, Calif., pp. 17-20.)


Matrix proteins (MPs) are transmembrane and extracellular proteins which function in formation, growth, remodeling, and maintenance of tissues and as important mediators and regulators of the inflammatory response. The expression and balance of MPs may be perturbed by biochemical changes that result from congenital, epigenetic, or infectious diseases. In addition, MPs affect leukocyte migration, proliferation, differentiation, and activation in the immune response. MPs are frequently characterized by the presence of one or more domains which may include collagen-like domains, EGF-like domains, immunoglobulin-like domains, and fibronectin-like domains. In addition, MPs may be heavily glycosylated and may contain an Arginine-Glycine-Aspartate (RGD) tripeptide motif which may play a role in adhesive interactions. MPs include extracellular proteins such as fibronectin, collagen, galectin, vitronectin and its proteolytic derivative somatomedin B; and cell adhesion receptors such as cell adhesion molecules (CAMs), cadherins, and integrins. (Reviewed in Ayad, S. et al. (1994) The Extracellular Matrix Facts Book, Academic Press, San Diego, Calif., pp. 2-16; Ruoslahti, E. (1997) Kidney Ita 51:1413-1417; Sjaastad, M. D. and Nelson, W. J. (1997) BioEssays 19:47-55.)


Mucins are highly glycosylated glycoproteins that are the major structural component of the mucus gel. The physiological functions of mucins are cytoprotection, mechanical protection, maintenance of viscosity in secretions, and cellular recognition. MUC6 is a human gastric mucin that is also found in gall bladder, pancreas, seminal vesicles, and female reproductive tract (Toribara, N. W. et al. (1997) J. Biol. Chem. 272:16398-16403). The MUC6 gene has been mapped to human chromosome 11 (Toribara, N. W. et al. (1993) J. Biol. Chem. 268:5879-5885). Hemomucin is a novel Drosophila surface mucin that may be involved in the induction of antibacterial effector molecules (Theopold, U. et al. (1996) J. Biol. Chef 217:12708-12715).


Tuftelins are one of four different enamel matrix proteins that have been identified so far. The other three known enamel matrix proteins are the amelogenins, enamelin and ameloblastin. Assembly of the enamel extracellular matrix from these component proteins is believed to be critical in producing a matrix competent to undergo mineral replacement. (Paine, C. T. et al. (1998) Connect Tissue Res. 38:257-267). Tuftelin mRNA has been found to be expressed in human ameloblastoma tumor, a non-mineralized odontogenic tumor (Deutsch, D. et al. (1998) Connect. Tissue Res. 39:177-184).


Olfactomedin-related proteins are extracellular matrix, secreted glycoproteins with conserved C-terminal motifs. They are expressed in a wide variety of tissues and in broad range of species, from Caenorhabditis elegans to Homo sapiens. Olfactomedin-related proteins comprise a gene family with at least 5 family members in humans. One of the five, TIGR/myocilin protein, is expressed in the eye and is associated with the pathogenesis of glaucoma (Kulkarmi, N. H. et al. (2000) Genet. Res. 76:41-50). Research by Yokoyama et al. (1996) found a 135-amino acid protein, termed AMY, having 96% sequence identity with rat neuronal olfactomedin-releated ER localized protein in a neuroblastoma cell line cDNA library, suggesting an essential role for AMY in nerve tissue (Yokoyama, M. et al. (1996) DNA Res. 3:311-320). Neuron-specific olfactomedin-related glycoproteins isolated from rat brain cDNA libraries show strong sequence similarity with olfactomedin. This similarity is suggestive of a matrix-related function of these glycoproteins in neurons and neurosecretory cells (Danielson, P. E. et al. (1994) J. Neurosci. Res. 38:468-478).


Mac-2 binding protein is a 90-kD serum protein (90K), a secreted glycoprotein isolated from both the human breast carcinoma cell line SK-BR-3, and human breast milk. It specifically binds to a human macrophage-associated lectin, Mac-2. Structurally, the mature protein is 567 amino acids in length and is proceeded by an 18-amino acid leader. There are 16 cysteines and seven potential N-linked glycosylation sites. The first 106 amino acids represent a domain very similar to an ancient protein superfamily defined by a macrophage scavenger receptor cysteine-rich domain (Koths, K. et al. (1993) J. Biol. Chem. 268:14245-14249). 90K is elevated in the serum of subpopulations of AIDS patients and is expressed at varying levels in prima tumor samples and tumor cell lines. Ullrich et al. (1994) have demonstrated that 90K stimulates host defense systems and can induce interleukin-2 secretion. This immune stimulation is proposed to be a result of oncogenic transformation, viral infection or pathogenic invasion (Ullrich, A. et al. (1994) J. Biol. Chem. 269:18401-18407).


Semaphorins are a large group of axonal guidance molecules consisting of at least 30 different members and are found in vertebrates, invertebrates, and even certain viruses. All semaphorins contain the sema domain which is approximately 500 amino acids in length. Neuropilin, a semaphorin receptor, has been shown to promote neurite outgrowth in vitro. The extracellular region of neuropilins consists of three different domains: CUB, discoidin, and MAM domains. The CUB and the MAM motifs of neuropilin have been suggested to have roles in protein-protein interactions and are thought to be involved in the binding of semaphorins through the sema and the C-terminal domains (reviewed in Raper, J. A. (2000) Curr. Opin. Neurobiol. 10:88-94). Plexins are neuronal cell surface molecules that mediate cell adhesion via a homophilic binding mechanism in the presence of calcium ions. Plexins have been shown to be expressed in the receptors and neurons of particular sensory systems (Ohta, K. et al. (1995) Cell 14:1189-1199). There is evidence that suggests that some plexins function to control motor and CNS axon guidance in the developing nervous system. Plexins, which themselves contain complete semaphorin domains, may be both the ancestors of classical semaphorins and binding partners for semnaphorins (Winberg, M. L. et al (1998) Cell 95:903-916).


Human pregnancy-specific beta 1-glycoprotein (trSG) is a family of closely related glycoproteins of molecular weights of 72 KDa, 64 KDa, 62 KDa, and 54 KDa. Together with the carcinoembryonic antigen, they comprise a subfamily within the immunoglobulin superfamily (Plouzek, C. A. and Chou, J. Y. (1991) Endocrinology 129:950-958) Different subpopulations of PSG have been found to be produced by the trophoblasts of the human placenta, and the amnionic and chorionic membranes (Plouzek, C. A. et al. (1993) Placenta 14:277-285).


Autocrine motility factor (AMP) is one of the motility cytokines regulating tumor cell migration; therefore identification of the signaling pathway coupled with it has critical importance. Autocrine motility factor receptor (AMFR) expression has been found to be associated with tumor progression in thymoma (Ohta Y. et al. (2000) Int. J. Oncol. 17:259-264). AMFR is a cell surface glycoprotein of molecular weight 78 KDa.


Hormones are signaling molecules that coordinately regulate basic physiological processes from embryogenesis throughout adulthood. These processes include metabolism, respiration, reproduction, excretion, fetal tissue differentiation and organogenesis, growth and development, homeostasis, and the stress response. Hormonal secretions and the nervous system are tightly integrated and interdependent. Hormones are secreted by endocrine glands, primarily the hypothalamus and pituitary, the thyroid and parathyroid, the pancreas, the adrenal glands, and the ovaries and testes.


The secretion of hormones into the circulation is tightly controlled. Hormones are often secreted in diurnal, pulsatile, and cyclic patterns. Hormone secretion is regulated by perturbations in blood biochemistry, by other upstream-acting hormones, by neural impulses, and by negative feedback loops. Blood hormone concentrations are constantly monitored and adjusted to maintain optimal, steady-state levels. Once secreted, hormones act only on those target cells that express specific receptors.


Most disorders of the endocrine system are caused by either hyposecretion or hypersecretion of hormones. Hyposecretion often occurs when a hormone's gland of origin is damaged or otherwise impaired. Hypersecretion often results from the proliferation of tumors derived from hormone-secreting cells. Inappropriate hormone levels may also be caused by defects in regulatory feedback loops or in the processing of hormone precursors. Endocrine malfunction may also occur when the target cell fails to respond to the hormone.


Hormones can be classified biochemically as polypeptides, steroids, eicosanoids, or amines. Polypeptide hormones, which include diverse hormones such as insulin and growth hormone, vary in size and function and are often synthesized as inactive precursors that are processed intracellularly into mature, active forms. Amine hormones, which include epinephrine and dopamine, are amino acid derivatives that function in neuroendocrine signaling. Steroid hormones, which include the cholesterol-derived hormones estrogen and testosterone, function in sexual development and reproduction. Eicosanoid hormones, which include prostaglandins and prostacyclins, are fatty acid derivatives that function in a variety of processes. Most polypeptide hormones and some amine hormones are soluble in the circulation where they are highly susceptible to proteolytic degradation within seconds after their secretion. Steroid hormones and eicosanoid hormones are insoluble and must be transported in the circulation by carrier proteins. The following discussion will focus primarily on polypeptide hormones.


Hormones secreted by the hypothalamus and pituitary gland play a critical role in endocrine function by coordinately regulating hormonal secretions from other endocrine glands in response to neural signals. Hypothalamic hormones include thyrotropin-releasing hormone, gonadotropin-releasing hormone, somatostatin, growth-hormone releasing factor, corticotropin-releasing hormone, substance P, dopamine, and prolactin-releasing hormone. These hormones directly regulate the secretion of hormones from the anterior lobe of the pituitary. Hormones secreted by the anterior pituitary include adrenocorticotropic hormone (ACH), melanocyte-stimulating hormone, somatotropic hormones such as growth hormone and prolactin, glycoprotein hormones such as thyroid-stimulating hormone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH), β-lipotropin, and β-endorphins. These hormones regulate hormonal secretions from the thyroid, pancreas, and adrenal glands, and act directly on the reproductive organs to stimulate ovulation and spermatogenesis. The posterior pituitary synthesizes and secretes antidiuretic hormone (ADH, vasopressin) and oxytocin.


Disorders of the hypothalamus and pituitary often result from lesions such as primary brain tumors, adenomas, infarction associated with pregnancy, hypophysectomy, aneurysms, vascular malformations, thrombosis, infections, immunological disorders, and complications due to head trauma. Such disorders have profound effects on the function of other endocrine glands. Disorders associated with hypopituitarism include hypogonadism, Sheehan syndrome, diabetes insipidus, Kallman's disease, Hand-Schuller-Christian disease, Letterer-Siwe disease, sarcoidosis, empty sella syndrome, and dwarfism. Disorders associated with hyperpituitarism include acromegaly, giantism, and syndrome of inappropriate ADH secretion (SIADH), often caused by benign adenomas.


Hormones secreted by the thyroid and parathyroid primarily control metabolic rates and the regulation of serum calcium levels, respectively. Thyroid hormones include calcitonin, somatostatin, and thyroid hormone. The parathyroid secretes parathyroid hormone. Disorders associated with hypothyroidism include goiter, myxedema, acute thyroiditis associated with bacterial infection, subacute thyroiditis associated with viral infection, autoimmune thyroiditis (Hashimoto's disease), and cretinism. Disorders associated with hyperthyroidism include thyrotoxicosis and its various forms, Grave's disease, pretibial myxedema, toxic multinodular goiter, thyroid carcinoma, and Plummer's disease. Disorders associated with hyperparathyroidism include Conn disease (chronic hypercalemia) leading to bone resorption and parathyroid hyperplasia.


Hormones secreted by the pancreas regulate blood glucose levels by modulating the rates of carbohydrate, fat, and protein metabolism. Pancreatic hormones include insulin, glucagon, amylin, γ-aminobutyric acid, gastrin, somatostatin, and pancreatic polypeptide. The principal disorder associated with pancreatic dysfunction is diabetes mellitus caused by insufficient insulin activity. Diabetes mellitus is generally classified as either Type I (insulin-dependent, juvenile diabetes) or Type II (non-insulin-dependent, adult diabetes). The treatment of both forms by insulin replacement therapy is well known. Diabetes mellitus often leads to acute complications such as hypoglycemia (insulin shock), coma, diabetic ketoacidosis, lactic acidosis, and chronic complications leading to disorders of the eye, kidney, skin, bone, joint, cardiovascular system, nervous system, and to decreased resistance to infection.


The anatomy, physiology, and diseases related to hormonal function are reviewed in McCance, K. L. and Huether, S. E. (1994) Pathophysiology: The Biological Basis for Disease in Adults and Children, Mosby-Year Book, Inc., St. Louis, Mo.; Greenspan, F. S. and Baxter, J. D. (1994) Basic and Clinical Endocrinology, Appleton and Lange, East Norwalk, Conn.


Growth factors are secreted proteins that mediate intercellular communication. Unlike hormones, which travel great distances via the circulatory system, most growth factors are primarily local mediators that act on neighboring cells. Most growth factors contain a hydrophobic N-terminal signal peptide sequence which directs the growth factor into the secretory pathway. Most growth factors also undergo post-translational modifications within the secretory pathway. These modifications can include proteolysis, glycosylation, phosphorylation, and intramolecular disulfide bond formation. Once secreted, growth factors bind to specific receptors on the surfaces of neighboring target cells, and the bound receptors trigger intracellular signal transduction pathways. These signal transduction pathways elicit specific cellular responses in the target cells. These responses can include the modulation of gene expression and the stimulation or inhibition of cell division, cell differentiation, and cell motility.


Growth factors fall into at least two broad and overlapping classes. The broadest class includes the large polypeptide growth factors, which are wide-ranging in their effects. These factors include epidermal growth factor (LGF), fibroblast growth factor (FGF), transforming growth factor-β (TGF-β), insulin-like growth factor (IGF), nerve growth factor (NGF), and platelet-derived growth factor (PDGF), each defining a family of numerous related factors. The large polypeptide growth factors, with the exception of NGF, act as mitogens on diverse cell types to stimulate wound healing, bone synthesis and remodeling, extracellular matrix synthesis, and proliferation of epithelial, epidermal, and connective tissues. Members of the TGF-β, EGF, and FGF families also function as inductive signals in the differentiation of embryonic tissue. NGF functions specifically as a neurotrophic factor, promoting neuronal growth and differentiation.


Another class of growth factors includes the hematopoietic growth factors, which are narrow in their target specificity. These factors stimulate the proliferation and differentiation of blood cells such as B-lymphocytes, T-lymphocytes, erythrocytes, platelets, eosinophils, basophils, neutrophils, macrophages, and their stem cell precursors. These factors include the colony-stimulating factors (G-CSF, M-CSF, GM-CSF, and CSF1-3), erythropoietin, and the cytokines. The cytolines are specialized hematopoietic factors secreted by cells of the immune system and are discussed in detail below.


Hormones travel through the circulation and bind to specific receptors on the surface of, or within, target cells. Although they have diverse biochemical compositions and mechanisms of action, hormones can be grouped into two categories. One category includes small lipophilic hormones that diffuse through the plasma membrane of target cells, bind to cytosolic or nuclear receptors, and form a complex that alters gene expression. Examples of these molecules include retinoic acid, thyroxine, and the cholesterol-derived steroid hormones such as progesterone, estrogen, testosterone, cortisol, and aldosterone. The second category includes hydrophilic hormones that function by binding to cell surface receptors that transduce signals across the plasma membrane. Examples of such hormones include amino acid derivatives such as catecholamines (epinephrine, norepinephrine) and histamine, and peptide hormones such as glucagon, insulin, gastrin, secretin, cholecystokinin, adrenocorticotropic hormone, follicle stimulating hormone, luteinizing hormone, thyroid stimulating hormone, and vasopressin. (See, for example, Lodish et al. (1995) Molecular Cell Biology, Scientific American Books Inc., New York, N.Y., pp. 856-864.)


Pro-opiomelanocortin (POMC) is the precursor polypeptide of corticotropin (ACTH), a hormone synthesized by the anterior pituitary gland, which functions in the stimulation of the adrenal cortex. POMC is also the precursor polypeptide of the hormone beta-lipotropin (beta-LPH). Each hormone includes smaller peptides with distinct biological activities: alpha-melanotropin (alpha-MSH) and corticotropin-like intermediate lobe peptide (CLIP) are formed from ACTH; gamma-lipotropin (gamma-LPH) and beta-endorphin are peptide components of beta-LPH; while beta-MSH is contained within gamma-LPH. Adrenal insufficiency due to ACTH deficiency, resulting from a genetic mutation in exons 2 and 3 of POMC results in an endocrine disorder characterized by early-onset obesity, adrenal insufficiency, and red hair pigmentation (Chretien, M. et al. (1979) Can. J. Biochem. 57:1111-1121; Krude, H. et al. (1998) Nat. Genet. 19:155-157; Online Mendelian Inheritance in Man (OMIM) 176830).


Growth and differentiation factors are secreted proteins which function in intercellular communication. Some factors require oligomerization or association with membrane proteins for activity. Complex interactions among these factors and their receptors trigger intracellular signal transduction pathways that stimulate or inhibit cell division, cell differentiation, cell signaling, and cell motility. Most growth and differentiation factors act on cells in their local environment (paracrine signaling). There are three broad classes of growth and differentiation factors. The first class includes the large polypeptide growth factors such as epidermal growth factor (EGF), fibroblast growth factor, transforming growth factor, insulin-like growth factor (IGF), and platelet-derived growth factor. EGF includes a 30-40 residue EGF repeat domain, composed of conserved cysteine and glycine residues, found in a variety of proteins involved in cell proliferation, including the leukocyte antigen CD97 and the Notch family proteins (Greener, M. (2000) Mol. Med. Today 6:139-140). IGF forms a heterotrimeric complex with IGF-binding-protein 3 and the acid-labile subunit (ALS). ALS is largely composed of 18-20 leucine-rich repeats of 24 amino acids (Leong, S. R. et al. (1992) Mol. Endocrinol. 6:870-876). The second class includes the hematopoietic growth factors such as the colony stimulating factors (CSFs). Hematopoietic growth factors stimulate the proliferation and differentiation of blood cells such as B-lymphocytes, T-lymphocytes, erythrocytes, platelets, eosinophils, basophils, neutrophils, macrophages, and their stem cell precursors. The third class includes small peptide factors such as bombesin, vasopressin, oxytocin, endothelin, transferrin, angiotensin II, vasoactive intestinal peptide, and bradykinin, which function as hormones to regulate cellular functions other than proliferation.


Growth and differentiation factors play critical roles in neoplastic transformation of cells in vitro and in tumor progression in vivo. Inappropriate expression of growth factors by tumor cells may contribute to vascularization and metastasis of tumors. During hematopoiesis, growth factor misregulation can result in anemias, leukemias, and lymphomas. Certain growth factors such as interferon are cytotoxic to tumor cells both in vivo and in vitro. Moreover, some growth factors and growth factor receptors are related both structurally and functionally to oncoproteins. In addition, growth factors affect transcriptional regulation of both proto-oncogenes and oncosuppressor genes. (Reviewed in Pimentel, E. (1994) Handbook of Growth Factors, CRC Press, Ann Arbor, Mich., pp. 1-9.)


In addition, some of the large polypeptide growth factors play crucial roles in the induction of the primordial germ layers in the developing embryo. This induction ultimately results in the formation of the embryonic mesoderm, ectoderm, and endoderm which in turn provide the framework for the entire adult body plan. Disruption of this inductive process would be catastrophic to embryonic development. One such growth factor, wnt, is a secreted glycoprotein that has activity as both a short-range inducer and as a long-range morphogen (for a review, see Howes, R. and S. Bray (2000) Current Biology 10:R222-R226). Wnt signaling is implicated in diseases including cancer and Alzheimer's Disease (Bienz, M. and H. Clevers (2000) Cell 103:311-320; Polakis, P. (2000) Genes Dev. 14:1837-1851; De Ferrari, G. V. and N. C. Inestrosa (2000) Brain Res. Brain. Res. Rev. 33:1-12). Chordin is a developmental protein that binds to ventralizing TGP-beta-like bone morphogenetic proteins (BMPs) and sequesters them in latent complexes, causing dorsalization of tissue (Pappano, W. N. et al., (1998) Genomics 52:236-239). Other developmental proteins that regulate BMPs include noggin, cerberus, dan, and gremlin (Schmitt, J. M. et al., (1999) J. Orthop. Res. 17:269-278).


The Slit protein, first identified in Drosophila, is critical in central nervous system midline formation and potentially in nervous tissue histogenesis and axonal pathfinding. Itoh et al. ((1998) Brain Res. Mol. Brain Res. 62:175-186) have identified mammalian homologues of the slit gene (human Slit-1, Slit-2, Slit-3 and rat Slit-1). The encoded proteins are putative secreted proteins containing EGF-like motifs and leucine-rich repeats, both of which are conserved protein-protein interaction domains. Slit-1, -2, and -3 mRNAs are expressed in the brain, spinal cord, and thyroid, respectively (Itoh, A. et al., supra). The Slit family of proteins are indicated to be functional ligands of glypican-1 in nervous tissue and it is suggested that their interactions may be critical in certain stages during central nervous system histogenesis (Liang, Y. et al. (1999) J. Biol. Chem. 274:17885-17892).


Neuropeptides and vasomediators (NP/VM) comprise a large family of endogenous signaling molecules. Included in this family are neuropeptides and neuropeptide hormones such as bombesin, neuropeptide Y, neurotensin, neuromedin N, melanocortins, opioids, galanin, somatostatin, tachykinins, urotensin II and related peptides involved in smooth muscle stimulation, vasopressin, vasoactive intestinal peptide, and circulatory system-borne signaling molecules such as angiotensin, complement, calcitonin, endothelins, formyl-methionyl peptides, glucagon, cholecystokinin and gastrin. NP/VMs can transduce signals directly, modulate the activity or release of other neurotransmitters and hormones, and act as catalytic enzymes in cascades. The effects of NP/VMs range from extremely brief to long-lasting. (Reviewed in Martin, C. R. et al. (1985) Endocrine Physiology, Oxford University Press, New York, N.Y., pp. 5762.)


NP/VMs are involved in numerous neurological and cardiovascular disorders. For example, neuropeptide Y is involved in hypertension, congestive heart failure, affective disorders, and appetite regulation. Somatostatin inhibits secretion of growth hormone and prolactin in the anterior pituitary, as well as inhibiting secretion in intestine, pancreatic acinar cells, and pancreatic beta-cells. A reduction in somatostatin levels has been reported in Alzheimer's disease and Parkinson's disease. Vasopressin acts in the kidney to increase water and sodium absorption, and in higher concentrations stimulates contraction of vascular smooth muscle, platelet activation, and glycogen breakdown in the liver. Vasopressin and its analogues are used clinically to treat diabetes insipidus. Endothelin and angiotensin are involved in hypertension, and drugs, such as captopril, which reduce plasma levels of angiotensin, are used to reduce blood pressure (Watson, S. and S. Arkinstall (1994) The G-protein Linked Receptor Facts Book, Academic Press, San Diego Calif., pp. 194; 252; 284; 55; 111).


Neuropeptides have also been shown to have roles in nociception (pain). Vasoactive intestinal peptide appears to play an important role in chronic neuropathic pain. Nociceptin, an endogenous ligand for for the opioid receptor-like 1 receptor, is thought to have a predominantly anti-nociceptive effect, and has been shown to have analgesic properties in different animal models of tonic or chronic pain (Dickinson, T. and Fleetwood-Walker, S. M. (1998) Trends Pharmacol. Sci. 19:346-348).


Cytokines comprise a family of signaling molecules that modulate the immune system and the inflammatory response. Cytokines are usually secreted by leukocytes, or white blood cells, in response to injury or infection. Cytokines function as growth and differentiation factors that act primarily on cells of the immune system such as B- and T-lymphocytes, monocytes, macrophages, and granulocytes. Like other signaling molecules, cytokines bind to specific plasma membrane receptors and trigger intracellular signal transduction pathways which alter gene expression patterns. There is considerable potential for the use of cytokines in the treatment of inflammation and immune system disorders.


Cytokine structure and function have been extensively characterized in vitro. Most cytokines are small polypeptides of about 30 kilodaltons or less. Over 50 cytokines have been identified from human and rodent sources. Examples of cytokine subfamilies include the interferons (IFN-α, -β, and -γ), the interleukins (IL1-IL13), the tumor necrosis factors (TNF-α and -β), and the chemokines. Many cytokines have been produced using recombinant DNA techniques, and the activities of individual cytokines have been determined in vitro. These activities include regulation of leukocyte proliferation, differentiation, and motility.


The activity of an individual cytokine in vitro may not reflect the full scope of that cytokine's activity in vivo. Cytokines are not expressed individually in vivo but are instead expressed in combination with a multitude of other cytokines when the organism is challenged with a stimulus. Together, these cytokines collectively modulate the immune response in a manner appropriate for that particular stimulus. Therefore, the physiological activity of a cytokine is determined by the stimulus itself and by complex interactive networks among co-expressed cytokines which may demonstrate both synergistic and antagonistic relationships.


Chemokines comprise a cytokine subfamily with over 30 members. (Reviewed in Wells, T. N. C. and Peitsch, M. C. (1997) J. Leukoc. Biol. 61:545-550.) Chemokines were initially identified as chemotactic proteins that recruit monocytes and macrophages to sites of inflammation. Recent evidence indicates that chemokines may also play key roles in hematopoiesis and HIV-1 infection. Chemokines are small proteins which range from about 6-15 kilodaltons in molecular weight. Chemokines are further classified as C, CC, CXC, or CX3C based on the number and position of critical cysteine residues. The CC chemokines, for example, each contain a conserved motif consisting of two consecutive cysteines followed by two additional cysteines which occur downstream at 24- and 16-residue intervals, respectively (ExPASy PROSITE database, documents PS00472 and PDOC00434). The presence and spacing of these four cysteine residues are highly conserved, whereas the intervening residues diverge significantly. However, a conserved tyrosine located about 15 residues downstream of the cysteine doublet seems to be important for chemotactic activity. Most of the human genes encoding CC chemokines are clustered on chromosome 17, although there are a few examples of CC chemokine genes that map elsewhere. Other chemokines include lymphotactin (C chemokine); macrophage chemotactic and activating factor (MCAF/MCP-1; CC chemokine); platelet factor 4 and IL8 (CXC chemokines); and fractalkine and neurotractin (CX3C chemokines). (Reviewed in Luster, A. D. (1998) N. Engl. J. Med. 338:436-445.)


Other proteins that contain signal peptides include secreted proteins with enzymatic activity. Such activity includes, for example, oxidoreductase/dehydrogenase activity, transferase activity, hydrolase activity, lyase activity, isomerase activity, or ligase activity. For example, matrix metalloproteinases are secreted hydrolytic enzymes that degrade the extracellular matrix and thus play an important role in tumor metastasis, tissue morphogenesis, and arthritis (Reponen, P. et al. (1995) Dev. Dyn. 202:388-396; Firestein, G. S. (1992) Curr. Opin. Rheumatol. 4:348-354; Ray, J. M. and Stetler-Stevenson, W. G. (1994) Eur. Respir. J. 7:2062-2072; and Mignatti, P. and Riflkin, D. B. (1993) Physiol. Rev. 73:161-195). The catalytic protein disulfide isomerase (PDI) is found in membrane-bound eukaryotic compartments such as the endoplasmic reticulum (ER). It facilitates disulfide bond exchange as well as correct glycosylation. Edman et al. (1995; Nature 317:267-70) reported that rat PDI is useful for the in vitro production and folding of recombinant human proteins. Likewise, purified PDI is also commercially useful for the production and folding of recombinant, therapeutic human proteins such as tissue plasminogen activator (tPA). Ceruloplasmin is a serum multicopper oxidase which plays a role in iron metabolism. Aceruloplasminemia is characterized by diabetes, retinal degeneration, and neurologic symptoms (for a review, see Gitlin, J. D. (1998) Pediatr. Res. 4:271-276). Additional examples are the acetyl-CoA synthetases which activate acetate for use in lipid synthesis or energy generation (Luong, A. et al. (2000) J. Biol. Chem. 275:26458-26466). The result of acetyl-CoA synthetase activity is the formation of acetyl-CoA from acetate and CoA. Acetyl-CoA sythetases share a region of sequence similarity identified as the AMP-binding domain signature. Acetyl-CoA synthetase has been shown to be associated with hypertension (Toh, H. (1991) Protein Seq. Data Anal. 4:111-117; and Iwai, N. et al. (1994) Hypertension 23:375-380).


A number of isomerases catalyze steps in protein folding, phototransduction, and various anabolic and catabolic pathways. One class of isomerases is known as peptidyl-prolyl cis-trans isomerases (PPIases). PPIases catalyze the cis to trans isomerization of certain proline imidic bonds in proteins. Two families of PPIases are the PK506 binding proteins (FKBPs), and cyclophilins (CyPs). FKBPs bind the potent immunosuppressants FK506 and rapamycin, thereby inhibiting signaling pathways in T-cells. Specifically, the PPIase activity of FKBPs is inhibited by binding of FK506 or rapamycin. There are five members of the FKBP family which are named according to their calculated molecular masses (FKBP12, FKBP13, FKBP25, FKBP52, and FKBP65), and localized to different regions of the cell where they associate with different protein complexes (Coss, M. et al. (1995) J. Biol. Chem. 270:29336-29341; Schreiber, S. L. (1991) Science 251:283-287).


The peptidyl-prolyl isomerase activity of CyP may be part of the signaling pathway that leads to T-cell activation. CyP isomerase activity is associated with protein folding and protein trafficking, and may also be involved in assembly/disassembly of protein complexes and regulation of protein activity. For example, in Drosophila, the CyP NinaA is required for correct localization of rhodopsins, while a mamnmalian CyP (Cyp40) is part of the Hsp90/Hsc70 complex that binds steroid receptors. The ammalian CypA has been shown to bind the gag protein from human immunodeficiency virus 1 (HIV-1), an interaction that can be inhibited by cyclosporin. Since cyclosporin has potent anti-HIV-1 activity, CypA may play an essential function in HIV-1 replication. Finally, Cyp40 has been shown to bind and inactivate the transcription factor c-Myb, an effect that is reversed by cyclosporin. This effect implicates CyPs in the regulation of transcription, transformation, and differentiation (Bergsma, D. J. et al (1991) J. Biol. Chem. 266:23204-23214; Hunter, T. (1998) Cell 92:141-143; and Leverson, J. D. and Ness, S. A. (1998) Mol. Cell. 1:203-211).


Gamma-carboxyglutamic acid (Gla) proteins rich in proline (PRGPs) are members of a family of vitamin K-dependent single-pass integral membrane proteins. These proteins are characterized by an extracellular amino terminal domain of approximately 45 amino acids rich in Gla. The intracellular carboxyl terminal region contains one or two copies of the sequence PPXY, a motif present in a variety of proteins involved in such diverse cellular functions as signal transduction, cell cycle progression, and protein turnover (Kulman, J. D. et al. (2001) Proc. Natl. Acad. Sci. USA 98:1370-1375). The process of post-translational modification of glutamic residues to form Gla is Vitamin K-dependent carboxylation. Proteins which contain Gla include plasma proteins involved in blood coagulation. These proteins are prothrombin, proteins C, S, and Z, and coagulation factors VII, IX, and X. Osteocalcin (bone-Gla protein, BGP) and matrix Gla-protein (MGP) also contain Gla (Friedman, P. A. and C. T. Przysiecki (1987) Int. J. Biochem. 19:1-7; C. Vermeer (1990) Biochem. J. 266:625-636).


Immunoglobulins


Antigen recognition molecules are key players in the sophisticated and complex immune systems which all vertebrates have developed to provide protection from viral, bacterial, fungal, and parasitic infections. A key feature of the immune system is its ability to distinguish foreign molecules, or antigens, from “self” molecules. This ability is mediated primarily by secreted and transmembrane proteins expressed by leukocytes (white blood cells) such as lymphocytes, granulocytes, and monocytes. Most of these proteins belong to the immunoglobulin (Ig) superfamily, members of which contain one or more repeats of a conserved structural domain. This Ig domain is comprised of antiparallel β sheets joined by a disulfide bond in an arrangement called the Ig fold. The criteria for a protein to be a member of the Ig superfamily is to have one or more Ig domains, which are regions of 70-110 amino acid residues in length homologous to either Ig variable-like (V) or Ig constant-like (C) domains. Members of the Ig superfamily include antibodies (Ab), T cell receptors (TCRs), class I and II major histocompatibility (MHC) proteins and immune cell-specific surface markers such as the “cluster of differentiation” or CD antigens, CD2, CD3, CD4, CD8, poly-Ig receptors, Fc receptors, neural cell-adhesion molecule (NCAM) and platelet-derived growth factor receptor (PDGFR).


