NZ540754A - Promotion or inhibition of angiogenesis and cardiovascularization - Google Patents

Promotion or inhibition of angiogenesis and cardiovascularization

Info

Publication number
NZ540754A
NZ540754A NZ54075499A NZ54075499A NZ540754A NZ 540754 A NZ540754 A NZ 540754A NZ 54075499 A NZ54075499 A NZ 54075499A NZ 54075499 A NZ54075499 A NZ 54075499A NZ 540754 A NZ540754 A NZ 540754A
Authority
NZ
New Zealand
Prior art keywords
polypeptide
seq
nucleic acid
acid sequence
antibody
Prior art date
Application number
NZ54075499A
Inventor
Kevin P Baker
Napoleone Ferrara
Hanspeter Gerber
Audrey Goddard
Paul J Godowski
Austin L Gurney
Kenneth J Hillan
Robert D Klein
Sophia S Kuo
Nicholas F Paoni
Victoria Smith
Colin K Watanabe
P Mickey Williams
William I Wood
Avi Ashkenazi
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Publication of NZ540754A publication Critical patent/NZ540754A/en

Links

Abstract

A composition comprises a polypeptide which has at least 80% sequence identity to an SEQ ID No: 216 or an antibody which specifically binds to the polypeptide. The polypeptide is useful for the promotion or inhibition of angiogenesis and cardiovascularization. (62) Divided Out of 535590

Description

<div class="application article clearfix" id="description"> <p class="printTableText" lang="en">540754 <br><br> ceW.65- <br><br> NEW ZEALAND PATENTS ACT, 1953 <br><br> No: Divided out of NZ 535590 <br><br> Date: Dated 30 November 1999 <br><br> COMPLETE SPECIFICATION <br><br> PROMOTION OR INHIBITION OF ANGIOGENESIS AND CARDIOVASCULARIZATION <br><br> We, GENENTECH, INC., 1 DNA Way, South San Francisco, California 94080-4990, USA, do hereby declare the invention for which we pray that a patent may be granted to us, and the method by which it is to be performed, to be particularly described in and by the following statement: <br><br> - 1 - <br><br> (followed by la) <br><br> PROMOTION OR INHIBITION OF ANGIOGENESIS AND CARDIOVASCULARIZATION <br><br> This application is a divisional of New Zealand specification No. 535590. <br><br> Background of the Invention <br><br> Field of the Invention <br><br> The present invention relates to compositions and methods useful for promoting or inhibiting angiogenesis and/or cardiovascularization in mammals in need of such biological effect. This includes the diagnosis and treatment of cardiovascular disorders as well as oncological disorders. <br><br> Description of Background <br><br> A. Cardiac Disorders and Factors <br><br> Heart failure affects approximately five million Americans, and new cases of heart failure number about 400.000 each year. It is the single most frequent cause of hospitalization for people age 65 and older in the United States. Recent advances in the management of acute cardiac diseases, including acute myocardial infarction, are 15 resulting in an expanding patient population that will eventually develop chronic heart failure. From 1979 to 1995, hospitalizations for congestive heart failure (CHF) rose from 377,000 to 872,000 (a 130 percent increase) and CHF deaths increased 116 percent. <br><br> CHF is a syndrome characterized by left ventricular dysfunction, reduced exercise tolerance, impaired quality of life, and markedly shortened life expectancy. The sine qua non of heart failure is an inability of the heart 20 to pump blood at a rate sufficient to meet the metabolic needs of the body's tissues (in other words, there is insufficient cardiac output). <br><br> At least four major compensatory mechanisms are activated in the setting Of heart failure to boost cardiac output, including peripheral vasoconstriction, increased heart rate, increased cardiac contractility, and increased plasma volume. These effects are mediated primarily by the sympathetic nervous system and the renin-angiotensin 25 system. See. Eichhorn, American Journal of Medicine. 104: 163-169 (1998). Increased output from the sympathetic nervous system increases vascular tone, heart rate, and contractility. Angiotensin II elevates blood pressure by 1) directly stimulating vascular smooth muscle contraction, 2) promoting plasma volume expansion by stimulating aldosterone and antidiuretic hormone secretion, 3) stimulating sympathetic-mediated vascular tone, and 4) catalyzing the degradation of bradykinin, which has vasodilatory and natriuretic activity. See, review by 30 Brown and Vaughan, Circulation. 97: 1411-1420 (1998). As noted below, angiotensin 11 may also have directly deleterious effects on the heart by promoting myocyte necrosis (impairing systolic function) and intracardiac fibrosis (impairing diastolic and in some cases systolic function). See, Weber, Circulation. 96:4065-4082 (1998). <br><br> A consistent feature of congestive heart failure (CHF) is cardiac hypertrophy, an enlargement of the heart la <br><br> 1 5 JUH 2008 RECEIVED <br><br> that is activated by both mechanical and hormonal stimuli and enables the heart to adapt to demands for increased cardiac output. Morgan and Baker. Circulation. 83: 13-25 (1991). This hypertrophic response is frequently associated with a variety of distinct pathological conditions such as hypertension, aortic stenosis, myocardial infarction, cardiomyopathy, valvular regurgitation, and intracardiac shunt, all of which result in chronic hemodynamic overload. <br><br> Hypertrophy is generally defined as an increase in size of an organ or structure independent of natural growth that does not involve tumor formation. Hypertrophy of the heart is due either to an increase in the mass of the individual cells (myocytes), or to an increase in the number of cells making up the tissue (hyperplasia), or both. While the enlargement of an embryonic heart is largely dependent on an increase in myocyte number (which continues until shortly after birth), post-natal cardiac myocytes lose their proliferative capacity. Further growth occurs through hypertrophy of the individual cells. <br><br> Adult myocyte hypertrophy is initially beneficial as a short term response to impaired cardiac function by permitting a decrease in the load on individual muscle fibers. With severe, long-standing overload, however, the hypertrophied cells begin to deteriorate and die. Katz, "Heart Failure", in: Katz A.M. ed., Physiology of the Heart (New York: Raven Press, 1992) pp. 638-668. Cardiac hypertrophy is a significant risk factor for both mortality and morbidity in the clinical course of heart failure. Katz. Trends Cardiovasc. Med.. 5: 37-44 (1995). For further details of the causes and pathology of cardiac hypertrophy see, e.g., Heart Disease. A Textbook of Cardiovascular Medicine. Braunwald, E. ed. (W.B. Saunders Co., 1988), Chapter 14, "Pathophysiology of Heart Failure." <br><br> On a cellular level, the heart is composed of myocytes and surrounding support cells, generically called non-myocytes. While non-myocytes are primarily fibroblast/mesenchymal cells, they also include endothelial and smooth muscle cells. Indeed, although myocytes make up most of the adult myocardial mass, they represent only about 30% of the total cell numbers present in heart. In response to hormonal, physiological, hemodynamic, and pathological stimuli, adult ventricular muscle cells can adapt to increased workloads through the activation of a hypertrophic process. This response is characterized by an increase in myocyte cell size and contractile protein content of individual cardiac muscle cells, without concomitant cell division and activation of embryonic genes, including the gene for atrial natriuretic peptide (ANP). Chien et al., FASEB J.. 5: 3037-3046 (1991); Chien el al., Annu. Rev. PhvsioL 55: 77-95 (1993). An increment in myocardial mass as a result of an increase in myocyte size that is associated with an accumulation of interstitial collagen within the extracellular matrix