Ig domains (V and C) are regions of conserved amino acid residues that give a polypeptide a globular tertiary structure called an immunoglobulin (or antibody) fold, which consists of two approximately parallel layers of β-sheets. Conserved cysteine residues form an intrachain disulfide-bonded loop, 55-75 amino acid residues in length, which connects the two layers of β-sheets. Each β-sheet has three or four anti-parallel β-strands of 5-10 amino acid residues. Hydrophobic and hydrophilic interactions of amino acid residues within the β-strands stabilize the Ig fold (hydrophobic on inward facing amino acid residues and hydrophilic on the amino acid residues in the outward facing portion of the strands). A V domain consists of a longer polypeptide than a C domain, with an additional pair of β-strands in the Ig fold.


A consistent feature of Ig superfamily genes is that each sequence of an Ig domain is encoded by a single exon. It is possible that the superfamily evolved from a gene coding for a single Ig domain involved in mediating cell-cell interactions. New members of the superfamily then arose by exon and gene duplications. Modern Ig superfamily proteins contain different numbers of V and/or C domains. Another evolutionary feature of this superfamily is the ability to undergo DNA rearrangements, a unique feature retained by the antigen receptor members of the family.


Many members of the Ig superfamily are integral plasma membrane proteins with extracellular Ig domains. The hydrophobic amino acid residues of their transmembrane domains and their cytoplasmic tails are very diverse, with little or no homology among Ig family members or to known signal-transducing structures. There are exceptions to this general superfamily description. For example, the cytoplasmic tail of PDGFR has tyrosine kinase activity. In addition Thy-1 is a glycoprotein found on thymocytes and T cells. This protein has no cytoplasmic tail, but is instead attached to the plasma membrane by a covalent glycophosphatidylinositol linkage.


Another common feature of many Ig superfamily proteins is the interactions between Ig domains which are essential for the function of these molecules. Interactions between Ig domains of a multimeric protein can be either homophilic or heterophilic (i.e., between the same or different Ig domains). Antibodies are multimeric proteins which have both homophilic and heterophilic interactions between Ig domains. Pairing of constant regions of heavy chains forms the Fc region of an antibody and pairing of variable regions of light and heavy chains form the antigen binding site of an antibody. Heterophilic interactions also occur between Ig domains of different molecules. These interactions provide adhesion between cells for significant cell-cell interactions in the immune system and in the developing and mature nervous system. (Reviewed in Abbas, A. K. et al. (1991) Cellular and Molecular Immunology, W.B. Saunders Company, Philadelphia, Pa., pp. 142-145.) Antibodies.


MHC proteins are cell surface markers that bind to and present foreign antigens to T cells. MHC molecules are classified as either class I or class II. Class I MHC molecules (MHC I) are expressed on the surface of almost all cells and are involved in the presentation of antigen to cytotoxic T cells. For example, a cell infected with virus will degrade intracellular viral proteins and express the protein fragments bound to MHC I molecules on the cell surface. The MHC I/antigen complex is recognized by cytotoxic T-cells which destroy the infected cell and the virus within. Class II MHC molecules are expressed primarily on specialized antigen-presenting cells of the immune system, such as B-cells and macrophages. These cells ingest foreign proteins from the extracellular fluid and express MHC II/antigen complex on the cell surface. This complex activates helper T-cells, which then secrete cytokines and other factors that stimulate the immune response. MHC molecules also play an important role in organ rejection following transplantation. Rejection occurs when the recipient's T-cells respond to foreign MHC molecules on the transplanted organ in the same way as to self MHC molecules bound to foreign antigen. (Reviewed in Alberts, B. et al. (1994) Molecular Biology of the Cell, Garland Publishing, New York, N.Y., pp. 1229-1246.)


Antibodies are multimeric members of the Ig superfamily which are either expressed on the surface of B-cells or secreted by B-cells into the circulation. Antibodies bind and neutralize foreign antigens in the blood and other extracellular fluids. The prototypical antibody is a tetramer consisting of two identical heavy polypeptide chains (H-chains) and two identical light polypeptide chains (L-chains) interlinked by disulfide bonds. This arrangement confers the characteristic Y-shape to antibody molecules. Antibodies are classified based on their H-chain composition. The five antibody classes, IgA, IgD, IgE, IgG and IgM, are defined by the α, δ, ε, γ, and μ H-chain types. There are two types of L-chains, κ, and λ, either of which may associate as a pair with any H-chain pair. IgG, the most common class of antibody found in the circulation, is tetrameric, while the other classes of antibodies are generally variants or multimers of this basic structure.


H-chains and L-chains each contain an N-terminal variable region and a C-terminal constant region. The constant region consists of about 110 amino acids in L-chains and about 330 or 440 amino acids in H-chains. The amino acid sequence of the constant region is nearly identical among H- or L-chains of a particular class. The variable region consists of about 110 amino acids in both H- and L-chains. However, the amino acid sequence of the variable region differs among H- or L-chains of a particular class. Within each H- or L-chain variable region are three hypervariable regions of extensive sequence diversity, each consisting of about 5 to 10 amino acids. In the antibody molecule, the H- and L-chain hypervariable regions come together to form the antigen recognition site. (reviewed in Alberts, B. et al. supra, pp. 1206-1213 and 1216-1217.)


Both H-chains and L-chains contain the repeated Ig domains of members of the Ig superfamily. For example, a typical H-chain contains four Ig domains, three of which occur within the constant region and one of which occurs within the variable region and contributes to the formation of the antigen recognition site. Likewise, a typical L-chain contains two Ig domains, one of which occurs within the constant region and one of which occurs within the variable region.


The immune system is capable of recognizing and responding to any foreign molecule that enters the body. Therefore, the immune system must be armed with a full repertoire of antibodies against all potential antigens. Such antibody diversity is generated by somatic rearrangement of gene segments encoding variable and constant regions. These gene segments are joined together by site-specific recombination which occurs between highly conserved DNA sequences that flank each gene segment. Because there are hundreds of different gene segments, millions of unique genes can be generated combinatorially. In addition, imprecise joining of these segments and an unusually high rate of somatic mutation within these segments further contribute to the generation of a diverse antibody population.


The discovery of new secreted proteins, and the polynucleotides encoding them, satisfies a need in the art by providing new compositions which are useful in the diagnosis, prevention, and treatment of cell proliferative, autoimmune/inflammatory, cardiovascular, neurological, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of secreted proteins.


SUMMARY OF THE INVENTION

The invention features purified polypeptides, secreted proteins, referred to collectively as “SECP” and individually as “SECP-1,” “SECP-2,” “SECP-3,” “SECP-4,” “SECP-5,” “SECT-6,” “SECP-7,” “SECP-8,” “SECP-9,” “SECP-10,” “SECP-11,” “SECP-12,” “SECP-13,” “SECP-14,” “SECP-15,” “SECP-16,” “SECP-17,” “SECP-18,” “SECP-19,” “SECP-20,” “SECP-21,” “SECP-22,” “SECP-23,” “SECP-24,” “SECP-25,” “SECP-26,” “SECP-27,” “SECP-28,” “SECP-29,” “SECP-30,” “SECP-31,” “SECP-32,” “SECP-33,” “SECP-34,” “SECP-35,” “SECP-36,” “SECP-37,” “SECP-38,” “SECP-39,” “SECP-40,” “SECP-41,” “SECP-42,” “SECP-43,” “SECP-44,” “SECP-45,” “SECP-46,” “SECP-47,” “SECP-48,” “SECP-49,” “SECP-50,” “SECP-51,” “SECP-52,” “SECP-53,” and “SECP-54.” In one aspect, the invention provides an isolated polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. In one alternative, the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:1-54.


The invention further provides an isolated polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. In one alternative, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ ID NO:1-54. In another alternative, the polynucleotide is selected from the group consisting of SEQ ID NO:55-108.


Additionally, the invention provides a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. In one alternative, the invention provides a cell transformed with the recombinant polynucleotide. In another alternative, the invention provides a transgenic organism comprising the recombinant polynucleotide.


The invention also provides a method for producing a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The method comprises a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recovering the polypeptide so expressed.


Additionally, the invention provides an isolated antibody which specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.


The invention further provides an isolated polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). In one alternative, the polynucleotide comprises at least 60 contiguous nucleotides.


Additionally, the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and optionally, if present, the amount thereof. In one alternative, the probe comprises at least 60 contiguous nucleotides.


The invention further provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.


The invention further provides a composition comprising an effective amount of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and a pharmaceutically acceptable excipient. In one embodiment, the composition comprises an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The invention additionally provides a method of treating a disease or condition associated with decreased expression of functional SECP, comprising administering to a patient in need of such treatment the composition.


The invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample. In one alternative, the invention provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with decreased expression of functional SECP, comprising administering to a patient in need of such treatment the composition.


Additionally, the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample. In one alternative, the invention provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with overexpression of functional SECP, comprising administering to a patient in need of such treatment the composition.


The invention further provides a method of screening for a compound that specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The method comprises a) combining the polypeptide with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide to the test compound, thereby identifying a compound that specifically binds to the polypeptide.


The invention further provides a method of screening for a compound that modulates the activity of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54. The method comprises a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.


The invention further provides a method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, the method comprising a) exposing a sample comprising the target polynucleotide to a compound, b) detecting altered expression of the target polynucleotide, and c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.


The invention further provides a method for assessing toxicity of a test compound, said method comprising a) treating a biological sample containing nucleic acids with the test compound; b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, iii) a polynucleotide having a sequence complementary to i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)iv). Hybridization occurs under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, iii) a polynucleotide complementary to the polynucleotide of i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Alternatively, the target polynucleotide comprises a fragment of a polynucleotide sequence selected from the group consisting of i)-v) above; c) quantifying the amount of hybridization complex; and d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.


BRIEF DESCRIPTION OF THE TABLES

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


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


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


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


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


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


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







DESCRIPTION OF THE INVENTION

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


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


Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any machines, materials, and methods similar or equivalent to those described herein can be used to practice or test the present invention, the preferred machines, materials and methods are now described. All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.


Definitions


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


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


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


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


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


“Amplification” relates to the production of additional copies of a nucleic acid sequence. Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art.


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


The term “antibody” refers to intact immunoglobulin molecules as well as to fragments thereof, such as Fab, F(ab′)2, and Fv fragments, which are capable of binding an epitopic determinant. Antibodies that bind SECP polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or oligopeptide used to immunize an animal (e.g., a mouse, a rat, or a rabbit) can be derived from the translation of RNA, or synthesized chemically, and can be conjugated to a carrier protein if desired. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal.


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


The term “aptamer” refers to a nucleic acid or oligonucleotide molecule that binds to a specific molecular target. Aptamers are derived from an in vitro evolutionary process (e.g., SELEX (Systematic Evolution of Ligands by EXponential Enrichment), described in U.S. Pat. No. 5,270,163), which selects for target-specific aptamer sequences from large combinatorial libraries. Aptamer compositions may be double-stranded or single-stranded, and may include deoxyribonucleotides, ribonucleotides, nucleotide derivatives, or other nucleotide-like molecules. The nucleotide components of an aptamer may have modified sugar groups (e.g., the 2′-OH group of a ribonucleotide may be replaced by 2′-F or 2′-NH2), which may improve a desired property, e.g., resistance to nucleases or longer lifetime in blood. Aptamers may be conjugated to other molecules, e.g., a high molecular weight carrier to slow clearance of the aptamer from the circulatory system. Aptamers may be specifically cross-linked to their cognate ligands, e.g., by photo-activation of a cross-linker. (See, e.g., Brody, E. N. and L. Gold (2000) J. Biotechnol:74:5-13.)


The term “intramer” refers to an aptamer which is expressed in vivo. For example, a vaccinia virus-based RNA expression system has been used to express specific RNA aptamers at high levels in the cytoplasm of leukocytes (Blind, M. et al. (1999) Proc. Natl. Acad. Sci. USA 96:3606-3610).


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


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


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


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


A “composition comprising a given polynucleotide sequence” and a “composition comprising a given amino acid sequence” refer broadly to any composition containing the given polynucleotide or amino acid sequence. The composition may comprise a dry formulation or an aqueous solution. Compositions comprising polynucleotide sequences encoding SECP or fragments of SECP may be employed as hybridization probes. The probes may be stored in freeze-dried form and may be associated with a stabilizing agent such as a carbohydrate. In hybridizations, the probe may be deployed in an aqueous solution containing salts (e.g., NaCl), detergents (e.g., sodium dodecyl sulfate; SDS), and other components (e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.).


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


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

Original ResidueConservative SubstitutionAlaGly, SerArgHis, LysAsnAsp, Gln, HisAspAsn, GluCysAla, SerGlnAsn, Glu, HisGluAsp, Gln, HisGlyAlaHisAsn, Arg, Gln, GluIleLeu, ValLeuIle, ValLysArg, Gln, GluMetLeu, IlePheHis, Met, Leu, Trp, TyrSerCys, ThrThrSer, ValTrpPhe, TyrTyrHis, Phe, TrpValIle, Leu, Thr


Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution; for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.


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


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


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


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


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


A “fragment” is a unique portion of SECP or the polynucleotide encoding SECP which is identical in sequence to but shorter in length than the parent sequence. A fragment may comprise up to the entire length of the defined sequence, minus one nucleotide/amino acid residue. For example, a fragment may comprise from 5 to 1000 contiguous nucleotides or amino acid residues. A fragment used as a probe, primer, antigen, therapeutic molecule, or for other purposes, may be at least 5, 10, 15, 16, 20, 25, 30, 40, 50, 60, 75, 100, 150, 250 or at least 500 contiguous nucleotides or amino acid residues in length. Fragments may be preferentially selected from certain regions of a molecule. For example, a polypeptide fragment may comprise a certain length of contiguous amino acids selected from the first 250 or 500 amino acids (or first 25% or 50%) of a polypeptide as shown in a certain defined sequence. Clearly these lengths are exemplary, and any length that is supported by the specification, including the Sequence Listing, tables, and figures, may be encompassed by the present embodiments.


A fragment of SEQ ID NO:55-108 comprises a region of unique polynucleotide sequence that specifically identifies SEQ ID NO:55-108, for example, as distinct from any other sequence in the genome from which the fragment was obtained. A fragment of SEQ ID NO:55-108 is useful, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ ID NO:55-108 from related polynucleotide sequences. The precise length of a fragment of SEQ ID NO:55-108 and the region of SEQ ID NO:55-108 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.


A fragment of SEQ ID NO:1-54 is encoded by a fragment of SEQ ID NO:55-108. A fragment of SEQ ID NO:1-54 comprises a region of unique amino acid sequence that specifically identifies SEQ ID NO:1-54. For example, a fragment of SEQ ID NO:1-54 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID NO:1-54. The precise length of a fragment of SEQ ID NO:1-54 and the region of SEQ ID NO:1-54 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.


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


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


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


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


Alternatively, a suite of commonly used and freely available sequence comparison algorithms is provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) (Altschul, S. F. et al. (1990) J. Mol. Biol. 215:403-410), which is available from several sources, including the NCBI, Bethesda, Md., and on the Internet at http://www.ncbi.nlm.nih.gov/BLAST/. The BLAST software suite includes various sequence analysis programs including “blastn,” that is used to align a known polynucleotide sequence with other polynucleotide sequences from a variety of databases. Also available is a tool called “BLAST 2 Sequences” that is used for direct pairwise comparison of two nucleotide sequences. “BLAST 2 Sequences” can be accessed and used interactively at http://www.ncbi.nlm.nih.gov/gorf/b12.html. The “BLAST 2 Sequences” tool can be used for both blastn and blastp (discussed below). BLAST programs are commonly used with gap and other parameters set to default settings. For example, to compare two nucleotide sequences, one may use blastn with the “BLAST 2 Sequences” tool Version 2.0.12 (Apr.-21-2000) set at default parameters. Such default parameters may be, for example:

    • Matrix: BLOSUM62
    • Reward for match: 1
    • Penalty for mismatch: −2
    • Open Gap: 5 and Extension Gap: 2 penalties
    • Gap×drop-off: 50
    • Expect: 10
    • Word Size: 11
    • Filter: on


Percent identity may be measured over the length of an entire defined sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined sequence, for instance, a fragment of at least 20, at least 30, at least 40, at least 50, at least 70, at least 100, or at least 200 contiguous nucleotides. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures, or Sequence Listing, may be used to describe a length over which percentage identity may be measured.


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


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


Percent identity between polypeptide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN version 3.12e sequence alignment program (described and referenced above). For pairwise alignments of polypeptide sequences using CLUSTAL V, the default parameters are set as follows: Ktuple=1, gap penalty=3, window=5, and “diagonals saved”=5. The PAM250 matrix is selected as the default residue weight table. As with polynucleotide alignments, the percent identity is reported by CLUSTAL V as the “percent similarity” between aligned polypeptide sequence pairs.


Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise comparison of two polypeptide sequences, one may use the “BLAST 2 Sequences” tool Version 2.0.12 (Apr.-21-2000) with blastp set at default parameters. Such default parameters may be, for example:

    • Matrix: BLOSUM62
    • Open Gap: 11 and Extension Gap: 1 penalties
    • Gap×drop-off. 50
    • Expect: 10
    • Word Size: 3
    • Filter: on


Percent identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.


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


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


“Hybridization” refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of complementarity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after the “washing” step(s). The washing step(s) is particularly important in determining the stringency of the hybridization process, with more stringent conditions allowing less non-specific binding, i.e., binding between pairs of nucleic acid strands that are not perfectly matched. Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve the desired stringency, and therefore hybridization specificity. Permissive annealing conditions occur, for example, at 68° C. in the presence of about 6×SSC, about 1% (w/v) SDS, and about 100 μg/ml sheared, denatured salmon sperm DNA.


Generally, stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5° C. to 20° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. An equation for calculating Tm, and conditions for nucleic acid hybridization are well known and can be found in Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3, Cold Spring Harbor Press, Plainview N.Y.; specifically see volume 2, chapter 9.


High stringency conditions for hybridization between polynucleotides of the present invention include wash conditions of 68° C. in the presence of about 0.2×SSC and about 0.1% SDS, for 1 hour. Alternatively, temperatures of about 65° C., 60° C., 55° C., or 42° C. may be used. SSC concentration may be varied from about 0.1 to 2×SSC, with SDS being present at about 0.1%. Typically, blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 μg/ml. Organic solvent, such as formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for RNA:DNA hybridizations. Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art. Hybridization, particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is strongly indicative of a similar role for the nucleotides and their encoded polypeptides.


The term “hybridization complex” refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (e.g., C0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed).


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


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


An “immunogenic fragment” is a polypeptide or oligopeptide fragment of SECP which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal. The term “immunogenic fragment” also includes any polypeptide or oligopeptide fragment of SECP which is useful in any of the antibody production methods disclosed herein or known in the art.


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


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


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


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


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


“Peptide nucleic acid” (PNA) refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition. PNAs preferentially bind complementary single stranded DNA or RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell.


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


“Probe” refers to nucleic acid sequences encoding SECP, their complements, or fragments thereof, which are used to detect identical, allelic or related nucleic acid sequences. Probes are isolated oligonucleotides or polynucleotides attached to a detectable label or reporter molecule. Typical labels include radioactive isotopes, ligands, chemiluminescent agents, and enzymes. “Primers” are short nucleic acids, usually DNA oligonucleotides, which may be annealed to a target polynucleotide by complementary base-pairing. The primer may then be extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification (and identification) of a nucleic acid sequence, e.g., by the polymerase chain reaction (PCR).


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


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


Oligonucleotides for use as primers are selected using software known in the art for such purpose. For example, OLIGO 4.06 software is useful for the selection of PCR primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases. Similar primer selection programs have incorporated additional features for expanded capabilities. For example, the PrimOU primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas Tex.) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope. The Primer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambridge Mass.) allows the user to input a “mispriming library,” in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarrays. (The source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.) The PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences. Hence, this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments. The oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of oligonucleotide selection are not limited to those described above.


A “recombinant nucleic acid” is a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two or more otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques such as those described in Sambrook, supra. The term recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion of a portion of the nucleic acid. Frequently, a recombinant nucleic acid may include a nucleic acid sequence operably linked to a promoter sequence. Such a recombinant nucleic acid may be part of a vector that is used, for example, to transform a cell.


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


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


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


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


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


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


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


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


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


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


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


A “transgenic organism,” as used herein, is any organism, including but not limited to animals and plants, in which one or more of the cells of the organism contains heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. The transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals. The isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation. Techniques for transferring the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook et al. (1989), supra.


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


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


The Invention


The invention is based on the discovery of new human secreted proteins (SECP), the polynucleotides encoding SECP, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative, autoimmune/inflammatory, cardiovascular, neurological, and developmental disorders.


Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the invention. Each polynucleotide and its corresponding polypeptide are correlated to a single Incyte project identification number (Incyte Project ID). Each polypeptide sequence is denoted by both a polypeptide sequence identification number (Polypeptide SEQ ID NO:) and an Incyte polypeptide sequence number (Incyte Polypeptide ID) as shown. Each polynucleotide sequence is denoted by both a polynucleotide sequence identification number (Polynucleotide SEQ ID NO:) and an Incyte polynucleotide consensus sequence number (Incyte Polynucleotide ID) as shown.


Table 2 shows sequences with homology to the polypeptides of the invention as identified by BLAST analysis against the GenBank protein (genpept) database. Columns 1 and 2 show the polypeptide sequence identification number (Polypeptide SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for polypeptides of the invention. Column 3 shows the GenBank identification number (GenBank ID NO:) of the nearest GenBank homolog. Column 4 shows the probability scores for the matches between each polypeptide and its homolog(s). Column 5 shows the annotation of the GenBak homolog(s) along with relevant citations where applicable, all of which are expressly incorporated by reference herein.


Table 3 shows various structural features of the polypeptides of the invention. Columns 1 and 2 show the polypeptide sequence identification number (SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for each polypeptide of the invention. Column 3 shows the number of amino acid residues in each polypeptide. Column 4 shows potential phosphorylation sites, and column 5 shows potential glycosylation sites, as determined by the MOTIFS program of the GCG sequence analysis software package (Genetics Computer Group, Madison Wis.). Column 6 shows amino acid residues comprising signature sequences, domains, and motifs, including the locations of signal peptides (as indicated by “Signal Peptide” and/or “signal_cleavage”.). Column 7 shows analytical methods for protein structure/function analysis and in some cases, searchable databases to which the analytical methods were applied.


Together, Tables 2 and 3 summarize the properties of polypeptides of the invention, and these properties establish that the claimed polypeptides are secreted proteins. For example, SEQ ID NO:2 is 99% identical to a novel human AMP-binding enzyme similar to acetyl-coenzyme A synthethase (acetate-coA ligase) (GenBank ID g6996429) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 5.8e-262, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:2 also contains an AMP-binding domain signature as determined by searching for statistically significant matches in the hidden Markov model (HM-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS, MOTIFS, and PROFILESCAN analyses provide further corroborative evidence that SEQ ID NO:2 is an AMP-binding enzyme (note that “AMP-binding domains” are shared regions of sequence similarity within a number of prokaryotic and eukaryotic enzymes which most likely act via an ATP-dependent covalent binding of AMP to their substrate, PROSITE:PDOC00427).


As a further example, SEQ ID NO:3 is 33% identical from residues E44 to L530 to bovine PDI (protein disulfide isomerase) (GenBank ID g163497) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 1.1e-70, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:3 also contains a thioredoxin domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS and PROFILESCAN analyses provide further corroborative evidence that SEQ ID NO:3 is a protein disulfide isomerase.


As a further example, SEQ ID NO:4 is 56% identical to human preceruloplasmin (GenBank ID g180256) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 0.0, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:4 contains a signal peptide and a multicopper oxidase active site domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) The presence of this domain is confirmed by BLIMPS, MOTIFS, and PROFILESCAN analyses, providing further corroborative evidence that SEQ ID NO:4 is a secreted multicopper oxidase.


In another example, SEQ ID NO:16 is 79% identical to human growth hormone hGH-V2 (GenBank ID g183178) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 5.6e-106, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:16 also contains a signal peptide and a somatotropin hormone family signature as determined by searching for statistically significant matches in the hidden Markov model (HM-based PFAM database of conserved protein family domains. (See Table 3.) The presence of these motifs is confirmed by BLIMPS, MOTIFS, SPSCAN, and PROFILESCAN analyses, providing further corroborative evidence that SEQ ID NO:16 is a secreted hormone.


As a further example, SEQ ID NO:27 is 49% identical to mouse Fca/m receptor (GenBank ID g11071950) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 2.2e-115, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:27 also contains an immunoglobulin domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from additional BLAST analyses provide further corroborative evidence that SEQ ID NO:27 is an immunoglobulin domain-containing receptor.


In another example, SEQ ID NO:41 is 99% identical to human chordin (GenBank ID g3822218) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 0.0, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO:41 also contains a von Wiliebrand factor growth regulator domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from MOTIFS analyses provide further corroborative evidence that SEQ ID NO:41 is a growth regulation molecule.


SEQ ID NO:50 contains a signal peptide as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 2.) The presence of the signal peptide is confirmed by data from SPSCAN. SEQ ID NO:1, SEQ ID NO:5-15, SEQ ID NO:17-26, SEQ ID NO:28-40, SEQ ID NO:42-49 and SEQ ID NO:51-54, which were analyzed and annotated in a similar manner, all contain signal peptides as determined by SPSCAN or HMMER analysis. The algorithms and parameters for the analysis of SEQ ID NO:1-54 are described in Table 7.


As shown in Table 4, the full length polynucleotide sequences of the present invention were assembled using cDNA sequences or coding (exon) sequences derived from genomic DNA, or any combination of these two types of sequences. Column 1 lists the polynucleotide sequence identification number (Polynucleotide SEQ ID NO:), the corresponding Incyte polynucleotide consensus sequence number (Incyte ID) for each polynucleotide of the invention, and the length of each polynucleotide sequence in basepairs. Column 2 shows the nucleotide start (5′) and stop (3′) positions of the cDNA and/or genomic sequences used to assemble the full length polynucleotide sequences of the invention, and of fragments of the polynucleotide sequences which are useful, for example, in hybridization or amplification technologies that identify SEQ ID NO:55-108 or that distinguish between SEQ ID NO:55-108 and related polynucleotide sequences.


The polynucleotide fragments described in Column 2 of Table 4 may refer specifically, for example, to Incyte cDNAs derived from tissue-specific cDNA libraries or from pooled cDNA libraries. Alternatively, the polynucleotide fragments described in column 2 may refer to GenBank cDNAs or ESTs which contributed to the assembly of the full length polynucleotide sequences. In addition, the polynucleotide fragments described in column 2 may identify sequences derived from the ENSEMBL (The Sanger Centre, Cambridge, UK) database (i.e., those sequences including the designation “ENST”). Alternatively, the polynucleotide fragments described in column 2 may be derived from the NCBI RefSeq Nucleotide Sequence Records Database (i.e., those sequences including the designation “NM” or “NT”) or the NCBI RefSeq Protein Sequence Records (i.e., those sequences including the designation “NP”). Alternatively, the polynucleotide fragments described in column 2 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an “exon stitching” algorithm. For example, a polynucleotide sequence identified as FL_XXXXXX_N1—N2—YYYYY_N3—N4 represents a “stitched” sequence in which XXXXXX is the identification number of the cluster of sequences to which the algorithm was applied, and YYYYY is the number of the prediction generated by the algorithm, and N1, 2, 3 . . . , if present, represent specific exons that may have been manually edited during analysis (See Example V). Alternatively, the polynucleotide fragments in column 2 may refer to assemblages of exons brought together by an “exon-stretching” algorithm. For example, a polynucleotide sequence identified as FLXXXXXX_gAAAAA_gBBBBB1_N is a “stretched” sequence, with XXXXXX being the Incyte project identification number, gAAAAA being the GenBank identification number of the human genomic sequence to which the “exon-stretching” algorithm was applied, gBBBBB being the GenBank identification number or NCBI RefSeq identification number of the nearest GenBank protein homolog, and N referring to specific exons (See Example V). In instances where a RefSeq sequence was used as a protein homolog for the “exon-stretching” algorithm, a RefSeq identifier (denoted by “NM,” “NP,” or “NT”) may be used in place of the GenBank identifier (i.e., gBBBBB).


Alternatively, a prefix identifies component sequences that were hand-edited, predicted from genomic DNA sequences, or derived from a combination of sequence analysis methods. The following Table lists examples of component sequence prefixes and corresponding sequence analysis methods associated with the prefixes (see Example IV and Example V).

PrefixType of analysis and/or examples of programsGNN,Exon prediction from genomic sequences using, forGFG,example, GENSCAN (Stanford University, CA, USA) orENSTFGENES (Computer Genomics Group, The Sanger Centre,Cambridge, UK).GBIHand-edited analysis of genomic sequences.FLStitched or stretched genomic sequences (seeExample V).INCYFull length transcript and exon prediction frommapping of EST sequences to the genome. Genomiclocation and EST composition data are combined topredict the exons and resulting transcript.


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


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


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


The invention also encompasses polynucleotides which encode SECP. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO:55-108, which encodes SECP. The polynucleotide sequences of SEQ ID NO:55-108, as presented in the Sequence Listing, embrace the equivalent RNA sequences, wherein occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.


The invention also encompasses a variant of a polynucleotide sequence encoding SECP. In particular, such a variant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding SECP. A particular aspect of the invention encompasses a variant of a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO:55-108 which has at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO:55-108. Any one of the polynucleotide variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of SECP.


In addition, or in the alternative, a polynucleotide variant of the invention is a splice variant of a polynucleotide sequence encoding SECP. A splice variant may have portions which have significant sequence identity to the polynucleotide sequence encoding SECP, but will generally have a greater or lesser number of polynucleotides due to additions or deletions of blocks of sequence arising from alternate splicing of exons during mRNA processing. A splice variant may have less than about 70%, or alternatively less than about 60%, or alternatively less than about 50% polynucleotide sequence identity to the polynucleotide sequence encoding SECP over its entire length; however, portions of the splice variant will have at least about 70%, or alternatively at least about 85%, or alternatively at least about 95%, or alternatively 100% polynucleotide sequence identity to portions of the polynucleotide sequence encoding SECP. For example, a polynucleotide comprising a sequence of SEQ ID NO:108 is a splice variant of a polynucleotide comprising a sequence of SEQ ID NO:94. Any one of the splice variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of SECP.


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


Although nucleotide sequences which encode SECP and its variants are generally capable of hybridizing to the nucleotide sequence of the naturally occurring SECP under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding SECP or its derivatives possessing a substantially different codon usage, e.g., inclusion of non-naturally occurring codons. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding SECP and its derivatives without altering the encoded amino acid sequences include the production of RNA transcripts having more desirable properties, such as a greater half-life, than transcripts produced from the naturally occurring sequence.


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


Also encompassed by the invention are polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ ID NO:55-108 and fragments thereof under various conditions of stringency. (See, e.g., Wahl, G. M. and S. L. Berger (1987) Methods Enzymol. 152:399-407; Kimmel, A. R. (1987) Methods Enzymol. 152:507-511.) Hybridization conditions, including annealing and wash conditions, are described in “Definitions.”


Methods for DNA sequencing are well known in the art and may be used to practice any of the embodiments of the invention. The methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical, Cleveland Ohio), Taq polymerase (Applied Biosystems), thermostable T7 polymerase (Amersham Pharmacia Biotech, Piscataway N.J.), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE amplification system (Life Technologies, Gaithersburg Md.). Preferably, sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno Nev.), PTC200 thermal cycler (MJ Research, Watertown Mass.) and ABI CATALYST 800 thermal cycler (Applied Biosystems). Sequencing is then carried out using either the ABI 373 or 377 DNA sequencing system (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (Molecular Dynamics, Sunnyvale Calif.), or other systems known in the art. The resulting sequences are analyzed using a variety of algorithms which are well known in the art. (See, e.g., Ausubel, F. M. (1997) Short Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., unit 7.7; Meyers, R. A. (1995) Molecular Biology and Biotechnology, Wiley VCH, New York N.Y., pp. 856-853.)


The nucleic acid sequences encoding SECP may be extended utilizing a partial nucleotide sequence and employing various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements. For example, one method which may be employed, restriction-site PCR, uses universal and nested primers to amplify unknown sequence from genomic DNA within a cloning vector. (See, e.g., Sarkar, G. (1993) PCR Methods Applic. 2:318-322.) Another method, inverse PCR, uses primers that extend in divergent directions to amplify unknown sequence from a circularized template. The template is derived from restriction fragments comprising a known genomic locus and surrounding sequences. (See, e.g., Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186.) A third method, capture PCR, involves PCR amplification of DNA fragments adjacent to known sequences in human and yeast artificial chromosome DNA. (See, e.g., Lagerstrom, M. et al. (1991) PCR Methods Applic. 1:111-119.) In this method, multiple restriction enzyme digestions and ligations may be used to insert an engineered double-stranded sequence into a region of unknown sequence before performing PCR. Other methods which may be used to retrieve unknown sequences are known in the art. (See, e.g., Parker, J. D. et al. (1991) Nucleic Acids Res. 19:3055-3060). Additionally, one may use PCR, nested primers, and PROMOTERFINDER libraries (Clontech, Palo Alto Calif.) to walk genomic DNA. This procedure avoids the need to screen libraries and is useful in finding intron/exon junctions. For all PCR-based methods, primers may be designed using commercially available software, such as OLIGO 4.06 primer analysis software (National Biosciences, Plymouth M or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the template at temperatures of about 68° C. to 72° C.


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


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


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


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


The nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara Calif.; described in U.S. Pat. No. 5,837,458; Chang, C.-C. et al. (1999) Nat. Biotechnol. 17:793-797; Christians, F. C. et al. (1999) Nat. Biotechnol. 17:259-264; and Crameri, A. et al. (1996) Nat. Biotechnol. 14:315-319) to alter or improve the biological properties of SECP, such as its biological or enzymatic activity or its ability to bind to other molecules or compounds. DNA shuffling is a process by which a library of gene variants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene variants with the desired properties. These preferred variants may then be pooled and further subjected to recursive rounds of DNA shuffling and selection/screening. Thus, genetic diversity is created through “artificial” breeding and rapid molecular evolution. For example, fragments of a single gene containing random point mutations may be recombined, screened, and then reshuffled until the desired properties are optimized. Alternatively, fragments of a given gene may be recombined with fragments of homologous genes in the same gene family, either from the same or different species, thereby maximizing the genetic diversity of multiple naturally occurring genes in a directed and controllable manner.


In another embodiment, sequences encoding SECP may be synthesized, in whole or in part, using chemical methods well known in the art. (See, e.g., Caruthers, M. H. et al. (1980) Nucleic Acids Symp. Ser. 7:215-223; and Horn, T. et al. (1980) Nucleic Acids Symp. Ser. 7:225-232.) Alternatively, SECP itself or a fragment thereof may be synthesized using chemical methods. For example, peptide synthesis can be performed using various solution-phase or solid-phase techniques. (See, e.g., Creighton, T. (1984) Proteins. Structures and Molecular Properties, WH Freeman, New York N.Y., pp. 55-60; and Roberge, J. Y. et al. (1995) Science 269:202-204.) Automated synthesis may be achieved using the ABI 431A peptide synthesizer (Applied Biosystems). Additionally, the amino acid sequence of SECP, or any part thereof, may be altered during direct synthesis and/or combined with sequences from other proteins, or any part thereof, to produce a variant polypeptide or a polypeptide having a sequence of a naturally occurring polypeptide.


The peptide may be substantially purified by preparative high performance liquid chromatography. (See, e.g., Chiez, R. M. and F. Z. Regnier (1990) Methods Enzymol. 182:392-421.) The composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing. (See, e.g., Creighton, supra, pp. 28-53.)


In order to express a biologically active SECP, the nucleotide sequences encoding SECP or derivatives thereof may be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host. These elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5′ and 3′ untranslated regions in the vector and in polynucleotide sequences encoding SECP. Such elements may vary in their strength and specificity. Specific initiation signals may also be used to achieve more efficient translation of sequences encoding SECP. Such signals include the ATG initiation codon and adjacent sequences, e.g. the Kozak sequence. In cases where sequences encoding SECP and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals including an in-frame ATG initiation codon should be provided by the vector. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular host cell system used. (See, e.g., Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162.)


Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding SECP and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. (See, e.g., Sambrook, J. et al. (1989) Molecular Cloning. A Laboratory Manual, Cold Spring Harbor Press, Plainview N.Y., ch. 4, 8, and 16-17; Ausubel, F. M. et al. (1995) Current Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., ch. 9, 13, and 16.)


A variety of expression vector/host systems may be utilized to contain and express sequences encoding SECP. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e.g., baculovirus); plant cell systems transformed with viral expression vectors (e.g., cauliflower mosaic virus, CAMV, or tobacco mosaic virus, ThV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems. (See, e.g., Sambrook, supra; Ausubel, supra; Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264:5503-5509; Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. USA 91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7:1937-1945; Takamatsu, N. (1987) EMBO J. 6:307-311; The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York N.Y., pp. 191-196; Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA 81:3655-3659; and Harrington, J. J. et al. (1997) Nat. Genet. 15:345-355.) Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids, may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population. (See, e.g., Di Nicola, M. et al. (1998) Cancer Gen. Ther. 5(6):350-356; Yu, M. et al. (1993) Proc. Natl. Acad. Sci. USA 90(13):6340-6344; Buller, R. M. et al. (1985) Nature 317(6040):813-815; McGregor, D. P. et al. (1994) Mol. Immunol. 31(3):219-226; and Verma, I. M. and N. Somia (1997) Nature 389:239-242.) The invention is not limited by the host cell employed.


In bacterial systems, a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding SECP. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding SECP can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla Calif.) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding SECP into the vector's multiple cloning site disrupts the lacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence. (See, e.g., Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264:5503-5509.) When large quantities of SECP are needed, e.g. for the production of antibodies, vectors which direct high level expression of SECP may be used. For example, vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used.


Yeast expression systems may be used for production of SECP. A number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH promoters, may be used in the yeast Saccharomyces cerevisiae or Pichia pastoris. In addition, such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation. (See, e.g., Ausubel, 1995, supra; Bitter, G. A. et al. (1987) Methods Enzymol. 153:516-544; and Scorer, C. A. et al. (1994) Bio/Technology 12:181-184.)


Plant systems may also be used for expression of SECP. Transcription of sequences encoding SECP may be driven by viral promoters, e.g., the 35S and 19S promoters of CaMV used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 6:307-311). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e.g., Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105.) These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. (See, e.g., The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York N.Y., pp. 191-196.)


In mammalian cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, sequences encoding SECP may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain infective virus which expresses SECP in host cells. (See, e.g., Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA 81:3655-3659.) In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells. SV40 or EBV-based vectors may also be used for high-level protein expression.


Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained in and expressed from a plasmid. HACs of about 6 kb to 10 Mb are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. (See, e.g., Harrington, J. J. et al. (1997) Nat. Genet. 15:345-355.)


For long term production of recombinant proteins in mammalian systems, stable expression of SECP in cell lines is preferred. For example, sequences encoding SECP can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media. The purpose of the selectable marker is to confer resistance to a selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be propagated using tissue culture techniques appropriate to the cell type.


Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase and adenine phosphoribosyltransferase genes, for use in tk- and apr cells, respectively. (See, e.g., Wigler, M. et al. (1977) Cell 11:223-232; Lowy, L et al. (1980) Cell 22:817-823.) Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate; neo confers resistance to the aminoglycosides neomycin and G418; and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. (See, e.g., Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. USA 77:3567-3570; Colbere-Garapin, F. et al. (1981) J. Mol. Biol. 150:1-14.) Additional selectable genes have been described, e.g., trpB and hisD, which alter cellular requirements for metabolites. (See, e.g., Hartman, S. C. and R. C. Mulligan (1988) Proc. Natl. Acad. Sci. USA 85:8047-8051.) Visible markers, e.g., anthocyanins, green fluorescent proteins (GFP; Clontech), B glucuronidase and its substrate β-glucuronide, or luciferase and its substrate luciferin may be used. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system. (See, e.g., Rhodes, C. A. (1995) Methods Mol. Biol. 55:121-131.)


Although the presence/absence of marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed. For example, if the sequence encoding SECP is inserted within a marker gene sequence, transformed cells containing sequences encoding SECP can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding SECP under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.


In general, host cells that contain the nucleic acid sequence encoding SECP and that express SECP may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations, PCR amplification, and protein bioassay or immunoassay techniques which include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein sequences.


Immunological methods for detecting and measuring the expression of SECP using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on SECP is preferred, but a competitive binding assay may be employed. These and other assays are well known in the art. (See, e.g., Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St. Paul Minn., Sect. IV; Coligan, J. E. et al. (1997) Current Protocols in Immunology, Greene Pub. Associates and Wiley-Interscience, New York N.Y.; and Pound, J. D. (1998) Immunochemical Protocols, Humana Press, Totowa N.J.)


A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding SECP include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide. Alternatively, the sequences encoding SECP, or any fragments thereof, may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Amersham Pharmacia Biotech, Promega (Madison Wis.), and US Biochemical. Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.


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


In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a “prepro” or “pro” form of the protein may also be used to specify protein targeting, folding, and/or activity. Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and W138) are available from the American Type Culture Collection (ATCC, Manassas Va.) and may be chosen to ensure the correct modification and processing of the foreign protein.


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


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


SECP of the present invention or fragments thereof may be used to screen for compounds that specifically bind to SECP. At least one and up to a plurality of test compounds may be screened for specific binding to SECP. Examples of test compounds include antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules.


In one embodiment, the compound thus identified is closely related to the natural ligand of SECP, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner. (See, e.g., Coligan, J. E. et al. (1991) Current Protocols in Immunology 1(2): Chapter 5.) Similarly, the compound can be closely related to the natural receptor to which SECP binds, or to at least a fragment of the receptor, e.g., the ligand binding site. In either case, the compound can be rationally designed using known techniques. In one embodiment, screening for these compounds involves producing appropriate cells which express SECP, either as a secreted protein or on the cell membrane. Preferred cells include cells from mammals, yeast, Drosophila, or E. coli. Cells expressing SECP or cell membrane fractions which contain SECP are then contacted with a test compound and binding, stimulation, or inhibition of activity of either SECP or the compound is analyzed.


An assay may simply test binding of a test compound to the polypeptide, wherein binding is detected by a fluorophore, radioisotope, enzyme conjugate, or other detectable label. For example, the assay may comprise the steps of combining at least one test compound with SECP, either in solution or affixed to a solid support, and detecting the binding of SECP to the compound. Alternatively, the assay may detect or measure binding of a test compound in the presence of a labeled competitor. Additionally, the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support.


SECP of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of SECP. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for SECP activity, wherein SECP is combined with at least one test compound, and the activity of SECP in the presence of a test compound is compared with the activity of SECP in the absence of the test compound. A change in the activity of SECP in the presence of the test compound is indicative of a compound that modulates the activity of SECP. Alternatively, a test compound is combined with an in vitro or cell-free system comprising SECP under conditions suitable for SECP activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of SECP may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened.


In another embodiment, polynucleotides encoding SECP or their mammalian homologs may be “knocked out” in an animal model system using homologous recombination in embryonic stem (ES) cells. Such techniques are well known in the art and are useful for the generation of animal models of human disease. (See, e.g., U.S. Pat. No. 5,175,383 and U.S. Pat. No. 5,767,337.) For example, mouse ES cells, such as the mouse 129/SvJ cell line, are derived from the early mouse embryo and grown in culture. The ES cells are transformed with a vector containing the gene of interest disrupted by a marker gene, e.g., the neomycin phosphotransferase gene (neo; Capecchi, M. R. (1989) Science 244:1288-1292). The vector integrates into the corresponding region of the host genome by homologous recombination. Alternatively, homologous recombination takes place using the Cre-loxP system to knockout a gene of interest in a tissue- or developmental stage-specific manner (Marth, J. D. (1996) Clin. Invest. 97:1999-2002; Wagner, K. U. et al. (1997) Nucleic Acids Res. 25:4323-4330). Transformed ES cells are identified and microinjected into mouse cell blastocysts such as those from the C57BL/6 mouse strain. The blastocysts are surgically transferred to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains. Transgenic animals thus generated may be tested with potential therapeutic or toxic agents.


Polynucleotides encoding SECP may also be manipulated in vitro in ES cells derived from human blastocysts. Human ES cells have the potential to differentiate into at least eight separate cell lineages including endoderm, mesoderm, and ectodermal cell types. These cell lineages differentiate into, for example, neural cells, hematopoietic lineages, and cardiomyocytes (Thomson, J. A. et al. (1998) Science 282:1145-1147).


Polynucleotides encoding SECP can also be used to create “knockin” humanized animals (pigs) or transgenic animals (mice or rats) to model human disease. With knockin technology, a region of a polynucleotide encoding SECP is injected into animal ES cells, and the injected sequence integrates into the animal cell genome. Transformed cells are injected into blastulae, and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease. Alternatively, a mammal inbred to overexpress SECP, e.g., by secreting SECP in its milk, may also serve as a convenient source of that protein (Janne, J. et al. (1998) Biotechnol. Annu. Rev. 4:55-74).


Therapeutics


Chemical and structural similarity, e.g., in the context of sequences and motifs, exists between regions of SECP and secreted proteins. In addition, the expression of SECP is closely associated with breast, reproductive, digestive, urinary, fibroblastic, diseased, tumorous, testicular, pituitary, adenoid, lymph node, monocyte, ileum, coronary artery endothelium, uterine endometrial and brain tissues. Examples can also be found in Table 6. Therefore, SECP appears to play a role in cell proliferative, autoimmune/inflammatory, cardiovascular, neurological, and developmental disorders. In the treatment of disorders associated with increased SECP expression or activity, it is desirable to decrease the expression or activity of SECP. In the treatment of disorders associated with decreased SECP expression or activity, it is desirable to increase the expression or activity of SECP.


Therefore, in one embodiment, SECP or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SECP. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCYD), myelofibrosis, paroxysmal noctumal hemoglobinuria, polycythernia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, a cancer of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma; a cardiovascular disorder such as congestive heart failure, ischemic heart disease, angina pectoris, myocardial infarction, hypertensive heart disease, degenerative valvular heart disease, calcific aortic valve stenosis, congenitally bicuspid aortic valve, mitral annular calcification, mitral valve prolapse, rheumatic fever and rheumatic heart disease, infective endocarditis, nonbacterial thrombotic endocarditis, endocarditis of systemic lupus erythematosus, carcinoid heart disease, cardiomyopathy, myocarditis, pericarditis, neoplastic heart disease, congenital heart disease, complications of cardiac transplantation, arteriovenous fistula, atherosclerosis, hypertension, vasculitis, Raynaud's disease, aneurysms, arterial dissections, varicose veins, thrombophlebitis and phlebothrombosis, vascular tumors, and complications of thrombolysis, balloon angioplasty, vascular replacement, and coronary artery bypass graft surgery; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; and a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss.


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


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


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


In a further embodiment, an antagonist of SECP may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of SECP. Examples of such disorders include, but are not limited to, those cell proliferative, autoimmune/inflammatory, cardiovascular, neurological, and developmental disorders described above. In one aspect, an antibody which specifically binds SECP may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express SECP.


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


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


An antagonist of SECP may be produced using methods which are generally known in the art. In particular, purified SECP may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind SECP. Antibodies to SECP may also be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies (i.e., those which inhibit dinner formation) are generally preferred for therapeutic use.


For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with SECP or with any fragment or oligopeptide thereof which has immunogenic properties. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol. Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are especially preferable.


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


Monoclonal antibodies to SECP may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D. et al. (1985) J. Immunol. Methods 81:3142; Cote, R. J. et al. (1983) Proc. Natl. Acad. Sci. USA 80:2026-2030; and Cole, S. P. et al. (1984) Mol. Cell Biol. 62:109-120.)


In addition, techniques developed for the production of “chimeric antibodies,” such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. (See, e.g., Morrison, S. L. et al. (1984) Proc. Natl. Acad. Sci. USA 81:6851-6855; Neuberger, M. S. et al. (1984) Nature 312:604-608; and Takeda, S. et al. (1985) Nature 314:452-454.) Alternatively, techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce SECP-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (See, e.g., Burton, D. R. (1991) Proc. Natl. Acad. Sci. USA 88:10134-10137.)


Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (See, e.g., Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Winter, G. et al. (1991) Nature 349:293-299.)


Antibody fragments which contain specific binding sites for SECP may also be generated. For example, such fragments include, but are not limited to, F(ab)2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab′)2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e.g., Huse, W. D. et al. (1989) Science 246:1275-1281.)


Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between SECP and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering SECP epitopes is generally used, but a competitive binding assay may also be employed (Pound, supra.


Various methods such as Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for SECP. Affinity is expressed as an association constant, Ka, which is defined as the molar concentration of SECP-antibody complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions. The Ka determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple SECP epitopes, represents the average affinity, or avidity, of the antibodies for SECP. The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular SECP epitope, represents a true measure of affinity. High-affinity antibody preparations with Ka ranging from about 109 to 1012 L/mole are preferred for use in immunoassays in which the SECP-antibody complex must withstand rigorous manipulations. Low-affinity antibody preparations with Ka ranging from about 106 to 107 L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of SECP, preferably in active form, from the antibody (Catty, D. (1988) Antibodies, Volume I: A Practical Approach, IRL Press, Washington D.C.; Liddell, J. E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies, John Wiley & Sons, New York N.Y.).


The titer and avidity of polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications. For example, a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml, preferably 5-10 mg specific antibody/ml, is generally employed in procedures requiring precipitation of SECP-antibody complexes. Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available. (See, e.g., Catty, supra, and Coligan et al. supra.)


In another embodiment of the invention, the polynucleotides encoding SECP, or any fragment or complement thereof, may be used for therapeutic purposes. In one aspect, modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified oligonucleotides) to the coding or regulatory regions of the gene encoding SECP. Such technology is well known in the art, and antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding SECP. (See, e.g., Agrawal, S., ed. (1996) Antisense Therapeutics, Humana Press Inc., Totawa N.J.)


In therapeutic use, any gene delivery system suitable for introduction of the antisense sequences into appropriate target cells can be used. Antisense sequences can be delivered intracellularly in the form of an expression plasmid which, upon transcription, produces a sequence complementary to at least a portion of the cellular sequence encoding the target protein. (See, e.g., Slater, J. E. et al. (1998) J. Allergy Clin. Immunol. 102(3):469-475; and Scanlon, K. J. et al. (1995) 9(13): 1288-1296.) Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors. (See, e.g., Miller, A. D. (1990) Blood 76:271; Ausubel, supra; Uckert, W. and W. Walther (1994) Pharmacol. Ther. 63(3):323-347.) Other gene delivery mechanisms include liposome-derived systems, artificial viral envelopes, and other systems known in the art. (See, e.g., Rossi, J. J. (1995) Br. Med. Bull. 51(1):217-225; Boado, R. J. et al. (1998) J. Pharm. Sci. 87(11):1308-1315; and Morris, M. C. et al. (1997) Nucleic Acids Res. 25(14):2730-2736.)


In another embodiment of the invention, polynucleotides encoding SECP may be used for somatic or germline gene therapy. Gene therapy may be performed to (i) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCID)-X1 disease characterized by X-linked inheritance (Cavazzana-Calvo, M. et al. (2000) Science 288:669-672), severe combined immunodeficiency syndrome associated with an inherited adenosine deaminase (ADA) deficiency (Blaese, R. M. et al. (1995) Science 270:475-480; Bordignon, C. et al. (1995) Science 270:470-475), cystic fibrosis (Zabner, J. et al. (1993) Cell 75:207-216; Crystal, R. G. et al. (1995) Hum. Gene Therapy 6:643-666; Crystal, R. G. et al. (1995) Hum. Gene Therapy 6:667-703), thalassamias, familial hypercholesterolemia, and hemophilia resulting from Factor III or Factor IX deficiencies (Crystal, R. G. (1995) Science 270:404-410; Verma, I. M. and N. Somia (1997) Nature 389:239-242)), (ii) express a conditionally lethal gene product (e.g., in the case of cancers which result from unregulated cell proliferation), or (iii) express a protein which affords protection against intracellular parasites (e.g., against human retroviruses, such as human immunodeficiency virus (HIV) (Baltimore, D. (1988) Nature 335:395-396; Poeschla, E. et al. (1996) Proc. Natl. Acad. Sci. USA 93:11395-11399), hepatitis B or C virus (HBV, HCV); fungal parasites, such as Candida albicans and Paracoccidioides brasiliensis; and protozoan parasites such as Plasmodium falciparum and Trypanosoma cruzi). In the case where a genetic deficiency in SECP expression or regulation causes disease, the expression of SECP from an appropriate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency.


In a further embodiment of the invention, diseases or disorders caused by deficiencies in SECP are treated by constructing mammalian expression vectors encoding SECP and introducing these vectors by mechanical means into SECP-deficient cells. Mechanical transfer technologies for use with cells in vivo or ex vitro include (i) direct DNA microinjection into individual cells, (ii) ballistic gold particle delivery, (iii) liposome-mediated transfection, (iv) receptor-mediated gene transfer, and (v) the use of DNA transposons (Morgan, R. A. and W. F. Anderson (1993) Annu. Rev. Biochem. 62:191-217; Ivics, Z. (1997) Cell 91:501-510; Boulay, J-L. and H. Récipon (1998) Curr. Opin. Biotechnol. 9:445-450).


Expression vectors that may be effective for the expression of SECP include, but are not limited to, the PCDNA 3.1, EPITAG, PRCCMV2, PREP, PVAX, PCR2-TOPOTA vectors (Invitrogen, Carlsbad Calif.), PCMV-SCRIPT, PCMV-TAG, PEGSH/PERV (Stratagene, La Jolla Calif.), and PTET-OFF, PTET-ON, PTRE2, PTRE2-LUC, PTK-HYG (Clontech, Palo Alto Calif.). SECP may be expressed using (i) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or β-actin genes), (ii) an inducible promoter (e.g., the tetracycline-regulated promoter (Gossen, M. and H. Bujard (1992) Proc. Natl. Acad. Sci. USA 89:5547-5551; Gossen, M. et al. (1995) Science 268:1766-1769; Rossi, F. M. V. and H. M. Blau (1998) Curr. Opin. Biotechnol. 9:451-456), commercially available in the T-REX plasmid (Invitrogen)); the ecdysone-inducible promoter (available in the plasmids PVGRXR and PIND; Invitrogen); the FK506/rapamycin inducible promoter; or the RU486/mifepristone inducible promoter (Rossi, F. M. V. and H. M. Blau, supra)), or (iii) a tissue-specific promoter or the native promoter of the endogenous gene encoding SECP from a normal individual.


Commercially available liposome transformation kits (e.g., the PERFECT LIPID TRANSFECTION KIT, available from Invitrogen) allow one with ordinary skill in the art to deliver polynucleotides to target cells in culture and require minimal effort to optimize experimental parameters. In the alternative, transformation is performed using the calcium phosphate method (Graham, F. L. and A. J. Eb (1973) Virology 52:456-467), or by electroporation (Neumann, E. et al. (1982) EMBO J. 1:841-845). The introduction of DNA to primary cells requires modification of these standardized mammalian transfection protocols.


In another embodiment of the invention, diseases or disorders caused by genetic defects with respect to SECP expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding SECP under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (ii) appropriate RNA packaging signals, and (iii) a Rev-responsive element (RRE) along with additional retrovirus cis-acting RNA sequences and coding sequences required for efficient vector propagation. Retrovirus vectors (e.g., PFB and PFBNEO) are commercially available (Stratagene) and are based on published data (Riviere, I. et al. (1995) Proc. Natl. Acad. Sci. USA 92:6733-6737), incorporated by reference herein. The vector is propagated in an appropriate vector producing cell line (VPCL) that expresses an envelope gene with a tropism for receptors on the target cells or a promiscuous envelope protein such as VSVg (Armentano, D. et al. (1987) J. Virol. 61:1647-1650; Bender, M. A. et al. (1987) J. Virol. 61:1639-1646; Adam, M. A. and A. D. Miller (1988) J. Virol. 62:3802-3806; Dull, T. et al. (1998) J. Virol. 72:8463-8471; Zufferey, R. et al. (1998) J. Virol. 72:9873-9880). U.S. Pat. No. 5,910,434 to Rigg (“Method for obtaining retrovirus packaging cell lines producing high transducing efficiency retroviral supernatant”) discloses a method for obtaining retrovirus packaging cell lines and is hereby incorporated by reference. Propagation of retrovirus vectors, transduction of a population of cells (e.g., CD4+ T-cells), and the return of transduced cells to a patient are procedures well known to persons skilled in the art of gene therapy and have been well documented (Ranga, U. et al. (1997) J. Virol. 71:7020-7029; Bauer, G. et al. (1997) Blood 89:2259-2267; Bonyhadi, M. L. (1997) J. Virol. 71:4707-4716; Ranga, U. et al. (1998) Proc. Natl. Acad. Sci. USA 95:1201-1206; Su, L. (1997) Blood 89:2283-2290).


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


In another alternative, a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding SECP to target cells which have one or more genetic abnormalities with respect to the expression of SECP. The use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing SECP to cells of the central nervous system, for which HSV has a tropism. The construction and packaging of herpes-based vectors are well known to those with ordinary skill in the art. A replication-competent herpes simplex virus (HSV) type 1-based vector has been used to deliver a reporter gene to the eyes of primates (Liu, X. et al. (1999) Exp. Eye Res. 169:385-395). The construction of a HSV-1 virus vector has also been disclosed in detail in U.S. Pat. No. 5,804,413 to DeLuca (“Herpes simplex virus strains for gene transfer”), which is hereby incorporated by reference. U.S. Pat. No. 5,804,413 teaches the use of recombinant HSV d92 which consists of a genome containing at least one exogenous gene to be transferred to a cell under the control of the appropriate promoter for purposes including human gene therapy. Also taught by this patent are the construction and use of recombinant HSV strains deleted for ICP4, ICP27 and ICP22. For HSV vectors, see also Goins, W. F. et al. (1999) J. Virol. 73:519-532 and Xu, H. et al. (1994) Dev. Diol. 163:152-161, hereby incorporated by reference. The manipulation of cloned herpesvirus sequences, the generation of recombinant virus following the transfection of multiple plasmids containing different segments of the large herpesvirus genomes, the growth and propagation of herpesvirus, and the infection of cells with herpesvirus are techniques well known to those of ordinary skill in the art.


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


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


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


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


Complementary ribonucleic acid molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding SECP. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as 17 or SP6. Alternatively, these cDNA constructs that synthesize complementary RNA, constitutively or inducibly, can be introduced into cell lines, cells, or tissues.


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


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


At least one, and up to a plurality, of test compounds may be screened for effectiveness in altering expression of a specific polynucleotide. A test compound may be obtained by any method commonly known in the art, including chemical modification of a compound known to be effective in altering polynucleotide expression; selection from an existing, commercially-available or proprietary library of naturally-occurring or non-natural chemical compounds; rational design of a compound based on chemical and/or structural properties of the target polynucleotide; and selection from a library of chemical compounds created combinatorially or randomly. A sample comprising a polynucleotide encoding SECP is exposed to at least one test compound thus obtained. The sample may comprise, for example, an intact or permeabilized cell, or an in vitro cell-free or reconstituted biochemical system. Alterations in the expression of a polynucleotide encoding SECP are assayed by any method commonly known in the art. Typically, the expression of a specific nucleotide is detected by hybridization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding SECP. The amount of hybridization may be quantified, thus forming the basis for a comparison of the expression of the polynucleotide both with and without exposure to one or more test compounds. Detection of a change in the expression of a polynucleotide exposed to a test compound indicates that the test compound is effective in altering the expression of the polynucleotide. A screen for a compound effective in altering expression of a specific polynucleotide can be carried out, for example, using a Schizosaccharomyces pombe gene expression system (Atkins, D. et al. (1999) U.S. Pat. No. 5,932,435; Arndt, G. M. et al. (2000) Nucleic Acids Res. 28:E15) or a human cell line such as HeLa cell (Clarke, M. L. et al. (2000) Biochem. Biophys. Res. Commun. 268:8-13). A particular embodiment of the present invention involves screening a combinatorial library of oligonucleotides (such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides) for antisense activity against a specific polynucleotide sequence (Bruice, T. W. et al. (1997) U.S. Pat. No. 5,686,242; Bruice, T. W. et al. (2000) U.S. Pat. No. 6,022,691).


Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient. Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See, e.g., Goldman, C. K. et al. (1997) Nat. Biotechnol. 15:462-466.)


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


An additional embodiment of the invention relates to the administration of a composition which generally comprises an active ingredient formulated with a pharmaceutically acceptable excipient. Excipients may include, for example, sugars, starches, celluloses, gums, and proteins. Various formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton Pa.). Such compositions may consist of SECP, antibodies to SECP, and mimetics, agonists, antagonists, or inhibitors of SECP.


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


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


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


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


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


A therapeutically effective dose refers to that amount of active ingredient, for example SECP or fragments thereof, antibodies of SECP, and agonists, antagonists or inhibitors of SECP, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the ED50 (the dose therapeutically effective in 50% of the population) or LD50 (the dose lethal to 50% of the population) statistics. The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the LD50/ED50 ratio. Compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used to formulate a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that includes the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.


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


Normal dosage amounts may vary from about 0.1 μg to 100,000 μg, up to a total dose of about 1 gram, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc.


Diagnostics


In another embodiment, antibodies which specifically bind SECP may be used for the diagnosis of disorders characterized by expression of SECP, or in assays to monitor patients being treated with SECP or agonists, antagonists, or inhibitors of SECP. Antibodies useful for diagnostic purposes may be prepared in the same manner as described above for therapeutics. Diagnostic assays for SECP include methods which utilize the antibody and a label to detect SECP in human body fluids or in extracts of cells or tissues. The antibodies may be used with or without modification, and may be labeled by covalent or non-covalent attachment of a reporter molecule. A wide variety of reporter molecules, several of which are described above, are known in the art and may be used.


A variety of protocols for measuring SECP, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of SECP expression. Normal or standard values for SECP expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to SECP under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of SECP expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.


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


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


Probes may also be used for the detection of related sequences, and may have at least 50% sequence identity to any of the SECP encoding sequences. The hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ ID NO:55-108 or from genomic sequences including promoters, enhancers, and introns of the SECP gene.


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


Polynucleotide sequences encoding SECP may be used for the diagnosis of disorders associated with expression of SECP. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, a cancer of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid; and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma; a cardiovascular disorder such as congestive heart failure, ischemic heart disease, angina pectoris, myocardial infarction, hypertensive heart disease, degenerative valvular heart disease, calcific aortic valve stenosis, congenitally bicuspid aortic valve, mitral annular calcification, mitral valve prolapse, rheumatic fever and rheumatic heart disease, infective endocarditis, nonbacterial thrombotic endocarditis, endocarditis of systemic lupus erythematosus, carcinoid heart disease, cardiomyopathy, myocarditis, pericarditis, neoplastic heart disease, congenital heart disease, complications of cardiac transplantation, arteriovenous fistula, atherosclerosis, hypertension, vasculitis, Raynaud's disease, aneurysms, arterial dissections, varicose veins, thrombophlebitis and phlebothrombosis, vascular tumors, and complications of thrombolysis, balloon angioplasty, vascular replacement, and coronary artery bypass graft surgery; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; and a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss. The polynucleotide sequences encoding SECP may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and multiformat ELISA-like assays; and in microarrays utilizing fluids or tissues from patients to detect altered SECP expression. Such qualitative or quantitative methods are well known in the art.


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


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


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


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


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


In a particular aspect, oligonucleotide primers derived from the polynucleotide sequences encoding SECP may be used to detect single nucleotide polymorphisms (SNPs). SNPs are substitutions, insertions and deletions that are a frequent cause of inherited or acquired genetic disease in humans. Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods. In SSCP, oligonucleotide primers derived from the polynucleotide sequences encoding SECP are used to amplify DNA using the polymerase chain reaction (PCR). The DNA may be derived, for example, from diseased or normal tissue, biopsy samples, bodily fluids, and the like. SNPs in the DNA cause differences in the secondary and tertiary structures of PCR products in single-stranded form, and these differences are detectable using gel electrophoresis in non-denaturing gels. In fSCCP, the oligonucleotide primers are fluorescently labeled, which allows detection of the amplimers in high-throughput equipment such as DNA sequencing machines. Additionally, sequence database analysis methods, termed in silico SNP (is SNP), are capable of identifying polymorphisms by comparing the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence. These computer-based methods filter out sequence variations due to laboratory preparation of DNA and sequencing errors using statistical models and automated analyses of DNA sequence chromatograms. In the alternative, SNPs may be detected and characterized by mass spectrometry using, for example, the high throughput MASSARRAY system (Sequenom, Inc., San Diego Calif.).


Methods which may also be used to quantify the expression of SECP include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid; and interpolating results from standard curves. (See, e.g., Melby, P. C. et al. (1993) J. Immunol. Methods 159:235-244; Duplaa, C. et al. (1993) Anal. Biochem. 212:229-236.) The speed of quantitation of multiple samples may be accelerated by running the assay in a high-throughput format where the oligomer or polynucleotide of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.


In further embodiments, oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as elements on a microarray. The microarray can be used in transcript imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as described below. The microarray may also be used to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic basis of a disorder, to diagnose a disorder, to monitor progression/regression of disease as a function of gene expression, and to develop and monitor the activities of therapeutic agents in the treatment of disease. In particular, this information may be used to develop a pharmacogenomic profile of a patient in order to select the most appropriate and effective treatment regimen for that patient. For example, therapeutic agents which are highly effective and display the fewest side effects may be selected for a patient based on his/her pharmacogenomic profile.


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


A particular embodiment relates to the use of the polynucleotides of the present invention to generate a transcript image of a tissue or cell type. A transcript image represents the global pattern of gene expression by a particular tissue or cell type. Global gene expression patterns are analyzed by quantifying the number of expressed genes and their relative abundance under given conditions and at a given time. (See Seilhamer et al., “Comparative Gene Transcript Analysis,” U.S. Pat. No. 5,840,484, expressly incorporated by reference herein.) Thus a transcript image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcripts or reverse transcripts of a particular tissue or cell type. In one embodiment, the hybridization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements comprise a subset of a plurality of elements on a microarray. The resultant transcript image would provide a profile of gene activity.


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


Transcript images which profile the expression of the polynucleotides of the present invention may also be used in conjunction with in vitro model systems and preclinical evaluation of pharmaceuticals, as well as toxicological testing of industrial and naturally-occurring environmental compounds. All compounds induce characteristic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E. F. et al. (1999) Mol. Carcinog. 24:153-159; Steiner, S. and N. L. Anderson (2000) Toxicol. Lett. 112-113:467-471, expressly incorporated by reference herein). If a test compound has a signature similar to that of a compound with known toxicity, it is likely to share those toxic properties. These fingerprints or signatures are most useful and refined when they contain expression information from a large number of genes and gene families. Ideally, a genome-wide measurement of expression provides the highest quality signature. Even genes whose expression is not altered by any tested compounds are important as well, as the levels of expression of these genes are used to normalize the rest of the expression data. The normalization procedure is useful for comparison of expression data after treatment with different compounds. While the assignment of gene function to elements of a toxicant signature aids in interpretation of toxicity mechanisms, knowledge of gene function is not necessary for the statistical matching of signatures which leads to prediction of toxicity. (See, for example, Press Release 00-02 from the National Institute of Environmental Health Sciences, released Feb. 29, 2000, available at http://www.niehs.nih.gov/oc/news/toxchip.htm.) Therefore, it is important and desirable in toxicological screening using toxicant signatures to include all expressed gene sequences.


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


Another particular embodiment relates to the use of the polypeptide sequences of the present invention to analyze the proteome of a tissue or cell type. The term proteome refers to the global pattern of protein expression in a particular tissue or cell type. Each protein component of a proteome can be subjected individually to further analysis. Proteome expression patterns, or profiles, are analyzed by quantifying the number of expressed proteins and their relative abundance under given conditions and at a given time. A profile of a cell's proteome may thus be generated by separating and analyzing the polypeptides of a particular tissue or cell type. In one embodiment, the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectric focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Steiner and Anderson, supra). The proteins are visualized in the gel as discrete and uniquely positioned spots, typically by staining the gel with an agent such as Coomassie Blue or silver or fluorescent stains. The optical density of each protein spot is generally proportional to the level of the protein in the sample. The optical densities of equivalently positioned protein spots from different samples, for example, from biological samples either treated or untreated with a test compound or therapeutic agent, are compared to identify any changes in protein spot density related to the treatment. The proteins in the spots are partially sequenced using, for example, standard methods employing chemical or enzymatic cleavage followed by mass spectrometry. The identity of the protein in a spot may be determined by comparing its partial sequence, preferably of at least 5 contiguous amino acid residues, to the polypeptide sequences of the present invention. In some cases, further sequence data may be obtained for definitive protein identification.


A proteomic profile may also be generated using antibodies specific for SECP to quantify the levels of SECP expression. In one embodiment, the antibodies are used as elements on a microarray, and protein expression levels are quantified by exposing the microarray to the sample and detecting the levels of protein bound to each array element (Lueking, A. et al. (1999) Anal. Biochem. 270:103-111; Mendoze, L. G. et al. (1999) Biotechniques 27:778-788). Detection may be performed by a variety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or amino-reactive fluorescent compound and detecting the amount of fluorescence bound at each array element.


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


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


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


Microarrays may be prepared, used, and analyzed using methods known in the art. (See, e.g., Brennan, T. M. et al. (1995) U.S. Pat. No. 5,474,796; Schena, M. et al. (1996) Proc. Natl. Acad. Sci. USA 93:10614-10619; Baldeschweiler et al. (1995) PCT application WO95/251116; Shalon, D. et al. (1995) PCT application WO95/35505; Heller, R. A. et al. (1997) Proc. Natl. Acad. Sci. USA 94:2150-2155; and Heller, M. J. et al. (1997) U.S. Pat. No. 5,605,662.) Various types of microarrays are well known and thoroughly described in DNA Microarrays: A Practical Approach, M. Schena, ed. (1999) Oxford University Press, London, hereby expressly incorporated by reference.


In another embodiment of the invention, nucleic acid sequences encoding SECP may be used to generate hybridization probes useful in mapping the naturally occurring genomic sequence. Either coding or noncoding sequences may be used, and in some instances, noncoding sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a multi-gene family may potentially cause undesired cross hybridization during chromosomal mapping. The sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, e.g., human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial P1 constructions, or single chromosome cDNA libraries. (See, e.g., Harrington, J. J. et al. (1997) Nat. Genet. 15:345-355; Price, C. M. (1993) Blood Rev. 7:127-134; and Trask, B. J. (1991) Trends Genet. 7:149-154.) Once mapped, the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the inheritance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP). (See, for example, Lander, E. S. and D. Botstein (1986) Proc. Natl. Acad. Sci. USA 83:7353-7357.)


Fluorescent in situ hybridization (FISH) may be correlated with other physical and genetic map data. (See, e.g., Heinz-Ulrich, et al. (1995) in Meyers, supra, pp. 965-968.) Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMIM) World Wide Web site. Correlation between the location of the gene encoding SECP on a physical map and a specific disorder, or a predisposition to a specific disorder, may help define the region of DNA associated with that disorder and thus may further positional cloning efforts.


In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps. Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the exact chromosomal locus is not known. This information is valuable to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the gene or genes responsible for a disease or syndrome have been crudely localized by genetic linkage to a particular genomic region, e.g., ataxia-telangiectasia to 11q22-23, any sequences mapping to that area may represent associated or regulatory genes for further investigation. (See, e.g., Gatti, R. A. et al. (1988) Nature 336:577-580.) The nucleotide sequence of the instant invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals.


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


Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest. (See, e.g., Geysen, et al. (1984) PCT application WO84/03564.) In this method, large numbers of different small test compounds are synthesized on a solid substrate. The test compounds are reacted with SECP, or fragments thereof, and washed. Bound SECP is then detected by methods well known in the art. Purified SECP can also be coated directly onto plates for use in the aforementioned drug screening techniques. Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.


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


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


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


The disclosures of all patents, applications and publications, mentioned above and below, including U.S. Ser. No. 60/262,932, U.S. Ser. No. 60/265,926, U.S. Ser. No. 60/255,639, U.S. Ser. No. 60/257,852, U.S. Ser. No. 60/260,105, U.S. Ser. No. 60/263,090 and U.S. Ser. No. 60/263,096 are expressly incorporated by reference herein.


EXAMPLES

I. Construction of cDNA Libraries


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


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


In some cases, Stratagene was provided with RNA and constructed the corresponding cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Life Technologies), using the recommended procedures or similar methods known in the art. (See, e.g., Ausubel, 1997, supra, units 5.1-6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Life Technologies), PCDNA2.1 plasmid (Invitrogen, Carlsbad Calif.), PBK-CMV plasmid (Stratagene), PCR2-TOPOTA plasmid (Invitrogen), PCMV-ICIS plasmid (Stratagene), pIGEN (Incyte Genomics, Palo Alto Calif.), pRARE (Incyte Genomics), or pINCY (Incyte Genomics), or derivatives thereof. Recombinant plasmids were transformed into competent E. coli cells including XL1-Blue, XL1-BlueMRF, or SOLR from Stratagene or DH5α, DH10B, or ElectroMAX DH10B from Life Technologies.


II. Isolation of cDNA Clones


Plasmids obtained as described in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Miniprep purification kit (Edge Biosystems, Gaithersburg Md.); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid purification systems or the R.E.A.L. PREP 96 plasmid purification kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4° C.


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


III. Sequencing and Analysis


Incyte cDNA recovered in plasmids as described in Example H were sequenced as follows. Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (Applied Biosystems) thermal cycler or the PTC-200 thermal cycler (MJ Research) in conjunction with the HYDRA microdispenser (Robbins Scientific) or the MICROLAB 2200 (Hamilton) liquid transfer system. cDNA sequencing reactions were prepared using reagents provided by Amersham Pharmacia Biotech or supplied in ABI sequencing kits such as the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems). Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were carried out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (Applied Biosystems) in conjunction with standard ABI protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, 1997, supra, unit 7.7). Some of the cDNA sequences were selected for extension using the techniques disclosed in Example VIII.


The polynucleotide sequences derived from Incyte cDNAs were validated by removing vector, linker, and poly(A) sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programming, and dinucleotide nearest neighbor analysis. The Incyte cDNA sequences or translations thereof were then queried against a selection of public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM; PROTEOME databases with sequences from Homo sapiens, Rattus norvegicus, Mus musculus, Caenorhabditis elegans, Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Candida albicans (Incyte Genomics, Palo Alto Calif.); and hidden Markov model (HMM)-based protein family databases such as PFAM. (HMM is a probabilistic approach which analyzes consensus primary structures of gene families. See, for example, Eddy, S. R. (1996) Curr. Opin. Struct. Biol. 6:361-365.) The queries were performed using programs based on BLAST, FASTA, BLIMPS, and HMMER. The Incyte cDNA sequences were assembled to produce full length polynucleotide sequences. Alternatively, GenBank cDNAs, GenBank ESTs, stitched sequences, stretched sequences, or Genscan-predicted coding sequences (see Examples IV and V) were used to extend Incyte cDNA assemblages to full length. Assembly was performed using programs based on Phred, Phrap, and Consed, and cDNA assemblages were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA. The full length polynucleotide sequences were translated to derive the corresponding full length polypeptide sequences. Alternatively, a polypeptide of the invention may begin at any of the methionine residues of the full length translated polypeptide. Full length polypeptide sequences were subsequently analyzed by querying against databases such as the GenBank protein databases (genpept), SwissProt, the PROTEOME databases, BLOCKS, PRINTS, DOMO, PRODOM, Prosite, and hidden Markov model (HMM)-based protein family databases such as PFAM. Full length polynucleotide sequences are also analyzed using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco Calif.) and LASERGENE software (DNASTAR). Polynucleotide and polypeptide sequence alignments are generated using default parameters specified by the CLUSTAL algorithm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences.


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


The programs described above for the assembly and analysis of full length polynucleotide and polypeptide sequences were also used to identify polynucleotide sequence fragments from SEQ ID NO:55-108. Fragments from about 20 to about 4000 nucleotides which are useful in hybridization and amplification technologies are described in Table 4, column 2.


IV. Identification and Editing of Coding Sequences from Genomic DNA


Putative secreted proteins were initially identified by running the Genscan gene identification program against public genomic sequence databases (e.g., gbpri and gbhtg). Genscan is a general-purpose gene identification program which analyzes genomic DNA sequences from a variety of organisms (See Burge, C. and S. Karlin (1997) J. Mol. Biol. 268:78-94, and Burge, C. and S. Karlin (1998) Curr. Opin. Struct. Biol. 8:346-354). The program concatenates predicted exons to form an assembled cDNA sequence extending from a methionine to a stop codon. The output of Genscan is a FASTA database of polynucleotide and polypeptide sequences. The maximum range of sequence for Genscan to analyze at once was set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode secreted proteins, the encoded polypeptides were analyzed by querying against PFAM models for secreted proteins. Potential secreted proteins were also identified by homology to Incyte cDNA sequences that had been annotated as secreted proteins. These selected Genscan-predicted sequences were then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences were then edited by comparison to the top BLAST hit from genpept to correct errors in the sequence predicted by Genscan, such as extra or omitted exons. BLAST analysis was also used to find any Incyte cDNA or public cDNA coverage of the Genscan-predicted sequences, thus providing evidence for transcription. When Incyte cDNA coverage was available, this information was used to correct or confirm the Genscan predicted sequence. Full length polynucleotide sequences were obtained by assembling Genscan-predicted coding sequences with Incyte cDNA sequences and/or public cDNA sequences using the assembly process described in Example III. Alternatively, full length polynucleotide sequences were derived entirely from edited or unedited Genscan-predicted coding sequences.


V. Assembly f Genomic Sequence Data with cDNA Sequence Data


“Stitched” Sequences


Partial cDNA sequences were extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example III were mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster was analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that were subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval was present on more than one sequence in the cluster were identified, and intervals thus identified were considered to be equivalent by transitivity. For example, if an interval was present on a cDNA and two genomic sequences, then all three intervals were considered to be equivalent. This process allows unrelated but consecutive genomic sequences to be brought together, bridged by cDNA sequence. Intervals thus identified were then “stitched” together by the stitching algorithm in the order that they appear along their parent sequences to generate the longest possible sequence, as well as sequence variants. Linkages between intervals which proceed along one type of parent sequence (cDNA to cDNA or genomic sequence to genomic sequence) were given preference over linkages which change parent type (cDNA to genomic sequence). The resultant stitched sequences were translated and compared by BLAST analysis to the genpept and gbpri public databases. Incorrect exons predicted by Genscan were corrected by comparison to the top BLAST hit from genpept. Sequences were further extended with additional cDNA sequences, or by inspection of genomic DNA, when necessary.


“Stretched” Sequences


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


VI. Chromosomal Mapping of SECP Encoding Polynucleotides


The sequences which were used to assemble SEQ ID NO:55-108 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith-Waterman algorithm. Sequences from these databases that matched SEQ ID NO:55-108 were assembled into clusters of contiguous and overlapping sequences using assembly algorithms such as Phrap (Table 7). Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Généthon were used to determine if any of the clustered sequences had been previously mapped. Inclusion of a mapped sequence in a cluster resulted in the assignment of all sequences of that cluster, including its particular SEQ ID NO:, to that map location.


Map locations are represented by ranges, or intervals, of human chromosomes. The map position of an interval, in centiMorgans, is measured relative to the terminus of the chromosome's p-arm. (The centiMorgan (cM) is a unit of measurement based on recombination frequencies between chromosomal markers. On average, 1 cM is roughly equivalent to 1 megabase (Mb) of DNA in humans, although this can vary widely due to hot and cold spots of recombination.) The cM distances are based on genetic markers mapped by Généthon which provide boundaries for radiation hybrid markers whose sequences were included in each of the clusters. Human genome maps and other resources available to the public, such as the NCBI “GeneMap'99” World Wide Web site (http://www.ncbi.nlm.nih.gov/genemap/), can be employed to determine if previously identified disease genes map within or in proximity to the intervals indicated above.


In this manner, SEQ ID NO:58 was mapped to chromosome 3 within the interval from 160.0 to 187.1 centiMorgans. SEQ ID NO:59 was mapped to chromosome 15 within the interval from 59.3 centiMorgans to the q-terminus. SEQ ID NO:60 was mapped to chromosome 15 within the interval from 39.5 to 59.3 centiMorgans. SEQ ID NO:61 was mapped to chromosome 3 within the interval from 67.9 to 77.4 centiMorgans. SEQ ID NO:62 was mapped to chromosome 16 at 473.44 centiMorgans. SEQ ID NO:63 was mapped to chromosome 9 within the interval from 75.8 to 136.7 centiMorgans. SEQ ID NO:64 was mapped to chromosome 19. SEQ ID NO:65 was mapped to chromosome 1 within the interval from 196.5 to 205.1 centiMorgans. SEQ ID NO:66 was mapped to chromosome 5 within the interval from 138.7 to 141.4 centiMorgans. SEQ ID NO:67 was mapped to chromosome 2 within the interval from 223.1 to 231.8 centiMorgans. SEQ ID NO:68 was mapped to chromosome 2 within the interval from 223.1 to 231.8 centiMorgans. SEQ ID NO:69 was mapped to chromosome 17 within the interval from 62.2 centiMorgans to the q-terminus. SEQ ID NO:75 was mapped to chromosome 15 within the interval from 59.3 centiMorgans to the q terminus. SEQ ID NO:76 was mapped to chromosome 13 within the interval from the p-terminus to 36.6 centiMorgans. SEQ ID NO:77 was mapped to the short arm of chromosome 8 within the cytogenetic band 23.3. SEQ ID NO:78 was mapped to chromosome 11 within the interval from 102.6 to 131.7 centiMorgans. SEQ ID NO:79 was mapped to chromosome 3 within the interval from 49.5 to 64.4 centiMorgans. SEQ ID NO:80 was mapped to chromosome 5 within the interval from 104.5 to 121.4 centiMorgans.


VII. Analysis of Polynucleotide Expression


Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (See, e.g., Sambrook, supra, ch. 7; Ausubel (1995) supra, ch. 4 and 16.)


Analogous computer techniques applying BLAST were used to search for identical or related molecules in cDNA databases such as GenBank or LIFESEQ (Incyte Genomics). This analysis is much faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or similar. The basis of the search is the product score, which is defined as:
BLASTScore×PercentIdentity5×minimum{length(Seq.1),length(Seq.2)}


The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. The product score is a normalized value between 0 and 100, and is calculated as follows: the BLAST score is multiplied by the percent nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences). The BLAST score is calculated by assigning a score of +5 for every base that matches in a high-scoring segment pair (HSP), and −4 for every mismatch. Two sequences may share more than one HSP (separated by gaps). If there is more than one HSP, then the pair with the highest BLAST score is used to calculate the product score. The product score represents a balance between fractional overlap and quality in a BLAST alignment. For example, a product score of 100 is produced only for 100% identity over the entire length of the shorter of the two sequences being compared. A product score of 70 is produced either by 100% identity and 70% overlap at one end, or by 88% identity and 100% overlap at the other. A product score of 50 is produced either by 100% identity and 50% overlap at one end, or 79% identity and 100% overlap.


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


VIII. Extension of SECP Encoding Polynucleotides


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


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


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


The concentration of DNA in each well was determined by dispensing 100 μl PICOGREEN quantitation reagent (0.25% (v/v) PICOGREEN; Molecular Probes, Eugene Oreg.) dissolved in 1×TE and 0.5 μl of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton Mass.), allowing the DNA to bind to the reagent. The plate was scanned in a Fluoroskan II (Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 μl to 10 μl aliquot of the reaction mixture was analyzed by electrophoresis on a 1% agarose gel to determine which reactions were successful in extending the sequence.


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


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


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


IX. Labeling and Use of Individual Hybridization Probes


Hybridization probes derived from SEQ ID NO:55-108 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments. Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 software (National Biosciences) and labeled by combining 50 pmol of each oligomer, 250 μCi of [γ-32P] adenosine triphosphate (Amersham Pharmacia Biotech), and T4 polynucleotide kinase (DuPont NEN, Boston Mass.). The labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech). An aliquot containing 107 counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl II, Eco RI, Pst I, Xba I, or Pvu II (DuPont NEN).


The DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schieicher & Schuell, Durham N-ti). Hybridization is carried out for 16 hours at 40° C. To remove nonspecific signals, blots are sequentially, washed at room temperature under conditions of up to, for example, 0.1× saline sodium citrate and 0.5% sodium dodecyl sulfate. Hybridization patterns are visualized using autoradiography or an alternative imaging means and compared.


X. Microarrays


The linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectric printing (inkjet printing, See, e.g., Baldeschweiler, supra.), mechanical microspotting technologies, and derivatives thereof. The substrate in each of the aforementioned technologies should be uniform and solid with a non-porous surface (Schena (1999), supra). Suggested substrates include silicon, silica, glass slides, glass chips, and silicon wafers. Alternatively, a procedure analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures. A typical array may be produced using available methods and machines well known to those of ordinary skill in the art and may contain any appropriate number of elements. (See, e.g., Schena, M. et al. (1995) Science 270:467-470; Shalon, D. et al. (1996) Genome Res. 6:639-645; Marshall, A. and J. Hodgson (1998) Nat. Biotechnol. 16:27-31.)


Full length cDNAs, Expressed Sequence Tags (ESTs), or fragments or oligomers thereof may comprise the elements of the microarray. Fragments or oligomers suitable for hybridization can be selected using software well known in the art such as LASERGENE software (DNASTAR). The array elements are hybridized with polynucleotides in a biological sample. The polynucleotides in the biological sample are conjugated to a fluorescent label or other molecular tag for ease of detection. After hybridization, nonhybridized nucleotides from the biological sample are removed, and a fluorescence scanner is used to detect hybridization at each array element. Alternatively, laser desorbtion and mass spectrometry may be used for detection of hybridization. The degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed. In one embodiment, microarray preparation and usage is described in detail below.


Tissue or Cell Sample Preparation


Total RNA is isolated from tissue samples using the guanidinium thiocyanate method and poly(A)+ RNA is purified using the oligo-(dT) cellulose method. Each poly(A)+ RNA sample is reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/μl oligo-(dT) primer (21mer), 1× first strand buffer, 0.03 units/μL RNase inhibitor, 500 μM dATP, 500 μM dGTP, 500 μM dTTP, 40 μM dCTP, 40 μM dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech). The reverse transcription reaction is performed in a 25 ml volume containing 200 ng poly(A)+ RNA with GEMBRIGHT kits (Incyte). Specific control poly(A)+ RNAs are synthesized by in vitro transcription from non-coding yeast genomic DNA. After incubation at 37° C. for 2 hr, each reaction sample (one with Cy3 and another with Cy5 labeling) is treated with 2.5 ml of 0.5M sodium hydroxide and incubated for 20 minutes at 85° C. to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (CLONTECH Laboratories, Inc. (CLONTECH), Palo Alto Calif.) and after combining, both reaction samples are ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The sample is then dried to completion using a SpeedVAC (Savant Instruments Inc., Holbrook N.Y.) and resuspended in 14 μl 5×SSC/0.2% SDS.


Microarray Preparation


Sequences of the present invention are used to generate array elements. Each array element is amplified from bacterial cells containing vectors with cloned cDNA inserts. PCR amplification uses primers complementary to the vector sequences flanking the cDNA insert. Array elements are amplified in thirty cycles of PCR from an initial quantity of 1-2 ng to a final quantity greater than 5 μg. Amplified array elements are then purified using SEPHACRYL400 (Amersham Pharmacia Biotech).


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


Array elements are applied to the coated glass substrate using a procedure described in U.S. Pat. No. 5,807,522, incorporated herein by reference. 1 μl of the array element DNA, at an average concentration of 100 ng/μl, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 nl of array element sample per slide.


Microarrays are UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene). Microarrays are washed at room temperature once in 0.2% SDS and three times in distilled water. Non-specific binding sites are blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (PBS) (Tropix, Inc., Bedford Mass.) for 30 minutes at 60° C. followed by washes in 0.2% SDS and distilled water as before.


Hybridization


Hybridization reactions contain 9 it of sample mixture consisting of 0.2 μg each of Cy3 and Cy5 labeled cDNA synthesis products in 5×SSC, 0.2% SDS hybridization buffer. The sample mixture is heated to 65° C. for 5 minutes and is aliquoted onto the microarray surface and covered with an 1.8 cm2 coverslip. The arrays are transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber is kept at 100% humidity internally by the addition of 140 μl of 5×SSC in a corner of the chamber. The chamber containing the arrays is incubated for about 6.5 hours at 60° C. The arrays are washed for 10 min at 45° C. in a first wash buffer (1×SSC, 0.1% SDS), three times for 10 minutes each at 45° C. in a second wash buffer (0.1×SSC), and dried.


Detection


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


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


The sensitivity of the scans is typically calibrated using the signal intensity generated by a cDNA control species added to the sample mixture at a known concentration. A specific location on the array contains a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100.000. When two samples from different sources (e.g., representing test and control cells), each labeled with a different fluorophore, are hybridized to a single array for the purpose of identifying genes that are differentially expressed, the calibration is done by labeling samples of the calibrating cDNA with the two fluorophores and adding identical amounts of each to the hybridization mixture.


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


A grid is superimposed over the fluorescence signal image such that the signal from each spot is centered in each element of the grid. The fluorescence signal within each element is then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis is the GEMTOOLS gene expression analysis program (Incyte).


XI. Complementary Polynucleotides


Sequences complementary to the SECP-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring SECP. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of SECP. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5′ sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the SECP-encoding transcript.


XII. Expression of SECP


Expression and purification of SECP is achieved using bacterial or virus-based expression systems. For expression of SECP in bacteria, cDNA is subcloned into an appropriate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcription. Examples of such promoters include, but are not limited to, the trp-lac (tac) hybrid promoter and the T5 or T7 bacteriophage promoter in conjunction with the lac operator regulatory element. Recombinant vectors are transformed into suitable bacterial hosts, e.g., BL21(DE3). Antibiotic resistant bacteria express SECP upon induction with isopropyl beta-D-thiogalactopyranoside (IPTG). Expression of SECP in eukaryotic cells is achieved by infecting insect or mammalian cell lines with recombinant Autopraphica californica nuclear polyhedrosis virus (AcMNPV), commonly known as baculovirus. The nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding SECP by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates. Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of cDNA transcription. Recombinant baculovirus is used to infect Snodoptera frugiperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases. Infection of the latter requires additional genetic modifications to baculovirus. (See Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. USA 91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7:1937-1945.)


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


XIII. Functional Assays


SECP function is assessed by expressing the sequences encoding SECP at physiologically elevated levels in mammalian cell culture systems. cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA expression. Vectors of choice include PCMV SPORT (Life Technologies) and PCR3.1 (Invitrogen, Carlsbad Calif.), both of which contain the cytomegalovirus promoter. 5-10 μg of recombinant vector are transiently transfected into a human cell line, for example, an endothelial or hematopoietic cell line, using either liposome formulations or electroporation. 1-2 μg of an additional plasmid containing sequences encoding a marker protein are co-transfected. Expression of a marker protein provides a means to distinguish transfected cells from nontransfected cells and is a reliable predictor of cDNA expression from the recombinant vector. Marker proteins of choice include, e.g., Green Fluorescent Protein (GFP; Clontech), CD64, or a CD64-GFP fusion protein. Flow cytometry (FCM), an automated, laser optics-based technique, is used to identify transfected cells expressing GFP or CD64-GFP and to evaluate the apoptotic state of the cells and other cellular properties. FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death. These events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide; changes in cell size and granularity as measured by forward light scatter and 90 degree side light scatter; down-regulation of DNA synthesis as measured by decrease in bromodeoxyuridine uptake; alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell surface. Methods in flow cytometry are discussed in Ormerod, M. G. (1994) Flow Cytometry, Oxford, New York N.Y.


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


XIV. Production of SECP Specific Antibodies


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


Alternatively, the SECP amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Methods for selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions are well described in the art. (See, e.g., Ausubel, 1995, supra, ch. 11.)


Typically, oligopeptides of about 15 residues in length are synthesized using an ABI 431A peptide synthesizer (Applied Biosystems) using FMOC chemistry and coupled to KLH (Sigma-Aldrich, St. Louis Mo.) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) to increase immunogenicity. (See, e.g., Ausubel, 1995, supra.) Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide and anti-SECP activity by, for example, binding the peptide or SECP to a substrate, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG.


XV. Purification of Naturally Occurring SECP Using Specific Antibodies


Naturally occurring or recombinant SECP is substantially purified by immunoaffinity chromatography using antibodies specific for SECP. An immunoaffinity column is constructed by covalently coupling anti-SECP antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.


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


XVI. Identification of Molecules WHICH Interact with SECP


SECP, or biologically active fragments thereof, are labeled with 125I Bolton-Hunter reagent. (See, e.g., Bolton, A. E. and W. M. Hunter (1973) Biochem. J. 133:529-539.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled SECP, washed, and any wells with labeled SECP complex are assayed. Data obtained using different concentrations of SECP are used to calculate values for the number, affinity, and association of SECP with the candidate molecules.


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


SECP may also be used in the PATHCALLING process (CuraGen Corp., New Haven Conn.) which employs the yeast two-hybrid system in a high-throughput manner to determine all interactions between the proteins encoded by two large libraries of genes (Nandabalan, K. et al. (2000) U.S. Pat. No. 6,057,101).


XVII. Demonstration of SECP Activity


An assay for growth stimulating or inhibiting activity of SECP measures the amount of DNA synthesis in Swiss mouse 3T3 cells (McKay, I. and Leigh, I., eds. (1993) Growth Factors: A Practical Approach, Oxford University Press, New York, N.Y.). In this assay, varying amounts of SECP are added to quiescent 3T3 cultured cells in the presence of [3]thymidine, a radioactive DNA precursor. SECP for this assay can be obtained by recombinant means or from biochemical preparations. Incorporation of [3H]thymidine into acid-precipitable DNA is measured over an appropriate time interval, and the amount incorporated is directly proportional to the amount of newly synthesized DNA. A linear dose-response curve over at least a hundred-fold SECP concentration range is indicative of growth modulating activity. One unit of activity per milliliter is defined as the concentration of SECP producing a 50% response level, where 100% represents maximal incorporation of [3H]thymidine into acid-precipitable DNA.


Alternatively, an assay for SECP activity measures the stimulation or inhibition of neurotransmission in cultured cells. Cultured CHO fibroblasts are exposed to SECP. Following endocytic uptake of SECP, the cells are washed with fresh culture medium, and a whole cell voltage-clamped Xenopus myocyte is manipulated into contact with one of the fibroblasts in SECP-free medium Membrane currents are recorded from the myocyte. Increased or decreased current relative to control values are indicative of neuromodulatory effects of SECP (Morimoto, T. et al. (1995) Neuron 15:689-696).


Alternatively, an assay for SECP activity measures the amount of SECP in secretory, membrane-bound organelles. Transfected cells as described above are harvested and lysed. The lysate is fractionated using methods known to those of skill in the art, for example, sucrose gradient ultracentrifugation. Such methods allow the isolation of subcellular components such as the Golgi apparatus, ER, small membrane-bound vesicles, and other secretory organelles. Immunoprecipitations from fractionated and total cell lysates are performed using SECP-specific antibodies, and immunoprecipitated samples are analyzed using SDS-PAGE and immunoblotting techniques. The concentration of SECP in secretory organelles relative to SECP in total cell lysate is proportional to the amount of SECP in transit through the secretory pathway.


Alternatively, AMP binding activity is measured by combining SECP with 32P-labeled AMP. The reaction is incubated at 37° C. and terminated by addition of trichloroacetic acid. The acid extract is neutralized and subjected to gel electrophoresis to remove unbound label. The radioactivity retained in the gel is proportional to SECP activity.


Alternatively, the activity of purified SECP can be tested by introducing the molecule into an in vitro production system for tissue plasminogen activator (tPA). Any statistically significant improvement of correctly folded tPA in the presence as compared to the absence of SECP would indicate that SECP is active and functioning correctly.


Alternatively, SECP activity may be measured by the enzymatic activity they possess. For SEQ ID NO:4, for example, SECP activity is measured as ferroxidase activity at pH 6 in 0.3 M acetate buffer. The appearance of ferric ions is monitored at 315 nm (Bonomi, F. et al. (1996) J. Biol. Inorg. Chem. 1:67-72). For SEQ ID NO:6, for example, SECP activity is measured by the phosphorylation of galactose. SECP is incubated for 5 minutes in a 100 μl reaction containing 200 μM 3H-galactose (30,000 cpm), 5 mM ATP, 5 mM MgCl2, 5 mM NaF, 100 mM Tris-HCl buffer, pH 8.5. The reaction is stopped by heating at 100° C. for 1 min, and the incubation mixture applied to a DE52 column. The column is washed with at least 5 column volumes of 10 mM (NH4)HCO3 to remove unbound material. Galactose-P is eluted with 500 mM (NH4)HCO3 and assayed for radioactive content by scintillation counting (Pastuszak, I. et al. (1996) J. Biol. Chem. 271:23653-23656). For SEQ ID NO:9, for example, SECP activity is measured by the amount of cobalamin bound using the isotope dilution method of Nexø, and Gimsing, employing human IF as the binding protein (1981, Scand. J. Clin. Lab. Invest. 41:465-468). For SEQ ID NO:10, for example, SECP activity is measured by the hydrolysis of appropriate synthetic peptide substrates conjugated with various chromogenic molecules in which the degree of hydrolysis is quantified by spectrophotometric (or fluorometric) absorption of the released chromophore (Beynon, R. J. and J. S. Bond (1994) Proteolytic Enzymes: A Practical Approach, Oxford University Press, New York, N.Y., pp. 25-55). Peptide substrates are designed according to the category of protease activity as endopeptidase (serine, cysteine, aspartic proteases, or metalloproteases), aminopeptidase (leucine aminopeptidase), or carboxypeptidase (carboxypeptidases A and B, procollagen C-proteinase). Commonly used chromogens are 2-naphthylamine, 4-nitroaniline, and furylacrylic acid. Assays are performed at ambient temperature and contain an aliquot of the enzyme and the appropriate substrate in a suitable buffer. Reactions are carried out in an optical cuvette, and the increase/decrease in absorbance of the chromogen released during hydrolysis of the peptide substrate is measured. The change in absorbance is proportional to the enzyme activity in the assay.


Alternatively, SECP activity can be measured as enzyme activity. For SEQ ID NO:20, for example, activity is proportional to the hydrolysis of glucosamine-6-sulfate by SECP which can be measured by the method of Robertson et al. (1992, Biochem. J. 288:539-544).


In another alternative, SECP can be assayed by its interaction with the insulin-like growth factor complex. For SEQ ID NO:17, for example, 125I-labeled SECP is incubated for 2 h with 10 ng of IGF-I or -II and a range from 0 to 10 ng of IGFBP-3 in 50 mM sodium phosphate buffer, pH 6.5, at 22° C. (final volume 0.3 ml). SECP complexed to IGFBP-3 is precipitated using IGFBP-3 antiserum and radioactivity in each tube measured (Janosi, J. B. M. et al. (1999) J. Biol. Chem. 274:5292-5298).


XVII. Demonstration of Immunoglobulin Activity


An assay for SECP activity measures the ability of SECP to recognize and precipitate antigens from serum. This activity can be measured by the quantitative precipitin reaction. (Golub, E. S. et al. (1987) Immunology: A Synthesis, Sinauer Associates, Sunderland, Mass., pages 113-115.) SECP is isotopically labeled using methods known in the art. Various serum concentrations are added to constant amounts of labeled SECP. SECP-antigen complexes precipitate out of solution and are collected by centrifugation. The amount of precipitable SECP-antigen complex is proportional to the amount of radioisotope detected in the precipitate. The amount of precipitable SECP-antigen complex is plotted against the serum concentration. For various serum concentrations, a characteristic precipitin curve is obtained, in which the amount of precipitable SECP-antigen complex initially increases proportionately with increasing serum concentration, peaks at the equivalence point, and then decreases proportionately with further increases in serum concentration. Thus, the amount of precipitable SECP-antigen complex is a measure of SECP activity which is characterized by sensitivity to both limiting and excess quantities of antigen.


Alternatively, an assay for SECP activity measures the expression of SECP on the cell surface. cDNA encoding SECP is transfected into a non-leukocytic cell line. Cell surface proteins are labeled with biotin (de la Fuente, M. A. et al. (1997) Blood 90:2398-2405). Immunoprecipitations are performed using SECP-specific antibodies, and immunoprecipitated samples are analyzed using SDS-PAGE and immunoblotting techniques. The ratio of labeled immunoprecipitant to unlabeled immunoprecipitant is proportional to the amount of SECP expressed on the cell surface.


Alternatively, an assay for SECP activity measures the amount of cell aggregation induced by overexpression of SECP. In this assay, cultured cells such as NIH3T3 are transfected with cDNA encoding SECP contained within a suitable mammalian expression vector under control of a strong promoter. Cotransfection with cDNA encoding a fluorescent marker protein, such as Green Fluorescent Protein (CLONTECH), is useful for identifying stable transfectants. The amount of cell agglutination, or clumping, associated with transfected cells is compared with that associated with untransfected cells. The amount of cell agglutination is a direct measure of SECP activity.


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

TABLE 1IncyteIncyteIncytePolypeptidePolypeptidePolynucleotidePolynucleotideProject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













TABLE 2









Incyte
GenBank ID NO:




Polypeptide
Polypeptide
or PROTEOME
Probability


SEQ ID NO:
ID
ID NO:
Score
Annotation



















1
95765CD1
g190183
2.6E−76
[Homo sapiens] opiomelanocortin






Krude, H. et al. (1998) Nature Genet. 19: 155-157


2
6399886CD1
g6996429
5.8E−262
dJ568C11.3 (novel AMP-binding enzyme similar to acetyl-coenzyme A






synthethase (acetate-coA ligase)) [Homo sapiens]






Luong, A. et al., (2000) J. Biol. Chem. 275: 26458-26466


3
6024420CD1
g163497
1.1E−70
[Bos taurus] PDI (E.C.5.3.4.1) (protein disulfide isomerase)






Yamauchi, K., et al. (1987) Biochem. Biophys. Res. Commun. 146, 1485-1492


4
7481067CD1
g180256
0
[Homo sapiens] preceruloplasmin (EC 1.16.3.1)






Waggoner, D. J. et al. (1999) Neurobiol. Dis. 9: 221-230;






Hellman, N. E. et al. (2000) Gut 47: 858-860


5
3378720CD1
g9859003
9.1E−65
[Homo sapiens] tumor metastasis-suppressor


16
1758413CD1
g183178
5.6E−106
[Homo sapiens] hGH-V2






Cooke, N. E. et al. (1988) J. Biol. Chem. 263: 9001-9006


17
7011042CD1
g184808
3.0E−11
[Homo sapiens] insulin-like growth factor binding protein complex,






acid-labile subunit






Leong, S. R. et al. (1992) Mol. Endocrinol. 6: 870-876


18
7427362CD1
g14530679
7.0E−70
WNT3A [Homo sapiens]






Saitoh, T., et al (2001) Biochem. Biophys. Res. Commun. 284: 1168-1175


19
7485304CD1
g2623871
2.1E−115
[Gallus gallus] Wnt-14 protein






Bergstein, I. et al. (1997) Genomics 46: 450-458


20
1422394CD1
g15430244
0
N-acetylglucosamine-6-sulfatase [Coturnix coturnix]






Dhoot, G. K., et al (2001) Science 293: 1663-6.


22
7473674CD1
g13620917
9.0E−46
mitochondrial ribosomal protein bMRP63 [Mus musculus]






Suzuki, T., et al (2001) J. Biol. Chem. 276: 33181-33195


26
7485334CD1
g10566471
1.2E−73
[Mus musculus] Gliacolin






Koide, T. et al. (2000) J. Biol. Chem. 275: 27957-27963


27
7220001CD1
g11071950
2.2E−115
[Mus musculus] (AB048834) Fca/m receptor






Shibuya, A., et al (2000) Nat. Immunol. 1: 441-446


28
5956275CD1
g7259265
4.4E−129
[Mus musculus] contains transmembrane (TM) region






Inoue, S., et al. (2000) Biochem. Biophys. Res. Commun. 268, 553-561


39
876751CD1
g15430246
0
nephronectin short isoform [Mus musculus]






Brandenberger, R., et al (2001) J. Cell Biol. 154: 447-458


40
2512510CD1
g14423349
0
membrane glycoprotein LIG-1 [Homo sapiens]


41
7486326CD1
g3822218
0
[Homo sapiens] chordin






Pappano, W. N. et al. (1998) Genomics 52: 236-239


48
2902793CD1
g15026974
2.0E−36
obscurin [Homo sapiens]






Young, P., et al (2001) J. Cell Biol. 154: 123-136


54
7503512CD1
g14423349
0
membrane glycoprotein LIG-1 [Homo sapiens]






















TABLE 3










Amino
Potential
Potential

Analytical


SEQ
Incyte
Acid
Phosphoryl-
Glycosy-

Methods


ID
Polypeptide
Resi-
ation
lation

and


NO:
ID
dues
Sites
Sites
Signature Sequences, Domains and Motifs
Databases





















1
095765CD1
235
S92 S211 Y189
N91
signal_cleavage: M1-E23
SPSCAN







Signal Peptide:
HMMER







M1-E23, M1-G26, M1-V24, M1-C28, M1-E30







Corticotropin ACTH domain: S106-F144, P188-T220
HMMER_PFAM







Transmembrane Domain: C6-W27
TMAP







N-terminus is non-cytosolic







Pro-opiomelanocortin signature
BLIMPS_PRINTS







PR00383: Y107-P117, V118-E133, D134-E143,







A181-W196, W196-M209







PRECURSOR CORTICOTROPIN LIPOTROPIN
BLAST_PRODOM







PRO-OPIOMELANOCORTIN POMC HORMONE







SIGNAL ENDORPHIN CLEAVAGE ON PAIR







PD004218: L12-Q68







PRO-OPIOMELANOTROPIN POM PRECURSOR
BLAST_PRODOM







SIGNAL







PD116389: R101-N229, C28-W84







HORMONE PRECURSOR CORTICOTROPIN
BLAST_PRODOM







LIPOTROPIN PRO-OPIOMELANOCORTIN POMC







CLEAVAGE ON PAIR OF BASIC







PD003250: S106-F144







CORTICOTROPIN LIPOTROPIN PRECURSOR
BLAST_PRODOM







PRO-OPIOMELANOCORTIN POMC ENDORPHIN







HORMONE CLEAVAGE ON PAIR OF







PD029102: D164-N229







CORTICOTROPIN-LIPOTROPIN
BLAST_DOMO







DM01793|P01190|1-83: M1-W84







CORTICOTROPIN-LIPOTROPIN
BLAST_DOMO







DM00964|P01190|171-264: K145-E235







CORTICOTROPIN-LIPOTROPIN
BLAST_DOMO







DM01793|P19402|1-79: M1-V78







CORTICOTROPIN-LIPOTROPIN
BLAST_DOMO







DM00964|PN0130|13-90: A167-E235


2
6399886CD1
689
S16 S409 S415
N109 N492
Signal Peptide: M1-A25
HMMER





S486 S601 S640
N554





T89 T97 T142





T150 T156 T318





T438 T546 T663





T664







AMP-binding enzyme: T142-V580
HMMER_PFAM







Transmembrane Domains:
TMAP







P173-A201, E463-P478







N-terminus is cytosolic







Putative AMP-binding domain signature: A272-V322
PROFILESCAN







Putative AMP-binding domain
BLIMPS_BLOCKS







BL00455: Y293-Q308







AMP-binding signature
BLIMPS_PRINTS







PR00154: E286-S297, T298-H306







LIGASE SYNTHETASE PROTEIN ENZYME
BLAST_PRODOM







BIOSYNTHESIS ANTIBIOTIC







PHOSPHOPANTETHEINE MULTIFUNCTIONAL







REPEAT ACYL-COA







PD000070: T142-T438, D482-I581







SYNTHETASE LIGASE ACETYL COENZYME A
BLAST_PRODOM







ENZYME ACETATE-COA ACETYL-COA







PROTEIN ACYL ACTIVATING







PD009307: Y58-L146







PUTATIVE AMP-BINDING DOMAIN
BLAST_DOMO







DM00073







S46276|96-631: L115-R649







P27550|82-615: L115-R649







S52154|15-546: I141-L648







P16928|102-634: L115-R649







Putative AMP-binding domain signature
MOTIFS







M291-K302


3
6024420CD1
584
S20 S71 S134
N58 N160
signal_cleavage: M1-S20
SPSCAN





S153 S358 S376
N238 N340





S417 S546 S558
N370 N436





T128 T241 T329
N540





T448 T481 T495





T557 Y356







Signal Peptide: M1-S20; M1-E23, M1-A26
HMMER







Thioredoxin domain: Q386-D451
HMMER_PFAM







Transmembrane Domain: P199-H216
TMAP







N-terminal is non-cytosolic







Thioredoxin family active site: L392-I440
PROFILESCAN







Thioredoxin family proteins signature
BLIMPS_BLOCKS







BL00194: F409-K421







ISOMERASE PRECURSOR PROTEIN SIGNAL
BLAST_PRODOM







REDOX ACTIVE CENTER DISULFIDE







ENDOPLASMIC RETICULUM REPEAT







PD001708: Q55-L367







REDOX ACTIVE CENTER PROTEIN
BLAST_PRODOM







ISOMERASE PRECURSOR THIOREDOXIN







SIGNAL DISULFIDE ENDOPLASMIC







ELECTRON







PD000175: V389-K496







PROTEIN DISULFIDE-ISOMERASE
BLAST_DOMO







DM00799|P09102|112-347: L148-D385







DM00799|P54399|129-364: E167-D385







DM00799|A54757|127-358: L148-D385







THIOREDOXIN FAMILY
BLAST_DOMO







DM00054|P09102|349-452: V389-L489


4
7481067CD1
1049
S38 S180 S221
N111 N137
signal_cleavage: M1-A19
SPSCAN





S241 S317 S318
N169 N245





S383 S551 S604
N270 N392





S638 S736 T155
N460 N475





T184 T198 T203
N549 N575





T208 T222 T311
N781 N829





T389 T414 T430
N830 N908





T452 T515 T570





T589 T661 T688





T783 T855 T1007





T1037 Y134 Y483





Y566 Y715







Signal Peptide: M1-P16, M1-W18, M1-A19
HMMER







Transmembrane Domain: I794-I809
TMAP







Multicopper oxidase: L900-R1041, L567-C705,
HMMER_PFAM







L218-C352







Multicopper oxidases signatures
PROFILESCAN







multicopper_oxidase1.prf: D310-V366, D660-E742,







D996-L1050







multicopper_oxidase2.prf: P1001-N1048







Multicopper oxidases signature 1: G680-Y700,
MOTIFS







G1016-Y1036







Multicopper oxidases signature
BLIMPS_BLOCKS







BL00079A: G93-N110, R817-T828, L975-F985,







D1014-M1032







FERROXIDASE REPEAT
BLAST_DOMO







DM00956|P00450|90-336: L89-L334, L438-L684,







P838-L1019







DM00956|P00450|445-697: H437-L684, L89-S318,







Y819-W1018,







H791-S831 DM00956|P00450|804-1038:







H791-Y1021, H437-G680, L89-L334







DM00956|P12259|83-308: G90-P249, G439-S577,







I794-V992,







E252-L334, D595-T675, S538-L552







FACTOR PRECURSOR GLYCOPROTEIN
BLAST_PRODOM







PLASMA REPEAT SIGNAL COAGULATION







BLOOD VIII CALCIUM







PD002090: H368-K559, R22-N205,







K707-P893







ATP/GTP-binding site motif A (P-loop):
MOTIFS







G335-S342


5
3378720CD1
383
S23 S90 S119
N107
signal_cleavage: M1-E57
SPSCAN





S247 S334 S340





T4 T19 T73





T78 T81 T109





Y345







Transmembrane Domains:
TMAP







H38-A61, C136-Y160, Y181-F198,







K201-R229, N261-F281, H294-L314







N terminus is non-cytosolic







PROTEIN TRANSMEMBRANE LONGEVITY
BLAST_PRODOM







ASSURANCE FACTOR UOG1 SIMILAR S







CEREVISIAE







PD006418: S119-L369


6
938824CD1
72


signal_cleavage: M1-A63
SPSCAN







Signal Peptide: M1-G29
HMMER







Transmembrane Domain: P9-A36
TMAP







N terminus is non-cytosolic







Galactokinase signature
PROFILESCAN







galactokinase.prf: S17-A67


7
1683721CD1
91
S41 T34 T53

signal_cleavage: M1-P23
SPSCAN







Signal Peptide: M1-P23
HMMER


8
1694122CD1
160
T19

signal_cleavage: M1-R62
SPSCAN


9
1970615CD1
95
S55

signal_cleavage: M1-A26
SPSCAN







Signal Peptide:
HMMER







M1-A26, M1-R27, M1-V28, M1-C30







Transmembrane Domain: Q4-C20
TMAP







N-terminus is non-cytosolic







Eukaryotic cobalamin-binding proteins signature
PROFILESCAN







cobalamin_binding.prf: D25-R72


10
2314152CD1
92
S11 S23 S33

signal_cleavage: M1-S19
SPSCAN





S52 T65 Y43







Signal Peptide: M1-R25, M1-S24
HMMER







Endopeptidase Clp active sites
PROFILESCAN







clpp_protease_ser.prf:







L32-G80


11
2886225CD1
71
S41 T37 Y67

signal_cleavage: M1-L29
SPSCAN







Signal Peptide: M1-L29
HMMER







Transmembrane Domain: H15-C33
TMAP







N-terminus is non-cytosolic







Vitamin K-dependent carboxylation domain
PROFILESCAN







glu_carboxylation.prf: M1-G70


12
6144418CD1
100
S46 S69 T50
N67 N85
signal_cleavage: M1-G36
SPSCAN





T60 T84







Signal Peptide: M1-A29, M1-V31
HMMER







Transmembrane Domain: L8-G36
TMAP







N-terminus is non-cytosolic


13
6834184CD1
122

N108
signal_cleavage: M1-E38
SPSCAN







Signal Peptide: M1-G25
HMMER







Transmembrane Domain: G8-T36, C81-I105
TMAP







N-terminus is non-cytosolic


14
6951005CD1
113
S25 T35 Y81

signal_cleavage: M1-D28
SPSCAN







Signal Peptide: M1-Q29, M1-D28
HMMER







Transmembrane Domain: S50-S72
TMAP







N-terminus is non-cytosolic


15
7250331CD1
85


signal_cleavage: M1-L27
SPSCAN







Signal Peptide:
HMMER







M1-P23, M1-V25, M1-R26, M1-L27, M1-C35







Immunoglobulins and major histocompatibility
PROFILESCAN







complex proteins signature ig_mhc.prf: L14-L64


16
1758413CD1
256
S88 S97 S111

signal_cleavage: M1-A26
SPSCAN





S132 S181





S194 T53







Signal Peptide: M1-A26
HMMER







Transmembrane Domain: T7-L35
TMAP







N-terminus is non-cytosolic







Somatotropin hormone family: L9-M151
HMMER_PFAM







Somatotropin, prolactin and related hormones
PROFILESCAN







somatotropin_1.prf: S88-H138







Somatotropin, prolactin and related hormones
MOTIFS







signature 1: C79-W112







Somatotropin, prolactin and related hormones
BLIMPS_BLOCKS







BL00266: L35-Y61, C79-V116, D135-M151,







P201-P224







Somatotropin hormone family signature PR00836:
BLIMPS_PRINTS







C79-E92, L101-L119







GROWTH HORMONE VARIANT II PRECURSOR
BLAST_PRODOM







GHV2 PLACENTA SIGNAL ALTERNATIVE







SPLICING







PD084405: P165-V256







HORMONE PRECURSOR SIGNAL PITUITARY
BLAST_PRODOM







GROWTH SOMATOTROPIN PROLACTIN







GLYCOPROTEIN PRL







PD000259: T7-M151







SOMATOTROPIN, PROLACTIN AND RELATED
BLAST_DOMO







HORMONES







DM00125|P09587|17-227: C17-P228







DM00125|P01243|17-212: C17-G163







DM00125|I67409|17-212: C17-G163







DM00125|P01242|17-212: C17-G163


17
7011042CD1
287
S16 S46 S91
N42 N176
signal_cleavage: M1-A18
SPSCAN





S147 T76 T105





T214 T242







Signal Peptide: M1-A18, M1-W17
HMMER







Leucine Rich Repeat: N254-P278, Q100-A122,
HMMER_PFAM







A171-D199, T205-P228, G123-S146, T76-E99,







S147-P170, K229-P253







Leucine Rich Repeat PR00019A: L124-L137
BLIMPS_PRINTS







Leucine zipper pattern: L59-L80, L66-L87
MOTIFS


18
7427362CD1
366
S89 T34 T81
N103
signal_cleavage: M1-A29
SPSCAN





T150 T215 T226





T257 T356 T363







Signal Peptide: M1-A29
HMMER







Transmembrane Domain: P6-L33
TMAP







N-terminus is non-cytosolic







wnt family of developmental signaling proteins:
HMMER_PFAM







A58-K365







Wnt-1 family signature
PROFILESCAN







wnt1.prf: M196-K245







Wnt-1 family signature: C216-C225
MOTIFS







Wnt-1 family proteins
BLIMPS_BLOCKS







BL00246: M196-Y248, N319-C364, A85-C104,







G118-D152, W163-E187







DEVELOPMENTAL GLYCOPROTEIN
BLAST_PRODOM







PRECURSOR SIGNAL WNT1 WNT5A WNT2







EXTRACELLULAR MATRIX







PD000810: C59-A195, E153-K365







WNT-1 FAMILY
BLAST_DOMO







DM00403|P27467|22-351: K67-K365







DM00403|P21551|32-368: L64-C364







DM00403|P28465|13-351: C59-K365







DM00403|P22727|24-363: A154-C364


19
7485304CD1
416
S72 S120 S228
N158
wnt family of developmental signaling proteins:
HMMER_PFAM





S238 T87 T136

Q113-K415





T203 T268 T413





Y342







Wnt-1 family signature wnt1.prf: D250-K298
PROFILESCAN







Wnt-1 family signature: C269-C278
MOTIFS







Wnt-1 family proteins
BLIMPS_BLOCKS







BL00246: A140-C159, G171-D205, W216-R240,







A249-Y301, S369-C414







PROTEIN DEVELOPMENTAL GLYCOPROTEIN
BLAST_PRODOM







PRECURSOR SIGNAL WNT1 WNT5A WNT2







EXTRACELLULAR MATRIX







PD000810: L119-A251, D206-K415







WNT-1 FAMILY
BLAST_DOMO







DM00403|P49340|30-391: L119-S365, A334-C414,







S69-Q106







DM00403|P21551|32-368: L119-C414







DM00403|P47793|24-351: C114-K415







DM00403|P22727|24-363: E211-L410, K118-G209


20
1422394CD1
871
S27 S206 S288
N64 N111
signal_cleavage: M1-C22
SPSCAN





S425 S468 S488
N131 N148





S505 S516 S520
N170 N197





S635 T24 T66
N240 N623





T96 T107 T367
N773 N783





T376 T400 T452





T484 T530 T611





T615 T781 Y645







Signal Peptide: M1-C22
HMMER







Transmembrane Domain: C5-L21, S353-D370
TMAP







N-terminus is cytosolic







Sulfatase: P43-T467
HMMER_PFAM







Sulfatases signature 1: P85-G97
MOTIFS







Sulfatases proteins
BLIMPS_BLOCKS







BL00523: P43-S59, C87-K98, G134-L144,







P214-H225, V289-G318, D363-G373, Y800-Q809







ARYLSULFATASE SIGNAL GLYCOPROTEIN
BLAST_PRODOM







LYSOSOME SULPHOHYDROLASE







MUCOPOLYSACCHARIDOSIS







PD001700: P43-I200, Q279-P378, K330-E392







N-ACETYLGLUCOSAMINE-6-SULFATASE
BLAST_PRODOM







PRECURSOR GLUCOSAMINE-6-SULFATASE







HYDROLASE LYSOSOME SIGNAL







GLYCOPROTEIN MUCOPOLYSACCHARIDOSIS







PD022780: C766-G837







SIMILAR TO SULFATASE
BLAST_PRODOM







PD122645: V385-Q487, H700-F765







ARYLSULFATASE; SULFATASE; PLANT;
BLAST_DOMO







DM08669|Q10723|23-520: R280-R490, R42-Q279,







N773-P843







DM08669|P14217|24-519: L278-G373, R42-P269,







S770-P843, E294-Y310







DM01026|P08842|24-548: V289-D390, R42-Y143







DM01026|P50473|63-522: E273-L389, R42-P153


21
1336022CD1
100
S3 S72 S93
N5
signal_cleavage: M1-V24
SPSCAN







Signal Peptide: M1-V24
HMMER







Transmembrane Domain: V4-P23
TMAP







N-terminus is non-cytosolic


22
7473674CD1
102
S28 S102 T79
N96
signal_cleavage: M1-G19
SPSCAN





T98


23
7475846CD1
117


signal_cleavage: M1-C32
SPSCAN


24
7475860CD1
150
S65 T58 T99
N46 N66
signal_cleavage: M1-A34
SPSCAN





T113 T138 Y143


25
7950941CD1
89
S10 S34 S39 S49

Signal Peptide: M1-G24
HMMER


26
7485334CD1
287
S111 S167 S255

signal_cleavage: M1-A15
SPSCAN





T28 T142 T192







Signal Peptide: M1-A20, M1-A21
HMMER







C1q domain: A160-L284
HMMER_PFAM







C1q domain signature: F177-Y207
MOTIFS







C1q domain proteins
BLIMPS_BLOCKS







BL01113: V174-I209, D243-K262, S277-P286,







G85-S111







Complement C1Q domain signature
BLIMPS_PRINTS







PR00007: P168-K194, F195-G214, D243-D264,







K275-Y285







Collagen triple helix repeat (20 copies):
HMMER_PFAM







P71-V129







Lysosome-associated membrane glycoproteins
PROFILESCAN







lamp_2.prf: E145-L175







PRECURSOR SIGNAL COLLAGEN REPEAT
BLAST_PRODOM







HYDROXYLATION GLYCOPROTEIN CHAIN







PLASMA EXTRACELLULAR MATRIX







PD002992: P168-L284







COLLAGEN ALPHA PRECURSOR REPEAT
BLAST_PRODOM







SIGNAL CONNECTIVE TISSUE







EXTRACELLULAR MATRIX







PD000007: G43-G118







PROCOLLAGEN TYPE XVII ALPHA 1 BULLOUS
BLAST_PRODOM







PEMPHIGOID AUTO-ANTIGEN CELL







ADHESION







PD071338: G43-G112







PRECOLLAGEN P PRECURSOR SIGNAL
BLAST_PRODOM







PD072959: G43-G128







C1Q DOMAIN
BLAST_DOMO







DM00777|S23297|465-674: P84-L283







DM00777|P23206|477-673: G88-P286







DM00777|P98085|222-418: G85-D287







DM00777|P27658|551-743: G88-P286


27
7220001CD1
578
S39 S108 S189
N212 N322
signal_cleavage: M1-A61
SPSCAN





S252 S297 S302





S406 S483 S494





S526 T6 T38 T88





T234 T272 T336





T350 T351 T438





T487 T525 T570





Y24







Signal Peptide: M46-A61, M46-P63, M46-Q64
HMMER







Immunoglobulm domain: G120-I200
HMMER_PFAM







Transmembrane Domain: S39-P67 R496-R518
TMAP







N-terminus is cytosolic







POLYMERIC IMMUNOGLOBULIN RECEPTOR
BLAST_PRODOM







PRECURSOR PLGR CONTAINS: SECRETORY







COMPONENT IMMUNOGLOBULIN FOLD







REPEAT







PD003917: G120-E203







(P-value = 5.4e−09)







IMMUNOGLOBULIN
BLAST_DOMO







DM00001|P01833|41-120: H128-G201







DM00001|P15083|41-120: H128-F208







DM00001|P01832|28-125: G120-G201







DM00001|S48841|41-120: H128-G201


28
5956275CD1
285
S109 S133 S256

signal_cleavage: M1-A28
SPSCAN





T38 T91 T100





T191 Y125







Signal Peptide: M1-A28
HMMER







Immunoglobulin domain: G42-V129
HMMER_PFAM







Transmembrane Domain: Q3-T31
TMAP







N-terminus is non-cytosolic







Cell attachment sequence: R197-D199
MOTIFS


29
346472CD1
72
S25

signal_cleavage: M1-C14
SPSCAN







Signal Peptide: M1-S16, M1-S20, M1-N21, M1-F22,
HMMER







M1-H24, M1-S25, M1-K27







Aminotransferases class-V pyridoxal-phosphate
PROFILESCAN







attachment site: S25-S72


30
643526CD1
72


signal_cleavage: M1-C18
SPSCAN







Signal Peptide: M1-C18, M1-S19, M1-E20, M1-S21,
HMMER







M1-G23, M1-S24, M1-P26







Transmembrane Domain: S24-V41
TMAP


31
1483418CD1
149
S65 S70

signal_cleavage: M1-H29
SPSCAN







Signal Peptide: M1-G32, M1-V34, M1-S35
HMMER







Transmembrane Domain: G32-P53
TMAP







N-terminus is non-cytosolic


32
2683477CD1
100
S71 T54 T81
N69
Signal Peptide: M7-L35
HMMER







Transmembrane Domain: S13-R38
TMAP







N-terminus is non-cytosolic


33
5580991CD1
78
S50

signal_cleavage: M1-C24
SPSCAN







Signal Peptide: M4-S22, M4-C24, M4-P25, M4-S26,
HMMER







M4-S29







Transmembrane Domain: F13-F41
TMAP







N-terminus is non-cytosolic







CBF/NF-Y subunits signatures: M4-S73
PROFILESCAN


34
5605931CD1
75
S65

Signal Peptide: M29-A43
HMMER







Signal Peptide: M1-Q28
HMMER







Transmembrane Domain: A5-F25
TMAP







N-terminus is non-cytosolic


35
6975241CD1
111
S6 S51 S75

signal_cleavage: M1-E22
SPSCAN







Signal Peptide: M1-E22, M1-K24, M1-K24, M1-G28,
HMMER







M1-I30


36
6988529CD1
72
S30 T35

signal_cleavage: M1-A15
SPSCAN







Signal Peptide: M1-A15, M1-T17, M1-A20
HMMER


37
6996808CD1
90
S34 S42 S68

signal_cleavage: M1-C45
SPSCAN







Signal Peptide: M26-C45, M17-L38, M17-S41
HMMER







Transmembrane Domains: Q12-F40, R44-I72
TMAP







N-terminus is non-cytosolic


38
7472689CD1
283
S66 S140 S197
N131
Signal Peptide: M21-G38
HMMER





T259


39
876751CD1
566
S111 S238 S407
N274
Signal_Peptide: M1-A19
SPSCAN





S485 S556 T167





T176 T308 T312





T316 T320 T346





T527 Y206







Signal Peptide: M1-A18, M1-E20
HMMER







EGF-Iike domain: C94-C128, C174-C213,
HMMER_PFAM







C219-C254, C134-C168, C61-C87







MAM domain: C423-E564
HMMER_PFAM







Anaphylatoxin domain proteins
BLIMPS_BLOCKS







BL01177: R99-L117, P163-S180







Calcium-binding EGF-like domain
BLIMPS_BLOCKS







BL01187: C105-Y120, C168-A179







PRECURSOR GLYCOPROTEIN SIGNAL
BLAST_PRODOM







TRANSMEMBRANE HYDROLASE PROTEIN







REPEAT RECEPTOR PHOSPHATASE







NEUROPILIN







PD001482: W432-E564







PROLINE-RICH PROTEIN 3
BLAST_DOMO







DM00215|P41479|30-111: P297-I369







Cell attachment sequence: R383-D385
MOTIFS







Aspartic acid and asparagine hydroxylation site:
MOTIFS







C105-C116 C187-C198 C232-C243







EGF-like domain signature 1: C76-C87
MOTIFS







EGF-like domain signature 2: C76-C87 C114-C128
MOTIFS







C241-C254







Calcium-binding EGF-like domain pattern signature:
MOTIFS







D90-C114 D170-C196 D215-C241


40
2512510CD1
1093
S81 S270 S326
N74 N150
Signal_Peptide: M1-A33
SPSCAN





S356 S379 S403
N246 N292





S473 S588 S705
N318 N684





S734 S822 S850





S970 S987 S998





T192 T214 T320





T370 T499 T538





T604 T609 T688





T736 T771 T818





T828 T848 T936





Y670







Signal Peptide: M1-A33, M1-A34
HMMER







Leucine Rich Repeat: S332-R355, S236-S259,
HMMER_PFAM







K308-S331, R212-N235, G407-K430, S356-D382,







N93-S114, S189-P211, K260-T283, S383-E406,







P164-S187, S140-P163, W69-P92, A284-Q307,







H116-K136







Leucine rich repeat C-terminal domain: D440-D490
HMMER_PFAM







Immunoglobulin domain: G707-M765, T613-A674,
HMMER_PFAM







G509-I579







Transmembrane Domain: R13-A33
TMAP







N terminus non-cytosolic







Bee Venom Hyaluronidase
BLIMPS_PRINTS







PR00847F: R738-V754







Leucine-rich repeat signature
BLIMPS_PRINTS







PR00019 L91-L104, L141-V154







MEMBRANE GLYCOPROTEIN
BLAST_PRODOM







PD129774: M765-S1093







PD172109: D491-F583







PD165826: E29-T70







SIMILARITY MULTIPLE LEUCINE RICH
BLAST_PRODOM







PD037237: L432-I610







IMMUNOGLOBULIN
BLAST_DOMO







DM00001|P35918|668-745: L699-A774







DM00001|A46065|668-745: L699-A774







Leucine zipper pattern: L52-L73 L59-L80
MOTIFS


41
7486326CD1
915
S38 S98 S130
N217 N351
Signal_Peptide: M1-P23
SPSCAN





S132 S151 S153
N365 N434





S195 S201 S245





S306 S353 S488





S596 S751 S882





T304 T367 T592





T775 T859 T902





T904 Y152 Y745







Signal Peptide: M1-P23, M1-G26
HMMER







von Willebrand factor (growth regulator) type C
HMMER_PFAM







domain: C51-C125, C832-C892, C744-C810,







C665-C722







Transmembrane Domain: P5-R25
TMAP







N terminus non-cytosolic







CHORDIN
BLAST_PRODOM







PD018424: Q440-L618, G232-Y439, F141-P292,







A413-V605







PD069130: P811-C892PD015143: P662-D727







VON WILLEBRAND FACTOR TYPE C REPEAT
BLAST_DOMO







DM00551|A55195|22-117: P31-P126







DM00551|A55195|752-835: E724-K808, P656-C719







DM00551|A55195|637-751: R660-C722, R738-C810,







T592-L618







DM00551|A55195| 836-920: Q809-C892







Cell attachment sequence R165-D167
MOTIFS







VWFC domain signature: C686-C722 C771-C810
MOTIFS







Leucine zipper pattern: L315-L336
MOTIFS


42
1221545CD1
113
S66 S71 T44
N88
Signal_Peptide: M1-T45
SPSCAN





T100







Signal Peptide: M1-D18, M1-G20
HMMER







Transmembrane Domain: T4-R24
TMAP







N terminus cytosolic


43
124737CD1
91
S30 S43

Signal_Peptide: M1-A26
SPSCAN







Signal Peptide: M1-A26, M1-L31
HMMER


44
1510784CD1
83
S33 S61 S71

Signal_Peptide: M1-S61
SPSCAN







Signal Peptide: M1-H17, M3-P23
HMMER







Defensin signature: PR00217A L58-C67
BLIMPS_PRINTS


45
1901257CD1
128
S28 T31 T38

Signal_Peptide: M1-S27
SPSCAN







Signal Peptide: M7-S34, M46-S74
HMMER







Transmembrane Domain: A4-A25
TMAP







N terminus non-cytosolic


46
2044370CD1
84
S31 S49 T22
N38
Signal_Peptide: M1-G21
SPSCAN







Signal Peptide: M1-G21
HMMER







Transmembrane Domain: L3-Y23
TMAP







Sushi (SCR complement) domain:
BLIMPS_PFAM







PF00084A: C69-P73


47
2820933CD1
109
S33 S52 S97

Signal_Peptide: M1-S21
SPSCAN







Signal Peptide: M1-S21, M1-E31
HMMER


48
2902793CD1
159
S85 S154 T122

Signal_Peptide: M1-A23
SPSCAN







Signal Peptide: M1-C25
HMMER







Immunoglobulin domain: G54-C112
HMMER_PFAM







Transmembrane Domain: V4-S21
TMAP







N terminus non-cytosolic


49
7486536CD1
242
S86 S100 S104
N76
Signal Peptide: M1-L19, M1-N21
HMMER





S115 S139 S229





T30 T134 Y176







Transmembrane Domain: T6-L29
TMAP







N terminus non-cytosolic


50
8137305CD1
542
S74, S90, S95,
N41, N333
Signal cleavage: M1-F28
SPSCAN





S111, S126,





T225, Y245,





T231, S320,





S338, S379,





S424, T536







Signal Peptide: M1-C34
HMMER







Transmembrane Domain: I7-L32;
TMAP







N-terminus is non-cytoplasmic


51
3793128CD1
105
S30, T37
N55
Signal cleavage: M1-R22
SPSCAN







Signal Peptide: M1-S19
HMMER







Transmembrane Domain: P7-Y34;
TMAP







N-terminus is non-cytoplasmic


52
4001243CD1
102


Signal Peptide: M76-A95, M1-A27
HMMER


53
6986717CD1
129
S31, S77, T125
N83
Signal Peptide: M25-R48, M25-A53, M25-Q51
HMMER







Transmembrane Domain: P4-R19;
TMAP







N-terminus is non-cytoplasmic


54
7503512CD1
1070
S81 S247 S303
N74 N150
signal_cleavage: M1-A33
SPSCAN





S333 S356 S380
N223 N269





S450 S565 S682
N295 N661





S711 S799 S827





S947 S964 S975





T192 T297 T347





T476 T515 T581





T586 T665 T713





T748 T795 T805





T825 T913 Y647







Signal Peptide: M1-A33, M1-A34
HMMER







Leucine Rich Repeat: S309-R332, S213-S236,
HMMER_PFAM







K285-S308, S333-S356, G384-K407, N93-S114,







W69-P92, K237-T260, S360-E383, P164-S187,







S140-P163, S189-N212, A261-Q284, H116-K136







Leucine rich repeat C-terminal domain:
HMMER_PFAM







D417-D467







Leucine rich repeat N-terminal domain:
HMMER_PFAM







P40-P67







Immunoglobulin domain: G684-M742, T590-A651,
HMMER_PFAM







G486-I556







Cytosolic domain: Y793-S1070
TMHMMER







Transmembrane domain: V770-I792







Non-cytosolic domain: M1-T769







RECEPTOR INTERLEUKIN-1 P
BLIMPS_PRODOM







PD02870: F634-V666, L725-P759







MEMBRANE GLYCOPROTEIN
BLAST_PRODOM







PD129774: M742-S1070







PD172109: D468-F560







PD165826: E29-T70







KEK1 PRECURSOR T21D12.9 SPLICING SIGNAL
BLAST_PRODOM







ALTERNATIVE KEK2







PD037237: L409-I587







IMMUNOGLOBULIN
BLAST_DOMO







DM00001|P35918|668-745: L676-A751







DM00001|A46065|668-745: L676-A751







Leucine zipper pattern: L52-L73, L59-L80
MOTIFS

















TABLE 4








Polynucleotide



SEQ ID NO:/


Incyte ID/


Sequence Length
Sequence Fragments







55/095765CB1/1315
1-68, 1-341, 1-346, 1-359, 1-365, 1-367, 1-435, 1-566, 1-589, 1-625,



1-647, 2-367, 3-365, 3-367, 4-367, 5-367, 6-367, 7-68, 7-367, 9-367,



11-367, 67-367, 154-367, 244-367, 290-358, 361-547, 361-550,



362-957, 363-384, 363-387, 363-394, 363-399, 363-400, 363-403,



363-411, 363-417, 363-418, 363-424, 363-434, 363-436, 363-443,



363-444, 363-446, 363-450, 363-454, 363-459, 363-460, 363-461,



363-464, 363-467, 363-469, 363-473, 363-476, 363-477, 363-480,



363-484, 363-487, 363-489, 363-490, 363-491, 363-492, 363-496,



363-497, 363-498, 363-500, 363-501, 363-504, 363-509, 363-510,



363-511, 363-512, 363-515, 363-517, 363-518, 363-521, 363-526,



363-527, 363-528, 363-529, 363-530, 363-532, 363-533, 363-534,



363-536, 363-538, 363-539, 363-540, 363-541, 363-544, 363-545,



363-547, 363-549, 363-550, 363-551, 363-553, 363-554, 363-555,



363-556, 363-559, 363-560, 363-562, 363-563, 363-564, 363-565,



363-566, 363-567, 363-569, 363-571, 363-572, 363-574, 363-580,



363-582, 363-584, 363-586, 363-590, 363-592, 363-594, 363-595,



363-600, 363-604, 363-608, 363-611, 363-630, 363-925, 363-950,



363-956, 363-957, 365-447, 365-502, 365-510, 365-544, 365-550,



365-554, 365-560, 365-948, 367-443, 367-955, 379-956, 381-956,



388-957, 390-936, 394-956, 401-957, 414-955, 416-950,



421-956, 425-956, 433-554, 442-957, 444-957, 448-957, 451-956,



451-957, 452-955, 456-956, 458-953, 458-955, 461-956, 472-952,



473-957, 477-956, 479-956, 479-957, 484-956, 485-615, 487-956,



491-936, 493-957, 494-956, 495-956, 496-957, 499-954, 499-956,



499-957, 505-956, 505-957, 509-957, 512-949, 516-955, 529-956,



531-957, 537-956, 539-956, 539-957, 541-957, 545-955, 548-947,



548-956, 553-956, 555-953, 563-778, 563-912, 563-957, 564-955,



568-957, 569-965, 572-965, 580-957, 584-1206, 584-1315, 585-957,



586-949, 590-956, 593-956, 593-957, 594-965, 595-734, 595-957,



595-965, 597-743, 601-957, 605-794, 606-710, 607-882, 610-956,



624-954, 629-955, 630-947, 636-957, 641-957, 644-954, 651-921,



655-810, 657-929, 659-777, 659-798, 659-799, 659-940, 664-904,



671-947, 671-957, 678-897, 678-919, 690-835, 705-957, 723-956,



723-957, 729-956, 731-956, 763-955, 775-955, 778-957, 784-957,



805-957, 813-957, 823-957, 836-957, 850-957, 854-957, 881-954,



911-956


56/6399886CB1/3796
1-268, 1-287, 184-830, 186-653, 280-606, 280-753, 280-767, 280-836,



280-852, 280-924, 280-936, 365-765, 365-768, 365-794, 439-1252,



497-916, 571-1252, 621-1286, 645-836, 709-1568, 732-1214,



743-991, 773-1403, 810-1354, 810-1367, 810-1440, 843-1013,



847-1135, 856-1475, 858-1504, 867-1103, 928-1240, 942-1561,



961-1568, 985-1521, 1010-1130, 1010-1451, 1155-1438, 1158-1413,



1207-1495, 1207-1737, 1272-1533, 1272-1729, 1451-1529, 1506-1963,



1531-1871, 1534-1829, 1540-1786, 1543-1737, 1543-1814,



1597-1835, 1649-2212, 1669-2235, 1703-1891, 1711-2149, 1757-2033,



1802-2377, 1883-2433, 1914-2400, 1970-2635, 2001-2230,



2001-2677, 2014-2440, 2036-2249, 2036-2521, 2081-2503, 2086-2350,



2124-2563, 2151-2480, 2177-2453, 2184-2418, 2251-2875,



2257-2524, 2305-2534, 2313-2532, 2313-2938, 2314-3011, 2320-2540,



2331-2799, 2332-2647, 2390-2549, 2415-2612, 2459-2744,



2592-2715, 2602-2830, 2611-2886, 2681-2880, 2681-2928, 2684-2975,



2700-2955, 2710-2998, 2716-3047, 2720-3177, 2725-2947,



2745-2901, 2778-3062, 2792-3019, 2792-3188, 2794-2993, 2808-3112,



2811-3065, 2825-3106, 2826-3084, 2826-3104, 2831-3039,



2868-3094, 2868-3100, 2876-3144, 2883-3112, 2883-3141, 2883-3310,



2904-3133, 2904-3460, 2914-3165, 2951-3227, 2959-3180,



2977-3227, 2991-3245, 3026-3426, 3027-3314, 3115-3376, 3124-3390,



3129-3730, 3181-3406, 3199-3444, 3204-3445, 3217-3786,



3234-3494, 3240-3488, 3281-3492, 3315-3796, 3327-3775, 3341-3580,



3437-3681, 3483-3711, 3484-3738, 3572-3781, 3629-3796


57/6024420CB1/2983
1-186, 1-286, 1-506, 1-517, 1-631, 1-637, 1-711, 1-787, 2-591, 2-638,



2-667, 3-660, 4-689, 14-650, 70-526, 110-705, 202-837, 272-862,



281-825, 284-883, 298-1007, 319-948, 332-906, 334-884, 346-1151,



403-540, 408-1110, 448-1180, 462-729, 529-1218, 569-1233,



578-1254, 607-852, 608-1341, 612-1232, 664-1503, 668-1227,



714-1339, 740-1382, 781-1404, 811-1688, 882-1650, 914-1605,



916-1611, 931-1462, 947-1202, 955-1479, 998-1585, 999-1798,



1019-1566, 1077-1903, 1078-1637, 1081-1664, 1093-1614, 1127-1943,



1130-1810, 1141-1783, 1147-1873, 1179-1881, 1217-1782,



1264-1829, 1267-1889, 1271-1878, 1294-1918, 1317-1930, 1415-1878,



1709-2983, 1740-2008


58/7481067CB1/3840
1-222, 160-947, 250-418, 514-606, 775-1155, 781-861, 1027-1420,



1086-1379, 1321-1662, 1463-2176, 1463-2964, 1664-1856, 2038-2353,



2248-2294, 2250-2759, 2264-2506, 2423-2507, 2458-2696,



2550-2824, 2571-2825, 2584-3076, 2708-2984, 2713-2753, 2824-3128,



2937-3384, 2970-3384, 3059-3570, 3221-3840


59/3378720CB1/1570
1-293, 1-363, 1-404, 1-437, 1-452, 1-511, 1-521, 1-544, 1-553,



1-557, 1-565, 1-569, 6-276, 41-656, 49-720, 68-639, 89-709, 121-747,



215-841, 219-429, 225-755, 249-864, 296-906, 339-955, 380-934,



423-599, 450-750, 461-739, 634-1045, 647-791, 771-1317, 825-995,



841-1425, 875-1481, 911-1528, 911-1536, 978-1535, 984-1515,



994-1402, 1070-1526, 1093-1316, 1130-1570, 1344-1570


60/938824CB1/409
1-245, 1-294, 1-338, 207-409, 333-393


61/1683721CB1/953
1-189, 1-209, 1-439, 1-470, 1-581, 10-695, 26-658, 209-685, 251-573,



258-536, 269-489, 298-734, 321-839, 324-540, 360-521, 386-862,



388-742, 522-948, 556-953, 646-953


62/1694122CB1/890
1-286, 2-25, 2-479, 25-44, 25-311, 25-314, 25-331, 25-337, 25-338,



25-416, 27-331, 29-110, 51-145, 78-554, 94-529, 96-554, 120-546,



160-439, 160-653, 160-705, 161-435, 164-424, 201-860, 301-554,



301-857, 326-628, 335-621, 336-437, 336-554, 362-487, 362-584,



362-862, 362-881, 362-890, 424-554, 428-554, 445-554, 448-554,



452-693, 452-886, 467-554, 482-725, 516-554, 553-774, 553-807,



553-822, 554-829, 555-869


63/1970615CB1/1960
1-421, 84-333, 117-709, 144-700, 178-360, 178-374, 282-525, 285-709,



286-550, 286-852, 311-555, 423-708, 423-709, 583-736, 598-880,



598-881, 598-1013, 598-1015, 598-1074, 598-1094, 598-1120,



634-1049, 653-1211, 670-921, 678-929, 709-968, 719-1226, 728-1117,



731-978, 740-948, 740-952, 740-1006, 742-965, 742-1294,



747-1016, 762-1213, 850-1163, 869-1053, 922-1173, 922-1186, 983-1255,



1038-1282, 1049-1308, 1049-1327, 1050-1546, 1058-1342,



1060-1546, 1095-1337, 1117-1546, 1117-1603, 1182-1395, 1198-1891,



1202-1447, 1202-1749, 1219-1470, 1226-1466, 1287-1516,



1338-1625, 1351-1590, 1355-1652, 1369-1960, 1447-1640, 1447-1887,



1447-1901, 1472-1774, 1474-1733


64/2314152CB1/832
1-519, 11-466, 13-433, 29-187, 29-476, 29-486, 29-588, 29-628, 32-408,



34-490, 35-239, 35-348, 35-415, 36-655, 36-662, 49-548, 57-457,



58-643, 61-648, 66-760, 77-603, 91-552, 91-598, 94-644,



172-415, 219-832, 412-686, 501-757, 509-832, 525-774


65/2886225CB1/546
1-454, 273-533, 275-546


66/6144418CB1/890
1-123, 1-596, 42-619, 42-630, 60-709, 130-660, 188-658, 188-732,



222-737, 222-778, 222-890, 255-732, 258-686


67/6834184CB1/807
1-554, 1-687, 1-807


68/6951005CB1/677
1-564, 1-643, 4-677


69/7250331CB1/617
1-287, 3-397, 241-532, 269-598, 289-600, 307-497, 344-498,



366-597, 370-617, 441-586, 441-615


70/1758413CB1/795
1-456, 292-768, 292-795, 417-695, 573-707


71/7011042CB1/1677
1-895, 241-1100, 241-1660, 251-753, 896-1043, 920-1043, 1259-1677


72/7427362CB1/1402
1-1268, 268-527, 270-1268, 787-926, 875-1402


73/7485304CB1/1251
1-1251, 73-171, 255-1081


74/1422394CB1/4961
1-493, 34-650, 38-279, 38-311, 39-650, 48-597, 51-590, 304-484,



304-753, 309-899, 421-957, 488-710, 559-633, 582-971, 632-777,



717-1013, 738-1442, 748-1151, 748-1383, 756-951, 958-1606,



1126-1486, 1126-1815, 1273-1817, 1277-1828, 1291-1949, 1322-1922,



1336-1840, 1336-1859, 1336-1908, 1438-1714, 1442-2028, 1509-1757,



1509-2045, 1549-2251, 1754-2257, 1760-2378, 1905-2232,



1905-2438, 1905-2485, 1905-2491, 1905-2571, 1905-2626, 1908-2337,



1925-2480, 1967-2630, 1969-2201, 2010-2503, 2012-2506,



2034-2531, 2037-2626, 2044-2617, 2067-2282, 2070-2350, 2108-2715,



2117-2622, 2132-2324, 2132-2636, 2157-2816, 2183-2689,



2209-2626, 2244-2715, 2278-2790, 2301-2758, 2363-2644, 2363-2647,



2475-2751, 2479-2794, 2479-2884, 2487-2836, 2497-2791,



2498-2827, 2507-2858, 2680-2901, 2680-3121, 2702-3304, 2723-3251,



2774-3063, 2825-3066, 2830-3456, 2837-3405, 2844-3134,



2859-3128, 2861-3141, 2863-3149, 2871-3479, 3077-3336, 3077-3588,



3141-3409, 3141-3601, 3152-3414, 3182-3457, 3260-3554,



3277-3672, 3299-3596, 3299-3602, 3329-3556, 3329-3566, 3349-3612,



3377-3678, 3390-3613, 3408-3634, 3420-3691, 3506-3778,



3519-3673, 3595-4170, 3602-3798, 3625-3922, 3690-3884, 3697-3928,



3716-3979, 3716-4001, 3862-4072, 3865-4142, 3890-4136,



3967-4157, 4002-4269, 4029-4210, 4068-4311, 4070-4381,



4072-4316, 4099-4312, 4152-4403, 4158-4431, 4230-4401, 4230-4750,



4230-4811, 4250-4403, 4277-4817, 4299-4809, 4355-4590,



4355-4603, 4381-4784, 4460-4729, 4460-4816, 4460-4961, 4499-4777,



4501-4645, 4533-4776, 4633-4829, 4639-4874


75/1336022CB1/3298
1-494, 217-493, 220-494, 220-613, 220-630, 220-893, 223-831, 226-878,



228-833, 230-934, 234-874, 241-494, 244-613, 269-561, 360-613,



408-885, 465-658, 467-740, 495-1070, 549-1177, 566-827,



577-797, 633-895, 646-1194, 656-953, 668-857, 679-973, 694-885,



705-1428, 721-1177, 758-1079, 781-1362, 782-1009, 797-1044,



812-1122, 825-1101, 831-1122, 836-1108, 842-1070, 866-1100, 917-1125,



919-1610, 939-1177, 944-1561, 947-1222, 966-1234, 967-1134,



970-1436, 979-1535, 996-1232, 1017-1596, 1051-1629, 1074-1621,



1076-1623, 1076-1634, 1085-1345, 1097-1576, 1145-1637,



1159-1366, 1185-1436, 1200-1478, 1228-1671, 1239-1510, 1248-1399,



1249-1888, 1266-1573, 1355-1670, 1374-2001, 1375-1922,



1375-1976, 1424-1961, 1442-2078, 1468-2027, 1497-1977, 1543-2124,



1550-2186, 1624-2160, 1631-2137, 1637-2265, 1661-2205,



1665-2248, 1669-1961, 1678-1954, 1691-1962, 1691-2303, 1699-2305,



1715-2272, 1728-2335, 1740-1962, 1777-2348, 1840-2314,



1856-2086, 1862-2108, 1884-2510, 1885-2515, 1886-2213, 1906-2450,



1910-2426, 1954-1987, 1961-2213, 2017-2460, 2017-2551,



2042-2197, 2048-2507, 2092-2714, 2101-2314, 2167-2678, 2303-2839,



2376-2839, 2479-2895, 2479-2898, 2479-3109, 2538-3144,



2550-3282, 2550-3298, 2588-2879, 2589-2879, 2600-2839, 2603-2839,



2730-3002, 2841-3212


76/7473674CB1/833
1-321, 1-328, 44-463, 44-483, 46-367, 53-519, 68-236, 68-490,



91-537, 114-477, 128-415, 144-290, 269-653, 279-648, 282-485, 282-536,



282-583, 282-609, 282-654, 287-490, 288-654, 289-644, 289-654,



293-653, 300-620, 301-489, 301-645, 301-710, 319-727, 322-496,



323-446, 323-456, 323-470, 323-482, 323-618, 323-628, 323-630,



323-635, 323-637, 323-639, 323-645, 323-646, 323-647, 323-652,



323-653, 323-663, 323-717, 323-730, 323-731, 323-786, 324-647,



326-479, 331-507, 340-647, 341-730, 353-497, 369-497, 395-547,



395-638, 424-647, 426-654, 482-823, 504-654, 567-826, 640-833,



687-823


77/7475846CB1/920
1-920, 199-552


78/7475860CB1/964
1-553, 25-281, 240-535, 243-610, 255-872, 255-873, 268-872, 283-507,



285-849, 289-882, 291-592, 292-578, 293-511, 298-581, 298-722,



300-548, 301-563, 302-594, 309-753, 309-758, 309-760, 314-697,



314-783, 315-580, 315-591, 315-933, 316-678, 316-756, 316-785,



317-518, 317-580, 322-575, 322-586, 324-606, 325-570, 328-579,



330-614, 336-617, 342-623, 346-649, 347-607, 371-898, 377-946,



433-703, 477-820, 484-733, 493-964, 495-804, 512-910, 526-811,



568-807, 639-880, 639-964, 684-832, 762-962, 818-964


79/7950941CB1/701
1-397, 9-701


80/7485334CB1/1742
1-724, 126-1078, 752-995, 752-1279, 896-1188, 1002-1231, 1002-1507,



1038-1619, 1167-1454, 1167-1615, 1167-1742, 1282-1632


81/7220001CB1/2295
1-606, 1-762, 40-275, 149-553, 379-1700, 574-897, 736-1024,



736-1036, 738-1357, 1052-1657, 1117-1681, 1155-1690, 1159-1690,



1193-1839, 1202-1831, 1203-1796, 1226-1771, 1254-1820, 1324-1947,



1324-1969, 1347-1928, 1363-2014, 1367-2014, 1400-1867,



1423-1933, 1425-2084, 1432-2004, 1441-1867, 1499-2176, 1519-1700,



1547-2059, 1599-2093, 1600-2208, 1602-1806, 1605-2295,



1606-2201, 1645-2223, 2257-2285, 2257-2288, 2257-2290, 2257-2291,



2257-2292, 2257-2294, 2257-2295, 2258-2295


82/5956275CB1/911
1-676, 60-818, 61-349, 348-911


83/346472CB1/1806
1-259, 1-478, 1-1803, 115-677, 243-606, 292-495, 296-895, 313-977,



325-935, 353-997, 355-851, 361-1061, 362-935, 387-1063, 392-1010,



395-1077, 397-1029, 397-1051, 442-1006, 449-948, 463-979,



479-1190, 534-1032, 534-1100, 534-1102, 539-1042, 541-1191,



577-1094, 602-790, 615-950, 617-1298, 657-1047, 669-1293,



670-1298, 705-1110, 714-1314, 739-1367, 745-1271, 748-1367, 767-1282,



767-1286, 780-1367, 821-1116, 823-1421, 830-1481, 843-1555,



846-1443, 849-1506, 852-1496, 865-1404, 878-1528, 886-1563,



889-1596, 920-1451, 920-1460, 920-1463, 925-1227, 926-1392,



934-1475, 939-1595, 939-1625, 940-1483, 943-1504, 958-1171, 958-1519,



962-1479, 966-1566, 969-1468, 973-1314, 978-1489, 985-1511,



988-1417, 988-1535, 995-1675, 1001-1391, 1013-1286, 1013-1581,



1020-1170, 1122-1765, 1122-1793, 1167-1793, 1172-1443,



1188-1637, 1208-1793, 1214-1806, 1284-1767, 1294-1703, 1408-1684


84/643526CB1/603
1-539, 1-603, 4-603, 44-276, 44-535


85/1483418CB1/1888
1-105, 1-168, 1-193, 1-280, 1-301, 1-394, 1-564, 1-631, 51-522,



89-618, 125-632, 125-644, 125-657, 129-776, 209-717, 211-751, 258-874,



290-680, 297-823, 343-964, 359-1005, 387-561, 396-968, 415-823,



450-507, 460-1056, 470-711, 470-996, 518-1009, 568-1020,



568-1024, 597-764, 606-910, 606-1200, 611-1300, 640-1093, 640-1194,



656-1181, 669-1268, 678-1005, 684-1331, 689-954, 696-1219,



696-1298, 711-1284, 713-1138, 768-1047, 768-1058, 768-1304,



781-1314, 842-1366, 895-1434, 918-1477, 940-1467, 1035-1561,



1112-1664, 1131-1383, 1267-1725, 1307-1554, 1330-1529, 1334-1808,



1334-1888, 1340-1528, 1340-1842, 1366-1642, 1407-1679,



1411-1872, 1430-1888


86/2683477CB1/1576
1-254, 1-513, 1-531, 1-537, 1-557, 1-592, 1-617, 1-641, 1-643,



1-653, 1-692, 1-713, 1-818, 2-742, 16-717, 27-967, 361-1008, 408-1254,



425-1121, 467-1168, 478-1175, 560-1141, 572-1380, 574-1399,



595-1085, 629-1195, 655-1307, 699-1314, 754-1196, 773-1576,



787-1284, 809-1573, 879-1576, 971-1573, 985-1576, 995-1572,



1060-1576, 1075-1576


87/5580991CB1/415
1-256, 1-415


88/5605931CB1/762
1-705, 66-705, 72-748, 157-705, 166-705, 211-705, 271-762,



275-759, 358-708, 366-701, 454-705, 480-705


89/6975241CB1/654
1-654, 96-653


90/6988529CB1/505
1-464, 1-505


91/6996808CB1/841
1-514, 85-548, 85-745, 85-841


92/7472689CB1/1367
1-253, 192-448, 192-769, 192-781, 192-847, 284-331, 387-647, 444-546,



444-553, 444-587, 444-671, 444-674, 444-678, 444-699, 444-731,



444-734, 444-794, 444-810, 445-508, 481-1050, 483-963, 489-977,



490-765, 522-731, 529-863, 530-730, 543-937, 568-688, 572-1125,



604-859, 614-830, 627-1222, 630-1120, 663-1011, 667-1152,



672-1222, 686-1339, 715-1200, 721-927, 721-1259, 729-1290,



735-994, 759-1286, 770-1349, 796-1359, 818-1344, 837-1367, 861-1111,



861-1275, 861-1286, 861-1355, 873-1367, 890-1024, 897-1246,



897-1286, 909-1286, 911-1126, 912-1367, 914-1186, 922-1367,



941-1286, 944-1286, 951-1361, 955-1208, 960-1340, 1001-1304,



1001-1332, 1021-1286, 1022-1286, 1028-1286, 1035-1286, 1039-1367,



1041-1351, 1042-1295, 1075-1259, 1084-1286, 1094-1367,



1096-1329, 1101-1286, 1117-1286, 1118-1286, 1144-1286, 1152-1361,



1168-1286, 1180-1286, 1180-1287, 1202-1286, 1239-1286


93/876751CB1/4595
1-594, 248-705, 249-910, 277-740, 289-741, 403-907, 417-988,



440-992, 453-1111, 481-1192, 498-992, 503-1111, 568-812, 568-1066,



599-1188, 642-1192, 665-1303, 675-1202, 997-1564, 1045-1569,



1093-1340, 1093-1658, 1130-1681, 1197-1579, 1330-1617, 1375-1653,



1502-2181, 1507-2168, 1538-2103, 1674-1918, 1675-2130,



1678-2164, 1850-2311, 1890-2119, 1964-2399, 2120-2369, 2120-2746,



2130-2171, 2330-2538, 2330-2804, 2335-2624, 2348-2623,



2356-2650, 2423-2666, 2423-2844, 2447-2637, 2483-3218, 2498-2683,



2501-2670, 2521-2834, 2551-2816, 2563-2757, 2587-3211,



2592-2939, 2597-2878, 2637-3216, 2668-2996, 2694-2939, 2696-2946,



2696-3294, 2725-2994, 2811-3050, 2817-3230, 2839-3111,



2840-3203, 2843-3166, 2892-3145, 2892-3207, 2934-3222, 2945-3099,



2946-3181, 2950-3259, 2966-3244, 2985-3138, 2985-3180,



2995-3223, 2995-3470, 3002-3226, 3021-3324, 3053-3284, 3071-3360,



3080-3367, 3208-3464, 3208-3471, 3208-3720, 3215-3470,



3220-3498, 3233-3522, 3233-3528, 3257-3570, 3294-3845, 3341-3593,



3341-3625, 3350-3606, 3350-3625, 3379-3570, 3390-3653,



3391-3632, 3431-3570, 3432-3734, 3482-3726, 3482-3746, 3492-3730,



3515-3863, 3572-3827, 3572-3829, 3574-3867, 3574-4072,



3593-3852, 3646-3827, 3661-3922, 3661-3931, 3684-3921,



3694-3916, 3711-3981, 3714-3991, 3717-3992, 3743-4032, 3770-4030,



3788-4036, 3789-3948, 3799-4049, 3817-4036, 3820-4053,



3833-4085, 3859-4069, 3863-4027, 3863-4136, 3863-4376, 3866-4106,



3867-4180, 3869-3930, 3870-4126, 3877-4152, 3909-4123,



3940-4566, 3960-4571, 4045-4320, 4088-4379, 4095-4366, 4143-4560,



4160-4390, 4188-4579, 4216-4595, 4246-4465, 4263-4493,



4269-4525, 4366-4595, 4390-4569, 4425-4568, 4428-4574


94/2512510CB1/4759
1-218, 124-348, 124-403, 124-474, 124-511, 124-523, 124-529,



124-547, 124-576, 124-595, 124-612, 124-615, 124-621, 124-628, 124-631,



124-645, 124-689, 124-698, 124-713, 124-718, 124-724, 124-731,



124-734, 124-777, 124-778, 124-813, 124-852, 125-709, 126-398,



132-583, 142-594, 142-609, 155-583, 181-663, 197-433, 242-482,



256-561, 338-865, 365-597, 390-656, 436-671, 436-839, 473-882,



475-869, 514-959, 563-806, 689-1241, 813-1483, 833-1469,



945-1339, 946-1492, 973-1329, 1036-1523, 1114-1501, 1139-1426,



1176-1430, 1185-1684, 1212-1883, 1215-1465, 1216-1818, 1250-1811,



1273-1508, 1293-1876, 1313-1929, 1416-1880, 1462-2059,



1494-1803, 1494-1823, 1496-2069, 1512-1691, 1529-2126, 1530-1811,



1565-2214, 1568-2155, 1747-2231, 1808-2326, 1838-2387,



1866-2157, 1883-2594, 1893-2513, 1893-2547, 1901-2071, 2043-2312,



2071-2487, 2100-2372, 2192-2493, 2248-2427, 2272-2528,



2286-2552, 2292-2592, 2432-2722, 2483-2717, 2500-2743, 2538-2831,



2617-2750, 2626-2899, 2653-3239, 2654-2858, 2656-2908,



2730-3003, 2732-2978, 2781-3038, 2781-3316, 2866-3098, 2876-3553,



2887-3045, 2985-3264, 3024-3273, 3039-3276, 3051-3285,



3055-3331, 3075-3281, 3080-3362, 3099-3362, 3141-3440, 3146-3360,



3152-3420, 3153-3427, 3153-3441, 3181-3442, 3241-3496,



3344-3577, 3400-3661, 3400-3665, 3409-3663, 3424-3689, 3431-3762,



3449-3713, 3452-3738, 3460-3729, 3473-3731, 3481-3745,



3502-3775, 3504-3718, 3527-3775, 3528-3752, 3546-3786, 3548-3777,



3570-3853, 3582-3801, 3582-3806, 3617-3813, 3617-3877,



3637-3991, 3669-3877, 3683-3876, 3690-3898, 3707-3947, 3707-3964,



3709-3921, 3738-3994, 3738-4303, 3750-3970, 3783-4044,



3816-4101, 3844-4107, 3943-4256, 3960-4179, 3973-4187, 3973-4190,



3973-4191, 3973-4209, 3973-4374, 3984-4240, 3991-4236,



4000-4271, 4007-4216, 4007-4443, 4057-4295, 4072-4334, 4072-4336,



4072-4605, 4072-4733, 4094-4732, 4100-4759, 4102-4369,



4144-4707, 4154-4466, 4155-4393, 4155-4735, 4155-4745, 4165-4747,



4170-4747, 4171-4759, 4179-4444, 4184-4419, 4189-4759,



4205-4759, 4211-4759, 4213-4759, 4239-4748, 4241-4759, 4250-4747,



4258-4740, 4259-4747, 4266-4759, 4272-4754, 4292-4755,



4299-4747, 4300-4749, 4300-4756, 4302-4747, 4304-4747, 4305-4751,



4308-4747, 4313-4751, 4319-4759, 4320-4759, 4321-4740,



4323-4759, 4327-4747, 4329-4749, 4330-4741, 4330-4747, 4334-4753,



4335-4749, 4338-4753, 4338-4759, 4339-4753, 4339-4759,



4340-4740, 4340-4747, 4341-4747, 4343-4747, 4344-4747, 4345-4756,



4350-4747, 4354-4747, 4356-4750, 4360-4759, 4362-4582,



4362-4682, 4362-4710, 4362-4734, 4362-4743, 4375-4751, 4376-4747,



4381-4753, 4388-4748, 4391-4747, 4391-4750, 4396-4738,



4397-4747, 4400-4747, 4401-4751, 4403-4747, 4423-4747, 4427-4748,



4428-4747, 4429-4741, 4435-4649, 4435-4737, 4435-4738,



4435-4745, 4440-4747, 4444-4754, 4451-4728, 4456-4747, 4456-4753,



4462-4748, 4498-4759, 4503-4748, 4528-4748, 4536-4747,



4538-4752, 4549-4759, 4551-4750, 4551-4759, 4568-4747, 4570-4748,



4576-4759, 4584-4759, 4587-4731, 4604-4747, 4610-4741,



4612-4747, 4622-4748, 4625-4747, 4625-4748, 4640-4747, 4642-4747,



4644-4748, 4650-4747, 4662-4741, 4662-4759, 4678-4747,



4685-4759


95/7486326CB1/3203
1-870, 411-3203


96/1221545CB1/1681
1-580, 69-613, 360-596, 377-763, 545-982, 569-775, 569-803,



583-1202, 657-901, 756-982, 784-1371, 986-1219, 1102-1615, 1204-1681,



1291-1511, 1339-1555, 1400-1614, 1446-1681


97/124737CB1/1207
1-529, 49-529, 309-1007, 319-729, 394-790, 402-566, 402-735,



406-568, 490-753, 502-988, 649-1179, 664-927, 671-1207, 697-963,



710-1092, 740-1175, 744-1092, 749-1092, 755-1015, 755-1207,



762-1207, 769-1179, 817-1008, 838-1092, 880-1092


98/1510784CB1/1544
1-196, 1-805, 316-894, 323-929, 366-783, 374-900, 374-916,



419-681, 446-962, 446-1005, 446-1038, 497-974, 534-886, 553-1008,



594-972, 658-1080, 669-1072, 717-1215, 1094-1523, 1094-1544,



1216-1447


99/1901257CB1/1519
1-248, 1-1519, 4-384, 79-583, 108-761, 481-645, 491-1007,



501-763, 545-1240, 708-1008, 714-1511, 784-1261, 787-1480, 802-1519,



808-1519, 817-1519, 839-1261, 839-1519, 841-1519, 853-1266,



878-1519, 901-1517, 924-1519, 931-1261, 945-1519, 1005-1519,



1007-1266, 1031-1261, 1241-1519


100/2044370CB1/525
1-235, 1-470, 1-525


101/2820933CB1/1062
1-862, 3-870, 431-1006, 440-1034, 440-1062, 443-995, 454-1009,



542-888, 663-966


102/2902793CB1/2155
1-2155, 66-129, 66-159, 66-304, 66-596, 66-653, 66-674, 92-736,



318-859, 318-860, 340-700, 372-973, 379-882, 379-923, 396-973,



405-973, 477-972, 487-937, 487-996, 487-998, 487-1053, 487-1054,



498-1083, 519-1002, 610-1001, 620-1016, 679-1189, 712-1259,



720-1096, 723-1252, 736-1436, 747-1252, 750-1252, 768-1309,



791-1178, 795-1052, 834-1122, 859-1420, 871-1380, 871-1385, 872-1355,



875-1252, 965-1287, 1002-1535, 1018-1597, 1047-1585, 1063-1463,



1157-1779, 1158-1586, 1184-1622, 1219-1704, 1240-1530,



1253-1717, 1253-1726, 1330-1933, 1333-1925, 1342-1977, 1386-1886,



1389-1631, 1394-1944, 1404-1977, 1446-2090, 1458-2078,



1487-2105, 1524-2071, 1621-2062, 1844-2155, 1978-2083


103/7486536CB1/1777
1-239, 1-259, 1-562, 10-255, 13-284, 19-248, 19-325, 19-451,



26-322, 49-265, 49-453, 140-367, 141-383, 148-368, 172-518, 174-553,



176-278, 181-281, 182-430, 182-454, 185-649, 189-449, 196-432,



196-825, 209-328, 217-486, 532-782, 634-914, 634-920, 634-1115,



634-1223, 644-926, 730-1013, 742-1003, 742-1013, 742-1055, 742-1300,



836-1135, 908-1128, 908-1377, 950-1192, 984-1199, 1055-1297,



1110-1390, 1117-1738, 1149-1753, 1171-1754, 1177-1663, 1181-1732,



1222-1733, 1302-1533, 1302-1754, 1302-1777, 1310-1758,



1324-1764, 1327-1622, 1333-1568, 1348-1755, 1454-1756, 1469-1696,



1511-1769, 1587-1765, 1590-1777, 1603-1765


104/8137305CB1/2587
1-308, 1-590, 1-899, 2-529, 5-481, 5-706, 27-611, 29-786,



30-611, 40-710, 41-276, 63-603, 87-873, 88-735, 89-688, 175-965, 309-609,



410-925, 481-1028, 550-806, 642-912, 676-1032, 693-936, 693-1024,



813-1102, 875-1451, 1042-1605, 1069-1228, 1170-1692, 1185-1403,



1290-1512, 1317-1947, 1346-1907, 1346-1998, 1381-1968,



1384-2097, 1427-2080, 1431-1686, 1463-1947, 1471-1955, 1548-1845,



1548-1991, 1559-2178, 1560-2068, 1566-1851, 1585-2176,



1592-1909, 1592-2009, 1604-1834, 1611-2217, 1674-2067, 1683-2151,



1689-2119, 1725-2276, 1734-1978, 1734-2119, 1734-2257,



1734-2301, 1734-2307, 1734-2587, 1740-2195, 1761-2201, 1775-2134,



1800-2211, 1802-2200


105/3793128CB1/1490
1-278, 1-452, 1-531, 1-534, 1-547, 1-583, 1-625, 1-707,



7-752, 91-728, 103-724, 110-524, 110-554, 110-575, 110-651, 110-677, 110-695,



110-739, 134-796, 193-576, 319-942, 435-1187, 436-970,



437-1067, 454-1190, 488-1163, 541-1134, 595-1270, 642-1173, 647-1162,



661-1280, 668-1271, 685-1271, 703-1376, 743-1435, 753-1286,



753-1454, 840-1490, 887-1349, 915-1374, 959-1484, 962-1484,



968-1437, 1005-1484, 1057-1484, 1073-1484, 1149-1484, 1177-1484,



1332-1484


106/4001243CB1/1174
1-292, 1-406, 1-442, 1-503, 1-570, 1-648, 52-303, 52-723,



86-644, 101-373, 101-583, 101-672, 113-321, 116-663, 285-854, 291-744,



291-898, 305-810, 415-1174, 427-710, 430-945, 436-723, 488-963,



504-720, 695-963


107/6986717CB1/818
1-556, 1-611, 13-546, 83-818


108/7503512CB1/4717
1-4679, 1-4690, 124-403, 124-474, 124-511, 124-523, 124-529,



124-547, 124-576, 124-595, 124-599, 126-398, 132-583, 132-599, 142-594,



142-599, 155-583, 171-360, 197-433, 242-482, 252-357, 252-360,



257-561, 366-478, 396-946, 397-946, 620-1172, 628-1180, 744-1413,



764-1400, 848-1500, 876-1270, 877-1423, 904-1260,



967-1454, 1032-1696, 1038-1658, 1045-1432, 1079-1357, 1107-1361,



1116-1615, 1146-1396, 1147-1749, 1148-1397, 1152-1814, 1181-1742,



1197-1745, 1204-1439, 1224-1806, 1347-1811, 1399-1968,



1418-1492, 1425-1950, 1425-1977, 1427-2000, 1443-1622, 1461-1742,



1496-2145, 1678-2162, 1769-2318, 1809-2230, 2031-2303,



2773-3462, 2797-3170, 2799-3170, 2807-3484, 2820-3351, 2824-3468,



2935-3513, 2955-3204, 2968-3205, 2982-3216, 2986-3262,



2999-3194, 3008-3212, 3011-3293, 3019-3399, 3020-3261, 3022-3553,



3045-3499, 3057-3500, 3059-3500, 3072-3371, 3077-3291,



3084-3494, 3086-3372, 3093-3572, 3096-3532, 3110-3434, 3112-3373,



3120-3468, 3139-3605, 3152-3217, 3166-3290, 3216-3525,



3252-3499, 3276-3508, 3276-3880, 3279-3861, 3326-3572, 3331-3592,



3331-3596, 3340-3536, 3340-3594, 3340-3601, 3355-3620,



3357-3609, 3362-3693, 3379-4037, 3382-4151, 3383-3644, 3383-3669,



3384-3658, 3391-3660, 3396-3913, 3403-3651, 3404-3662,



3404-3680, 3412-3676, 3433-3706, 3434-3728, 3434-3886, 3435-3649,



3443-3683, 3444-3742, 3456-3855, 3458-3694, 3458-3706,



3458-4047, 3459-3683, 3465-3750, 3477-3717, 3479-3708, 3491-4005,



3499-4117, 3501-3784, 3501-4125, 3513-3732, 3513-3737,



3515-3843, 3516-3806, 3528-3606, 3539-3830, 3542-3627, 3542-3807,



3542-3824, 3542-3882, 3542-4149, 3548-3744, 3548-3782,



3548-3808, 3568-3922, 3568-3978, 3571-4320, 3582-4060, 3586-3868,



3586-4184, 3588-4068, 3590-4295, 3600-3808, 3600-4261,



3603-3884, 3603-4075, 3603-4133, 3614-3807, 3621-3829, 3637-4192,



3638-3878, 3638-3895, 3640-3852, 3650-3933, 3654-4176,



3656-4212, 3659-3909, 3660-3950, 3667-3940, 3669-3925, 3669-4234,



3671-4053, 3671-4065, 3681-3901, 3707-4013, 3713-4166,



3714-3975, 3715-3971, 3717-3974, 3720-3873, 3721-3965, 3737-4296,



3738-4125, 3747-4032, 3756-4373, 3770-4118, 3772-4050,



3773-4212, 3775-4038, 3802-4095, 3808-4032, 3809-4390, 3813-4315,



3815-4059, 3815-4496, 3825-4084, 3825-4119, 3825-4120,



3828-4318, 3828-4462, 3829-4016, 3829-4043, 3831-4101, 3832-4343,



3838-4100, 3841-4091, 3847-4480, 3858-4082, 3860-4236,



3860-4303, 3866-4134, 3871-4132, 3874-4634, 3876-4187, 3877-4132,



3878-4164, 3892-4110, 3896-4608, 3901-4188, 3904-4118, 3904-4121,



3904-4122, 3904-4140, 3904-4305, 3904-4320, 3907-4157, 3908-4200,



3915-4171, 3920-4122, 3920-4345, 3922-4167,



3930-4225, 3931-4147, 3931-4186, 3931-4195, 3931-4201, 3931-4202,



3933-4165, 3934-4025, 3938-4147, 3938-4374, 3940-4201,



3942-4245, 3944-4238, 3947-4218, 3953-4561, 3956-4331, 3963-4628,



3967-4223, 3981-4265, 3982-4247, 3988-4226, 3988-4635,



3989-4545, 3990-4247, 3998-4211, 3998-4298, 3998-4673, 4000-4304,



4000-4606, 4003-4265, 4003-4267, 4003-4285, 4003-4536,



4003-4664, 4007-4230, 4013-4306, 4019-4552, 4025-4587, 4025-4631,



4025-4663, 4027-4289, 4031-4288, 4031-4310, 4031-4676,



4031-4705, 4033-4248, 4033-4253, 4033-4300, 4035-4561, 4043-4462,



4046-4341, 4049-4325, 4050-4280, 4052-4532, 4053-4324,



4059-4312, 4059-4356, 4071-4355, 4075-4638, 4084-4357, 4085-4642,



4086-4324, 4086-4676, 4087-4397, 4091-4329, 4094-4386,



4094-4663, 4094-4703, 4095-4346, 4096-4678, 4101-4678, 4103-4624,



4105-4574, 4109-4647, 4110-4375, 4111-4389, 4115-4350,



4115-4387, 4115-4398, 4115-4417, 4117-4359, 4120-4339, 4120-4674,



4128-4417, 4129-4425, 4133-4423, 4135-4367, 4138-4695,



4142-4697, 4144-4693, 4152-4486, 4170-4679, 4172-4711, 4174-4376,



4181-4678, 4189-4671, 4190-4459, 4190-4665, 4190-4678,



4190-4687, 4191-4363, 4192-4458, 4193-4439, 4197-4630, 4197-4702,



4200-4636, 4203-4685, 4206-4672, 4207-4678, 4212-4490,



4223-4686, 4230-4678, 4231-4676, 4231-4680, 4233-4678, 4235-4678,



4236-4682, 4239-4678, 4243-4619, 4244-4682, 4246-4586,



4250-4678, 4250-4692, 4251-4696, 4252-4671, 4254-4717, 4258-4678,



4260-4680, 4261-4672, 4261-4678, 4265-4684, 4266-4680,



4269-4684, 4269-4703, 4270-4684, 4270-4707, 4271-4671, 4271-4678,



4272-4678, 4274-4678, 4275-4678, 4276-4687, 4281-4678,



4285-4678, 4287-4681, 4289-4529, 4293-4513, 4293-4613, 4293-4641,



4293-4665, 4293-4674, 4295-4541, 4298-4581, 4306-4682,



4307-4678, 4312-4684, 4315-4558, 4319-4679, 4322-4678, 4322-4681,



4323-4597, 4327-4669, 4328-4678, 4331-4678, 4332-4682,



4334-4678, 4344-4597, 4351-4672, 4354-4678, 4355-4581, 4356-4625,



4358-4679, 4359-4678, 4360-4672, 4366-4580, 4366-4668,



4366-4669, 4366-4676, 4369-4655, 4375-4685, 4377-4478, 4377-4663,



4382-4659, 4387-4677, 4389-4668, 4393-4679, 4407-4644,



4429-4705, 4434-4679, 4459-4679, 4467-4678, 4469-4683, 4480-4708,



4482-4643, 4482-4681, 4482-4709, 4486-4575, 4499-4678,



4501-4679, 4515-4702, 4518-4662, 4535-4678, 4541-4672, 4556-4678,



4571-4678, 4573-4678, 4595-4672, 4595-4709, 4609-4678


















TABLE 5








Polynucleotide
Incyte
Representative


SEQ ID NO:
Project ID:
Library

















55
095765CB1
PITUNOT06


56
6399886CB1
PANCTUT01


57
6024420CB1
TESTNOT11


58
7481067CB1
BRSTNOT07


59
3378720CB1
KERANOT02


60
938824CB1
CERVNOT01


61
1683721CB1
PROSNOT15


62
1694122CB1
COLNNOT23


63
1970615CB1
PROSTUT09


64
2314152CB1
CONUTUT01


65
2886225CB1
UTRSTMR02


66
6144418CB1
BRANDIN01


67
6834184CB1
BRSTNON02


68
6951005CB1
BRAITDR02


69
7250331CB1
KIDNTUT15


71
7011042CB1
BRAZNOT01


72
7427362CB1
BRSTTMR01


73
7485304CB1
SEMVTDE01


74
1422394CB1
MIXDUNB01


75
1336022CB1
COLNNOT13


76
7473674CB1
LUNGNON07


78
7475860CB1
ADRENON04


79
7950941CB1
BRABNOE02


80
7485334CB1
BSTMNON02


81
7220001CB1
COLXTDT01


82
5956275CB1
BRAUNOR01


83
346472CB1
THYMNOT02


84
643526CB1
BRAIFEE05


85
1483418CB1
SINTBST01


86
2683477CB1
SINIUCT01


87
5580991CB1
UTRENON03


88
5605931CB1
MONOTXN03


89
6975241CB1
BRAHTDR04


90
6988529CB1
BRAIFER05


91
6996808CB1
BRAXTDR17


92
7472689CB1
LNODNOT12


93
876751CB1
THYRNOT03


94
2512510CB1
BRAUNOR01


96
1221545CB1
NEUTFMT01


97
124737CB1
THYMNON04


98
1510784CB1
SINTFER03


99
1901257CB1
BRSTTMT02


100
2044370CB1
HIPONON02


101
2820933CB1
ADRETUT06


102
2902793CB1
DRGCNOT01


103
7486536CB1
BRSTNOT05


104
8137305CB1
MIXDTUE01


105
3793128CB1
BRSTNOT28


106
4001243CB1
PROSTMT03


107
6986717CB1
BRAIFER05


108
7503512CB1
BRSTNOT01


















TABLE 6








Library
Vector
Library Description







ADRENON04
PSPORT1
Normalized library was constructed from 1.36 × 1e6 independent




clones from an adrenal tissue library. Starting RNA was made




from adrenal gland tissue removed from a 20-year-old Caucasian




male, who died from head trauma. The library was normalized




in two rounds using conditions adapted from Soares et al. (PNAS




(1994) 91: 9228-9232) and Bonaldo et al. (Genome Res (1996)




6: 791-806), using a significantly longer (48-hours/round)




reannealing hybridization period.


ADRETUT06
pINCY
Library was constructed using RNA isolated from adrenal tumor




tissue removed from a 57-year-old Caucasian female during a




unilateral right adrenalectomy. Pathology indicated




pheochromocytoma, forming a nodular mass completely replacing the




medulla of the adrenal gland.


BRABNOE02
PBK-CMV
This 5′ biased random primed library was constructed using




RNA isolated from vermis tissue removed from a 35-year-old




Caucasian male who died from cardiac failure. Pathology




indicated moderate leptomeningeal fibrosis and multiple




microinfarctions of the cerebral neocortex. Patient history




included dilated cardiomyopathy, congestive heart failure,




cardiomegaly, and an enlarged spleen and liver. Patient




medications included simethicone, Lasix, Digoxin, Colace, Zantac,




captopril, and Vasotec.


BRAHTDR04
PCDNA2.1
This random primed library was constructed using RNA isolated




archaecortex, anterior hippocampus tissue removed from a 55-




year-old Caucasian female who died from cholangiocarcinoma.




Pathology indicated mild meningeal fibrosis predominately over




the convexities, scattered axonal spheroids in the white matter




of the cingulate cortex and the thalamus., and a few scattered




neurofibrillary tangles in the entorhinal cortex and the




periaqueductal gray region. Pathology for the associated tumor




tissue indicated well-differentiated cholangiocarcinoma of the




liver with residual or relapsed tumor. Patient history included




cholangiocarcinoma, post-operative Budd-Chiari syndrome, biliary




ascites, hydrothorax, dehydration, malnutrition, oliguria and




acute renal failure. Previous surgeries included cholecystectomy




and resection of 85% of the liver.


BRAIFEE05
PCDNA2.1
This 5' biased random primed library was constructed using RNA




isolated from brain tissue removed from a Caucasian male




fetus who was stillborn with a hypoplastic left heart at




23 weeks' gestation.


BRAIFER05
pINCY
Library was constructed using RNA isolated from brain tissue




removed from a Caucasian male fetus who was stillborn with a




hypoplastic left heart at 23 weeks' gestation.


BRAITDR02
PCDNA2.1
This random primed library was constructed using RNA isolated




from allocortex, neocortex, anterior and frontal cingulate tissue




removed from a 55-year-old Caucasian female who died from




cholangiocarcinoma. Pathology indicated mild meningeal fibrosis




predominately over the convexities, scattered axonal spheroids




in the white matter of the cingulate cortex and the thalamus, and




a few scattered neurofibrillary tangles in the entorhinal cortex




and the periaqueductal gray region. Pathology for the associated




tumor tissue indicated well-differentiated cholangiocarcinoma of




the liver with residual or relapsed tumor. Patient history




included cholangiocarcinoma, post-operative Budd-Chiari syndrome,




biliary ascites, hydrothorax, dehydration, malnutrition,




oliguria and acute renal failure. Previous surgeries included




cholecystectomy and resection of 85% of the liver.


BRANDIN01
pINCY
This normalized pineal gland tissue library was constructed from




.4 million independent clones from a pineal gland tissue library




from two different donors. Starting RNA was made from pooled




pineal gland tissue removed from two Caucasian females: a 68-




year-old (donor A) who died from congestive heart failure and a




79-year-old (donor B) who died from pneumonia.




Neuropathology for donor A indicated mild to moderate Alzheimer




disease, atherosclerosis, and multiple infarctions.




Neuropathology for donor B indicated severe Alzheimer disease,




arteriolosclerosis, cerebral amyloid angiopathy and multiple




infarctions. There were diffuse and neuritic amyloid plaques




and neurofibrillary tangles throughout the brain sections examined




in both donors. Patient history included diabetes mellitus,




rheumatoid arthritis, hyperthyroidism, amyloid heart disease, and




dementia in donor A; and pseudophakia, gastritis with bleeding,




glaucoma, peripheral vascular disease, COPD, delayed onset




tonic/clonic seizures, and transient ischemic attack in donor B.




The library was normalized in one round using conditions




adapted from Soares et al., PNAS (1994) 91: 9228-9232 and




Bonaldo et al., Genome Research 6 (1996): 791, except that a




significantly longer (48 hours/round) reannealing hybridization




was used.


BRAUNOR01
pINCY
This random primed library was constructed using RNA isolated




from striatum, globus pallidus and posterior putamen tissue




removed from an 81-year-old Caucasian female who died from a




hemorrhage and ruptured thoracic aorta due to atherosclerosis.




Pathology indicated moderate atherosclerosis involving the




internal carotids, bilaterally; microscopic infarcts of the frontal




cortex and hippocampus; and scattered diffuse amyloid plaques




and neurofibrillary tangles, consistent with age. Grossly, the




leptomeninges showed only mild thickening and hyalinization along




the superior sagittal sinus. The remainder of the leptomeninges




was thin and contained some congested blood vessels. Mild atrophy




was found mostly in the frontal poles and lobes, and temporal




lobes, bilaterally. Microscopically, there were pairs of




Alzheimer type II astrocytes within the deep layers of




the neocortex. There was increased satellitosis around neurons




in the deep gray matter in the middle frontal cortex. The




amygdala contained rare diffuse plaques and neurofibrillary




tangles. The posterior hippocampus contained a microscopic area of




cystic cavitation with hemosiderin-laden macrophages surrounded




by reactive gliosis. Patient history included sepsis,




cholangitis, post-operative atelectasis, pneumonia CAD,




cardiomegaly due to left ventricular hypertrophy, splenomegaly,




arteriolonephrosclerosis, nodular colloidal goiter, emphysema,




CHF, hypothyroidism, and peripheral vascular disease.


BRAXTDR17
PCDNA2.1
This random primed library was constructed using RNA isolated




from temporal neocortex tissue removed from a 55-year-old




Caucasian female who died from cholangiocarcinoma. Pathology




indicated mild meningeal fibrosis predominately over the




convexities, scattered axonal spheroids in the white matter of




the cingulate cortex and the thalamus, and a few scattered




neurofibrillary tangles in the entorhinal cortex and the




periaqueductal gray region. Pathology for the associated tumor




tissue indicated well-differentiated cholangiocarcinoma of the




liver with residual or relapsed tumor. Patient history included




cholangiocarcinoma, post-operative Budd-Chiari syndrome, biliary




ascites, hydrothorax, dehydration, malnutrition, oliguria and




acute renal failure. Previous surgeries included cholecystectomy




and resection of 85% of the liver.


BRAZNOT01
pINCY
Library was constructed using RNA isolated from striatum, globus




pallidus and posterior putamen tissue removed from a 45-




year-old Caucasian female who died from a dissecting aortic




aneurysm and ischemic bowel disease. Pathology indicated mild




arteriosclerosis involving the cerebral cortical white matter




and basal ganglia. Grossly, there was mild meningeal fibrosis and




mild focal atherosclerotic plaque in the middle cerebral artery,




as well as vertebral arteries bilaterally. Microscopically, the




cerebral hemispheres, brain stem and cerebellum reveal focal




areas in the white matter that contain blood vessels that were




barrel-shaped, hyalinized, with hemosiderin-laden macrophages in




the Virchow-Robin space. In addition, there were scattered




neurofibrillary tangles within the basolateral nuclei of the




amygdala. Patient history included mild atheromatosis of aorta and




coronary arteries, bowel and liver infarct due to aneurysm,




physiologic fatty liver associated with obesity, mild diffuse




emphysema, thrombosis of mesenteric and portal veins, cardiomegaly




due to hypertrophy of left ventricle, arterial hypertension,




acute pulmonary edema, splenomegaly, obesity (300 lb.), leiomyoma




of uterus, sleep apnea, and iron deficiency anemia.


BRSTNON02
pINCY
This normalized breast tissue library was constructed from 6.2 million




independent clones from a pool of two libraries from two




different donors. Starting RNA was made from breast tissue removed




from a 46-year-old Caucasian female during a bilateral




reduction mammoplasty (donor A), and from breast tissue removed




from a 60-year-old Caucasian female during a bilateral




reduction mammoplasty (donor B). Pathology indicated normal breast




parenchyma, bilaterally (A) and bilateral mammary




hypertrophy (B). Patient history included hypertrophy of breast,




obesity, lumbago, and glaucoma (A) and joint pain in the




shoulder, thyroid cyst, colon cancer, normal delivery and cervical




cancer (B). Family history included cataract, osteoarthritis,




uterine cancer, benign hypertension, hyperlipidemia, and alcoholic




cirrhosis of the liver, cerebrovascular disease, and type II




diabetes (A) and cerebrovascular accident, atherosclerotic




coronary artery disease, colon cancer, type II diabetes,




hyperlipidemia, depressive disorder, and Alzheimer's Disease.




The library was normalized in two rounds using conditions




adapted from Soares et al., PNAS (1994) 91: 9228-9232 and




Bonaldo et al., Genome Research 6 (1996): 791, except that a




significantly longer (48 hours/round) reannealing hybridization




was used.


BRSTNOT01
PBLUESCRIPT
Library was constructed using RNA isolated from the breast tissue




of a 56-year-old Caucasian female who died in a motor




vehicle accident.


BRSTNOT05
PSPORT1
Library was constructed using RNA isolated from breast tissue




removed from a 58-year-old Caucasian female during a




unilateral extended simple mastectomy. Pathology for the associated




tumor tissue indicated multicentric invasive grade 4 lobular




carcinoma. Patient history included skin cancer, rheumatic heart




disease, osteoarthritis, and tuberculosis. Family history




included cerebrovascular and cardiovascular disease, breast and




prostate cancer, and type I diabetes.


BRSTNOT07
pINCY
Library was constructed using RNA isolated from diseased breast




tissue removed from a 43-year-old Caucasian female during a




unilateral extended simple mastectomy. Pathology indicated mildly




proliferative fibrocystic changes with epithelial hyperplasia,




papillomatosis, and duct ectasia. Pathology for the associated




tumor tissue indicated invasive grade 4, nuclear grade 3 mammary




adenocarcinoma with extensive comedo necrosis. Family history




included epilepsy, cardiovascular disease, and type II diabetes.


BRSTNOT28
pINCY
Library was constructed using RNA isolated from diseased right




breast tissue removed from a 40-year-old Caucasian female




during a bilateral reduction mammoplasty. Pathology indicated




bilateral mild fibrocystic and proliferative changes. Patient




history included pure hypercholesterolemia. Family history




included acute myocardial infarction, atherosclerotic coronary artery




disease, type II diabetes, and prostate cancer.


BRSTTMR01
PCDNA2.1
This random primed library was constructed using RNA isolated




from right breast tissue removed from a 62-year-old Caucasian




female during open breast biopsy and unilateral extended simple




mastectomy. Pathology indicated benign breast parenchyma.




Pathology for the matched tumor tissue indicated residual grade




3 (of 4) ductal adenocarcinoma. The patient presented with




breast cancer. Patient history included benign neoplasm of the




large bowel and leg vein occlusion. Previous surgeries included




dilation and curettage and spinal canal exploration. Patient




medications included Lozal, Mevacor, and tamoxifen. Family history




included heart murmur in the mother; skin cancer in the sibling(s);




and prostate cancer in the grandfather.


BRSTTMT02
pINCY
Library was constructed using RNA isolated from diseased right




breast tissue removed from a 46-year-old Caucasian female




during a unilateral extended simple mastectomy and open breast




biopsy. Pathology indicated mildly proliferative fibrocystic




change, including intraductal duct ectasia, papilloma formation,




and ductal hyperplasia. Pathology for the associated tumor




tissue indicated multifocal ductal carcinoma in situ, both




comedo and non-comedo types, nuclear grade 2 with extensive




intraductal calcifications. Patient history included deficiency




anemia, normal delivery, chronic sinusitis, extrinsic asthma, and




kidney infection. Family history included type II diabetes,




benign hypertension, cerebrovascular disease, skin cancer, and




hyperlipidemia.


BSTMNON02
PSPORT1
This normalized brain stem library was constructed from 2.84




million independent clones from a brain stem library. Starting




RNA was made from the brain stem tissue of a 72-year-old Caucasian




male who died from myocardial infarction. Patient




history included coronary artery disease, insulin-dependent




diabetes mellitus, and arthritis. Normalization and hybridization




conditions were adapted from Soares et al. (PNAS (1994) 91: 9228).


CERVNOT01
PSPORT1
Library was constructed using RNA isolated from the uterine




cervical tissue of a 35-year-old Caucasian female during a vaginal




hysterectomy with dilation and curettage. Pathology indicated




mild chronic cervicitis. Family history included atherosclerotic




coronary artery disease and type II diabetes.


COLNNOT13
pINCY
Library was constructed using RNA isolated from ascending colon




tissue of a 28-year-old Caucasian male with moderate




chronic ulcerative colitis.


COLNNOT23
pINCY
Library was constructed using RNA isolated from diseased colon




tissue removed from a 16-year-old Caucasian male during a




total colectomy with abdominal/perineal resection. Pathology




indicated gastritis and pancolonitis consistent with the acute phase




of ulcerative colitis. Inflammation was more severe in the




transverse colon, with inflammation confined to the mucosa. There




was only mild involvement of the ascending and sigmoid colon,




and no significant involvement of the cecum, rectum, or




terminal ileum. Family history included irritable bowel syndrome.


COLXTDT01
pINCY
Library was constructed using RNA isolated from colon tissue




removed from the appendix of a 37-year-old Black female during




myomectomy, dilation and curettage, right fimbrial region biopsy,




and incidental appendectomy. Pathology indicated an




unremarkable appendix. Pathology for the associated tumor




tissue indicated multiple (12) uterine leiomyomata. Patient history




included premenopausal menorrhagia and sarcoidosis of the lung.




Family history included acute myocardial infarction and




atherosclerotic coronary artery disease.


CONUTUT01
pINCY
Library was constructed using RNA isolated from sigmoid mesentery




tumor tissue obtained from a 61-year-old female during a




total abdominal hysterectomy and bilateral salpingo-oophorectomy




with regional lymph node excision. Pathology indicated a




metastatic grade 4 malignant mixed mullerian tumor present in




the sigmoid mesentery at two sites.


DRGCNOT01
pINCY
Library was constructed using RNA isolated from dorsal root




ganglion tissue removed from the cervical spine of a 32-year-old




Caucasian male who died from acute pulmonary edema and




bronchopneumonia, bilateral pleural and pericardial effusions, and




malignant lymphoma (natural killer cell type). Patient history




included probable cytomegalovirus infection, hepatic congestion




and steatosis, splenomegaly, hemorrhagic cystitis, thyroid




hemorrhage, and Bell's palsy. Surgeries included colonoscopy, large




intestine biopsy, adenotonsillectomy, and nasopharyngeal endoscopy




and biopsy; treatment included radiation therapy.


HIPONON02
PSPORT1
This normalized hippocampus library was constructed from 1.13M




independent clones from a hippocampus tissue library. RNA




was isolated from the hippocampus tissue of a 72-year-old




Caucasian female who died from an intracranial bleed. Patient history




included nose cancer, hypertension, and arthritis. The normalization




and hybridization conditions were adapted from Soares et




al. (PNAS (1994) 91: 9228).


KERANOT02
PSPORT1
Library was constructed using RNA isolated from epidermal




breast keratinocytes (NHEK). NHEK (Clontech #CC-2501) is




human breast keratinocyte cell line derived from a 30-year-old black




female during breast-reduction surgery.


KIDNTUT15
pINCY
Library was constructed using RNA isolated from left kidney tumor




tissue removed from a 65-year-old Caucasian male during




an exploratory laparotomy and nephroureterectomy. Pathology




indicated grade 1 renal cell carcinoma within the upper pole of




the left kidney. Patient history included malignant melanoma of




the abdominal skin, benign neoplasm of colon, cerebrovascular




disease, and umbilical hernia. Family history included myocardial




infarction, atherosclerotic coronary artery disease, and




cerebrovascular disease, and prostate cancer.


LNODNOT12
pINCY
Library was constructed using RNA isolated from lymph node tissue




obtained from an 11-year-old Caucasian female who died




from a motor vehicle accident. Previous surgeries included tonsilectomy.


LUNGNON07
pINCY
This normalized lung tissue library was constructed from 5.1




million independent clones from a lung tissue library. Starting




RNA was made from RNA isolated from lung tissue. The library was




normalized in two rounds using conditions adapted from




Soares et al., PNAS (1994) 91: 9228-9232 and Bonaldo et al.,




Genome Research (1996) 6: 791, except that a significantly longer




(48 hours/round) reannealing hybridization was used.


MIXDTUE01
PBK-CMV
This 5′ biased random primed library was constructed using




pooled cDNA from seven donors. cDNA was generated using




mRNA isolated from placental tissue removed from a Caucasian




fetus (A), who died after 16 weeks' gestation from fetal demise




and hydrocephalus; from placental tissue removed from a Caucasian




male fetus (B), who died after 18 weeks' gestation from




fetal demise; from an untreated LNCaP cell line, derived from




prostate carcinoma with metastasis to the left supraclavicular




lymph nodes, removed from a 50-year-old Caucasian male (C);




from endometrial tissue removed from a 32-year-old female (D);




from diseased right ovary tissue removed from a 45-year-old




Caucasian female (E); from diseased right ovary tissue removed




from a 47-year-old Caucasian female (donor F) and from right




fallopian tube tumor tissue removed from an 85-year-old




Caucasian female (donor G). For donor A, patient history




included umbilical cord wrapped around the head (3 times) and the




shoulders (1 time). Serology was positive for anti-CMV. Family




history included multiple pregnancies and live births, and an




abortion in the mother. For donor B, serologies were negative.




For donor D, pathology indicated the endometrium was in




secretory phase. For donor E, pathology indicated stromal




hyperthecosis of the right and left ovaries. For donor F, pathology




indicated endometriosis. For donor G, pathology indicated poorly




differentiated mixed endometrioid (80%) and serous (20%)




adenocarcinoma of the right fallopian tube. Patient history




included medullary carcinoma of the thyroid.


MIXDUNB01
pINCY
Library was constructed using RNA isolated from myometrium




removed from a 41-year-old Caucasian female (A) during




vaginal hysterectomy with a dilatation and curettage and




untreated smooth muscle cells removed from the renal vein of a 57-




year-old Caucasian male. Pathology for donor A indicated




the myometrium and cervix were unremarkable. The endometrium




was secretory and contained fragments of endometrial polyps.




Benign endo- and ectocervical mucosa were identified in the




endocervix. Pathology for the associated tumor tissue




indicated uterine leiomyoma. Medical history included an unspecified




menstrual disorder, ventral hernia, normal delivery, a




benign ovarian neoplasm, and tobacco abuse in donor A. Previous




surgeries included a bilateral destruction of fallopian tubes,




removal of a solitary ovary, and an exploratory laparotomy in donor




A. Medications included ferrous sulfate in donor A.


MONOTXN03
pINCY
Normalized, treated monocyte tissue library was constructed from




7.6 million independent clones from a treated monocyte




library. Starting RNA was made from RNA isolated from treated




monocytes from peripheral blood obtained from a 42-year-old




female. The cells were treated with anti-interleukin-10 (anti-IL-10)




and lipopolysaccharide (LPS). The anti-IL-10 was added at




time 0 at 10 ng/ml and LPS was added at 1 hour at 5 ng/ml.




The monocytes were isolated from buffy coat by adherence to




plastic. Incubation time was 24 hours. cDNA synthesis was initiated




using a NotI-anchored oligo(dT) primer. The libraries were




normalized in two rounds using conditions adapted from Soares




et al., PNAS (1994) 91: 9228 and Bonaldo et al., Genome




Research (1996 6): 791, except that a significantly longer




(48-hours/round) reannealing hybridization was used. The libraries




were then linearized and recircularized to select for insert




containing clones as follows: plasmid DNA was prepped from




approximately 1 million clones from the normalized, treated




monocyte tissue libraries following soft agar transformation. The




DNA was linearized with NotI and insert containing clones were




size-selected by agarose gel electrophoresis and recircularized




by ligation.


NEUTFMT01
PBLUESCRIPT
Library was constructed using total RNA isolated from peripheral




blood granulocytes collected by density gradient centrifugation




through Ficoll-Hypaque. The cells were isolated from buffy coat




units obtained from unrelated male and female donors. Cells were




cultured in 10 nM fMLP for 30 minutes, lysed in GuSCN, and spun




through CsCl to obtain RNA for library construction. Because




this library was made from total RNA, it has an unusually high




proportion of unique singleton sequences, which may not all




come from polyA RNA species.


PANCTUT01
pINCY
Library was constructed using RNA isolated from pancreatic tumor




tissue removed from a 65-year-old Caucasian female during




radical subtotal pancreatectomy. Pathology indicated an invasive




grade 2 adenocarcinoma. Patient history included type II




diabetes, osteoarthritis, cardiovascular disease, benign neoplasm




in the large bowel, and a cataract. Previous surgeries included a




total splenectomy, cholecystectomy, and abdominal hysterectomy.




Family history included cardiovascular disease, type II




diabetes, and stomach cancer.


PITUNOT06
pINCY
Library was constructed using RNA isolated from pituitary gland




tissue removed from a 55-year-old male who died from




chronic obstructive pulmonary disease. Neuropathology indicated




there were no gross abnormalities, other than mild ventricular




enlargement. There was no apparent microscopic abnormality in




any of the neocortical areas examined, except for a number of




silver positive neurons with apical dendrite staining, particularly




in the frontal lobe. The significance of this was undetermined.




The only other microscopic abnormality was that there was




prominent silver staining with some swollen axons in the CA3




region of the anterior and posterior hippocampus. Microscopic




sections of the cerebellum revealed mild Bergmann's gliosis in




the Purkinje cell layer. Patient history included schizophrenia.


PROSNOT15
pINCY
Library was constructed using RNA isolated from diseased prostate




tissue removed from a 66-year-old Caucasian male during




radical prostatectomy and regional lymph node excision. Pathology




indicated adenofibromatous hyperplasia. Pathology for the




associated tumor tissue indicated an adenocarcinoma (Gleason




grade 2 + 3). The patient presented with elevated prostate specific




antigen (PSA). Family history included prostate cancer, secondary




bone cancer, and benign hypertension.


PROSTMT03
pINCY
The library was constructed using RNA isolated from right prostate




tissue removed from a 68-year-old Caucasian male during a




radical prostatectomy and regional lymph node excision. Pathology




for the associated tumor indicated adenocarcinoma, Gleason




grade 4 + 3, which formed a predominant mass involving the




left side peripherally. The patient presented with elevated prostate




specific antigen (PSA) and induration. Patient history included




pure hypercholesterolemia, kidney calculus, an unspecified




allergy, and atopicdermatitis. Family history included colon cancer.


PROSTUT09
pINCY
Library was constructed using RNA isolated from prostate tumor




tissue removed from a 66-year-old Caucasian male during a




radical prostatectomy, radical cystectomy, and urinary diversion.




Pathology indicated grade 3 transitional cell carcinoma. The




patient presented with prostatic inflammatory disease. Patient




history included lung neoplasm, and benign hypertension. Family




history included a malignant breast neoplasm, tuberculosis,




cerebrovascular disease, atherosclerotic coronary artery disease and




lung cancer.


SEMVTDE01
PCDNA2.1
This 5' biased random primed library was constructed using RNA




isolated from seminal vesicle tissue removed from a 63-year-




old Caucasian male during closed prostatic biopsy, radical




prostatectomy, and regional lymph node excision. Pathology for the




associated tumor tissue indicated Gleason grade 2 + 3




adenocarcinoma in the right side of the prostate. Adenofibromatous




hyperplasia was present. The patient presented with prostate




cancer, elevated prostate specific antigen and prostatic hyperplasia.




Patient history included kidney calculus, extrinsic asthma, benign




bowel neoplasm, backache, tremor, and tobacco abuse in




remission. Previous surgeries included adenotonsillectomy.




Patient medications included Ventolin and Vanceril. Family history




included atherosclerotic coronary artery disease and acute




myocardial infarction in the mother; atherosclerotic coronary artery




disease and acute myocardial infarction in the father; and




stomach cancer and extrinsic asthma in the grandparent(s).


SINIUCT01
pINCY
Library was constructed using RNA isolated from ileum tissue




obtained from a 42-year-old Caucasian male during a total intra-




abdominal colectomy and endoscopic jejunostomy. Previous surgeries




included polypectomy, colonoscopy, and spinal canal




exploration. Family history included cerebrovascular disease,




benign hypertension, atherosclerotic coronary artery disease, and




type II diabetes.


SINTBST01
pINCY
Library was constructed using RNA isolated from ileum tissue




obtained from an 18-year-old Caucasian female during bowel




anastomosis. Pathology indicated Crohn's disease of the ileum,




involving 15 cm of the small bowel. Family history included




cerebrovascular disease and atherosclerotic coronary artery disease.


SINTFER03
PCDNA2.1
This random primed library was constructed using RNA isolated




from small intestine tissue removed from a Caucasian male




fetus who died from fetal demise.


TESTNOT11
pINCY
Library was constructed using RNA isolated from testicular tissue




removed from a 16-year-old Caucasian male who died from




hanging. Patient history included drug use (tobacco, marijuana,




and cocaine use), and medications included Lithium, Ritalin,




and Paxil.


THYMNON04
PSPORT1
This normalized library was constructed from a thymus tissue




library. Starting RNA was made from thymus tissue removed




from a 3-year-old Caucasian male, who died from anoxia.




Serologies were negative. The patient was not taking any




medications. The library was normalized in two rounds using




conditions adapted from Soares et al., PNAS (1994) 91: 9228 and




Bonaldo et al., Genome Research (1996) 6: 791, except that a




significantly longer (48-hours/round) reannealing hybridization




was used.


THYMNOT02
PBLUESCRIPT
Library was constructed using polyA RNA isolated from thymus




tissue removed from a 3-year-old Caucasian male, who died




from drowning. Serologies were negative.


THYRNOT03
pINCY
Library was constructed using RNA isolated from thyroid tissue




removed from the left thyroid of a 28-year-old Caucasian




female during a complete thyroidectomy. Pathology indicated a




small nodule of adenomatous hyperplasia present in the left




thyroid. Pathology for the associated tumor tissue indicated




dominant follicular adenoma, forming a well-encapsulated mass in




the left thyroid.


UTRENON03
pINCY
This normalized library was constructed from 1.2 × 10e7




independent clones from a uterine endometrial tissue library. Starting




RNA was made from uterine endometrium tissue obtained from a




29-year-old Caucasian female during a vaginal hysterectomy




and cystocele repair. Pathology indicated the endometrium was




secretory and the cervix showed mild chronic cervicitis with




focal squamous metaplasia. Pathology for the associated tumor




tissue indicated an intramural uterine leiomyoma. Patient history




included hypothyroidism, pelvic floor relaxation, incomplete




T-12 injury (due to a motor vehicle accident) causing paraplegia




and self catheterization. Previous surgeries included a normal




delivery, a laminectomy, and a rhinoplasty. Family history




included benign hypertension, type II diabetes, and hyperlipidemia.




The libraries were normalized in two rounds using




conditions adapted from Soares et al., PNAS (1994) 91: 9228 and




Bonaldo et al., Genome Research (1996) 6: 791, except that a




significantly longer (48 hours/round) reannealing hybridization was used.


UTRSTMR02
PCDNA2.1
This random primed library was constructed using pooled cDNA




from two different donors. cDNA was generated using mRNA




isolated from endometrial tissue removed from a 32-year-old female




(donor A) and using mRNA isolated from myometrium




removed from a 45-year-old female (donor B) during vaginal




hysterectomy and bilateral salpingo-oophorectomy. In donor A,




pathology indicated the endometrium was secretory phase. The




cervix showed severe dysplasia (CIN III) focally involving the




squamocolumnar junction at the 1, 6 and 7 o'clock positions.




Mild koilocytotic dysplasia was also identified within the cervix. In




donor B, pathology for the matched tumor tissue indicated multiple




(23) subserosal, intramural, and submucosal leiomyomata.




Patient history included stress incontinence, extrinsic asthma




without status asthmaticus and normal delivery in donor B. Family




history included cerebrovascular disease, depression, and




atherosclerotic coronary artery disease in donor B.



















TABLE 7








Program
Description
Reference
Parameter Threshold







ABI FACTURA
A program that removes vector sequences and masks
Applied Biosystems,




ambiguous bases in nucleic acid sequences.
Foster City, CA.


ABI/PARACEL
A Fast Data Finder useful in comparing and
Applied Biosystems,
Mismatch < 50%


FDF
annotating amino acid or nucleic acid sequences.
Foster City, CA;




Paracel Inc.,




Pasadena, CA.


ABI
A program that assembles nucleic acid sequences.
Applied Biosystems,


AutoAssembler

Foster City, CA.


BLAST
A Basic Local Alignment Search Tool useful in
Altschul, S. F. et al.
ESTs: Probability



sequence similarity search for amino acid and
(1990) J. Mol. Biol.
value = 1.0E−8



nucleic acid sequences. BLAST includes five
215: 403-410;
or less; Full



functions: blastp, blastn, blastx, tblastn,
Altschul, S. F. et al.
Length sequences:



and tblastx.
(1997) Nucleic Acids
Probability value =




Res. 25: 3389-3402.
1.0E−10 or less


FASTA
A Pearson and Lipman algorithm that searches
Pearson, W. R. and
ESTs: fasta E



for similarity between a query sequence and
D. J. Lipman (1988)
value = 1.06E−6;



a group of sequences of the same type.
Proc. Natl. Acad Sci.
Assembled ESTs:



FASTA comprises as least five functions:
USA 85: 2444-2448;
fasta Identity =



fasta, tfasta, fastx, tfastx, and
Pearson, W. R. (1990)
95% or greater



ssearch.
Methods Enzymol. 183:
and Match




63-98; and Smith,
length = 200




T. F. and M. S. Waterman
bases or greater;




(1981) Adv. Appl. Math.
fastx E value =




2: 482-489.
1.0E−8 or less;





Full Length





sequences: fastx





score = 100





or greater


BLIMPS
A BLocks IMProved Searcher that matches a
Henikoff, S. and J. G.
Probability



sequence against those in BLOCKS, PRINTS,
Henikoff (1991)
value = 1.0E−3 or



DOMO, PRODOM, and PFAM databases to search
Nucleic Acids Res. 19:
less



for gene families, sequence homology, and
6565-6572; Henikoff,



structural fingerprint regions.
J. G. and S. Henikoff




(1996) Methods Enzymol.




266: 88-105; and




Attwood, T. K. et




al. (1997) J. Chem.




Inf. Comput. Sci. 37:




417-424.


HMMER
An algorithm for searching a query sequence
Krogh, A. et al. (1994)
PFAM hits: Probability



against hidden Markov model (HMM)-based
J. Mol. Biol.
value = 1.0E−3



databases of protein family consensus sequences,
235: 1501-1531;
or less; Signal



such as PFAM.
Sonnhammer, E. L. L.
peptide hits:




et al. (1988) Nucleic
Score = 0 or greater




Acids Res. 26: 320-322;




Durbin, R. et al.




(1998) Our World View,




in a Nutshell, Cambridge




Univ. Press, pp. 1-350.


ProfileScan
An algorithm that searches for structural and
Gribskov, M. et al.
Normalized quality



sequence motifs in protein sequences that match
(1988) CABIOS 4: 61-66;
score ≦ GCG



sequence patterns defined in Prosite.
Gribskov, M. et al.
specified “HIGH”




(1989) Methods
value for that




Enzymol. 183: 146-159;
particular Prosite




Bairoch, A. et al.
motif.




(1997) Nucleic Acids Res.
Generally, score =




25: 217-221.
1.4-2.1.


Phred
A base-calling algorithm that examines automated
Ewing, B. et al. (1998)



sequencer traces with high sensitivity and
Genome Res. 8: 175-185;



probability.
Ewing, B. and P. Green




(1998) Genome




Res. 8: 186-194.


Phrap
A Phils Revised Assembly Program including
Smith, T. F. and M. S.
Score = 120 or



SWAT and CrossMatch, programs based on efficient
Waterman (1981) Adv.
greater; Match



implementation of the Smith-Waterman algorithm,
Appl. Math. 2: 482-489;
length = 56 or



useful in searching sequence homology and
Smith, T. F. and M. S.
greater



assembling DNA sequences.
Waterman (1981) J. Mol.




Biol. 147: 195-197;




and Green, P.,




University of Washington,




Seattle, WA.


Consed
A graphical tool for viewing and editing Phrap
Gordon, D. et al. (1998)



assemblies.
Genome Res. 8: 195-202.


SPScan
A weight matrix analysis program that scans
Nielson, H. et al. (1997)
Score = 3.5



protein sequences for the presence of secretory
Protein Engineering 10:
or greater



signal peptides.
1-6; Claverie, J. M.




and S. Audic (1997)




CABIOS 12: 431-439.


TMAP
A program that uses weight matrices to delineate
Persson, B. and P. Argos



transmembrane segments on protein sequences and
(1994) J. Mol. Biol.



determine orientation.
237: 182-192; Persson,




B. and P. Argos (1996)




Protein Sci. 5: 363-371.


TMHMMER
A program that uses a hidden Markov model (HMM)
Sonnhammer, E. L. et al.



to delineate transmembrane segments on protein
(1998) Proc. Sixth



sequences and determine orientation.
Intl. Conf. On Intelligent




Systems for Mol.




Biol., Glasgow et al.,




eds., The Am. Assoc. for




Artificial Intelligence




Press, Menlo Park,




CA, pp. 175-182.


Motifs
A program that searches amino acid sequences for
Bairoch, A. et al. (1997)



patterns that matched those defined in Prosite.
Nucleic Acids Res.




25: 217-221; Wisconsin




Package Program Manual,




version 9, page M51-59,




Genetics Computer Group,




Madison, WI.









Claims
  • 1. An isolated polypeptide selected from the group consisting of: a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-53, c) a polypeptide consisting essentially of a naturally occurring amino acid sequence at least 91% identical to the amino acid sequence of SEQ ID NO:54, d) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, and e) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 2. An isolated polypeptide of claim 1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 3. An isolated polynucleotide encoding a polypeptide of claim 1.
  • 4. An isolated polynucleotide encoding a polypeptide of claim 2.
  • 5. An isolated polynucleotide of claim 4 comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108.
  • 6. A recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide of claim 3.
  • 7. A cell transformed with a recombinant polynucleotide of claim 6.
  • 8. A transgenic organism comprising a recombinant polynucleotide of claim 6.
  • 9. A method of producing a polypeptide of claim 1, the method comprising: a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide comprises a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 1, and b) recovering the polypeptide so expressed.
  • 10. A method of claim 9, wherein the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 11. An isolated antibody which specifically binds to a polypeptide of claim 1.
  • 12. An isolated polynucleotide selected from the group consisting of: a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:55-108, c) a polynucleotide complementary to a polynucleotide of a), d) a polynucleotide complementary to a polynucleotide of b), and e) an RNA equivalent of a)-d).
  • 13. An isolated polynucleotide comprising at least 60 contiguous nucleotides of a polynucleotide of claim 12.
  • 14. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising: a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and, optionally, if present, the amount thereof.
  • 15. A method of claim 14, wherein the probe comprises at least 60 contiguous nucleotides.
  • 16. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising: a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
  • 17. A composition comprising a polypeptide of claim 1 and a pharmaceutically acceptable excipient.
  • 18. A composition of claim 17, wherein the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 19. A method for treating a disease or condition associated with decreased expression of functional SECP, comprising administering to a patient in need of such treatment the composition of claim 17.
  • 20. A method of screening a compound for effectiveness as an agonist of a polypeptide of claim 1, the method comprising: a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting agonist activity in the sample.
  • 21. A composition comprising an agonist compound identified by a method of claim 20 and a pharmaceutically acceptable excipient.
  • 22. A method for treating a disease or condition associated with decreased expression of functional SECP, comprising administering to a patient in need of such treatment a composition of claim 21.
  • 23. A method of screening a compound for effectiveness as an antagonist of a polypeptide of claim 1, the method comprising: a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting antagonist activity in the sample.
  • 24. A composition comprising an antagonist compound identified by a method of claim 23 and a pharmaceutically acceptable excipient.
  • 25. A method for treating a disease or condition associated with overexpression of functional SECP, comprising administering to a patient in need of such treatment a composition of claim 24.
  • 26. A method of screening for a compound that specifically binds to the polypeptide of claim 1, the method comprising: a) combining the polypeptide of claim 1 with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide of claim 1 to the test compound, thereby identifying a compound that specifically binds to the polypeptide of claim 1.
  • 27. A method of screening for a compound that modulates the activity of the polypeptide of claim 1, the method comprising: a) combining the polypeptide of claim 1 with at least one test compound under conditions permissive for the activity of the polypeptide of claim 1, b) assessing the activity of the polypeptide of claim 1 in the presence of the test compound, and c) comparing the activity of the polypeptide of claim 1 in the presence of the test compound with the activity of the polypeptide of claim 1 in the absence of the test compound, wherein a change in the activity of the polypeptide of claim 1 in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide of claim 1.
  • 28. A method of screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a sequence of claim 5, the method comprising: a) exposing a sample comprising the target polynucleotide to a compound, under conditions suitable for the expression of the target polynucleotide, b) detecting altered expression of the target polynucleotide, and c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.
  • 29. A method of assessing toxicity of a test compound, the method comprising: a) treating a biological sample containing nucleic acids with the test compound, b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 12 under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide comprising a polynucleotide sequence of a polynucleotide of claim 12 or fragment thereof, c) quantifying the amount of hybridization complex, and d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.
  • 30. A diagnostic test for a condition or disease associated with the expression of SECP in a biological sample, the method comprising: a) combining the biological sample with an antibody of claim 11, under conditions suitable for the antibody to bind the polypeptide and form an antibody:polypeptide complex, and b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
  • 31. The antibody of claim 11, wherein the antibody is: a) a chimeric antibody, b) a single chain antibody, c) a Fab fragment, d) a F(ab′)2 fragment, or e) a humanized antibody.
  • 32. A composition comprising an antibody of claim 11 and an acceptable excipient.
  • 33. A method of diagnosing a condition or disease associated with the expression of SECP in a subject, comprising administering to said subject an effective amount of the composition of claim 32.
  • 34. A composition of claim 32, wherein the antibody is labeled.
  • 35. A method of diagnosing a condition or disease associated with the expression of SECP in a subject, comprising administering to said subject an effective amount of the composition of claim 34.
  • 36. A method of preparing a polyclonal antibody with the specificity of the antibody of claim 11, the method comprising: a) immunizing an animal with a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibodies from said animal, and c) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 37. A polyclonal antibody produced by a method of claim 36.
  • 38. A composition comprising the polyclonal antibody of claim 37 and a suitable carrier.
  • 39. A method of making a monoclonal antibody with the specificity of the antibody of claim 11, the method comprising: a) immunizing an animal with a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NO:1-54, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibody producing cells from the animal, c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells, d) culturing the hybridoma cells, and e) isolating from the culture monoclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 40. A monoclonal antibody produced by a method of claim 39.
  • 41. A composition comprising the monoclonal antibody of clam 40 and a suitable carrier.
  • 42. The antibody of claim 11, wherein the antibody is produced by screening a Fab expression library.
  • 43. The antibody of claim 11, wherein the antibody is produced by screening a recombinant immunoglobulin library.
  • 44. A method of detecting a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54 in a sample, the method comprising: a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and b) detecting specific binding, wherein specific binding indicates the presence of a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54 in the sample.
  • 45. A method of purifying a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54 from a sample, the method comprising: a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and b) separating the antibody from the sample and obtaining the purified polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-54.
  • 46. A microarray wherein at least one element of the microarray is a polynucleotide of claim 13.
  • 47. A method of generating an expression profile of a sample which contains polynucleotides, the method comprising: a) labeling the polynucleotides of the sample, b) contacting the elements of the microarray of claim 46 with the labeled polynucleotides of the sample under conditions suitable for the formation of a hybridization complex, and c) quantifying the expression of the polynucleotides in the sample.
  • 48. An array comprising different nucleotide molecules affixed in distinct physical locations on a solid substrate, wherein at least one of said nucleotide molecules comprises a first oligonucleotide or polynucleotide sequence specifically hybridizable with at least 30 contiguous nucleotides of a target polynucleotide, and wherein said target polynucleotide is a polynucleotide of claim 12.
  • 49. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 30 contiguous nucleotides of said target polynucleotide.
  • 50. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 60 contiguous nucleotides of said target polynucleotide.
  • 51. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to said target polynucleotide.
  • 52. An array of claim 48, which is a microarray.
  • 53. An array of claim 48, further comprising said target polynucleotide hybridized to a nucleotide molecule comprising said first oligonucleotide or polynucleotide sequence.
  • 54. An array of claim 48, wherein a linker joins at least one of said nucleotide molecules to said solid substrate.
  • 55. An array of claim 48, wherein each distinct physical location on the substrate contains multiple nucleotide molecules, and the multiple nucleotide molecules at any single distinct physical location have the same sequence, and each distinct physical location on the substrate contains nucleotide molecules having a sequence which differs from the sequence of nucleotide molecules at another distinct physical location on the substrate.
  • 56. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:1.
  • 57. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:2.
  • 58. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:3.
  • 59. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID. NO:4.
  • 60. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:5.
  • 61. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:6.
  • 62. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:7.
  • 63. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:8.
  • 64. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:9.
  • 65. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:10.
  • 66. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:11.
  • 67. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:12.
  • 68. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:13.
  • 69. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:14.
  • 70. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:15.
  • 71. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:16.
  • 72. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:17.
  • 73. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:18.
  • 74. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:19.
  • 75. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:20.
  • 76. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:21.
  • 77. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:22.
  • 78. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:23.
  • 79. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:24.
  • 80. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:25.
  • 81. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:26.
  • 82. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:27.
  • 83. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:28.
  • 84. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:29.
  • 85. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:30.
  • 86. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:31.
  • 87. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:32.
  • 88. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:33.
  • 89. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:34.
  • 90. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:35.
  • 91. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ED NO:36.
  • 92. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:37.
  • 93. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:38.
  • 94. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:39.
  • 95. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:40.
  • 96. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:41.
  • 97. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:42.
  • 98. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:43.
  • 99. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:44.
  • 100. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:45.
  • 101. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:46.
  • 102. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:47.
  • 103. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:48.
  • 104. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:49.
  • 105. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:50.
  • 106. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:51.
  • 107. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:52.
  • 108. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:53.
  • 109. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 54.
  • 110. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:55.
  • 111. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:56.
  • 112. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:57.
  • 113. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:58.
  • 114. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:59.
  • 115. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:59.
  • 116. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:61.
  • 117. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:62.
  • 118. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:63.
  • 119. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:64.
  • 120. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:65.
  • 121. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:66.
  • 122. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:67.
  • 123. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:68.
  • 124. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:69.
  • 125. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:70.
  • 126. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:71.
  • 127. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:71.
  • 128. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:73.
  • 129. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:74.
  • 130. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:75.
  • 131. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:76.
  • 132. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:77.
  • 133. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:78.
  • 134. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:79.
  • 135. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:80.
  • 136. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:81.
  • 137. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:82.
  • 138. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:83.
  • 139. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:84.
  • 140. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:85.
  • 141. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:86.
  • 142. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:87.
  • 143. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:88.
  • 144. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:89.
  • 145. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:90.
  • 146. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:91.
  • 147. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:92.
  • 148. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:93.
  • 149. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:94.
  • 150. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:95.
  • 151. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO-96.
  • 152. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:97.
  • 153. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:98.
  • 154. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:99.
  • 155. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:100.
  • 156. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:101.
  • 157. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:102.
  • 158. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:103.
  • 159. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:104.
  • 160. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:105.
  • 161. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:106.
  • 162. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:107.
  • 163. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:108.
PCT Information
Filing Document Filing Date Country Kind
PCT/US01/48517 12/12/2001 WO
Provisional Applications (7)
Number Date Country
60255639 Dec 2000 US
60257852 Dec 2000 US
60260105 Jan 2001 US
60262932 Jan 2001 US
60263096 Jan 2001 US
60263090 Jan 2001 US
60265926 Feb 2001 US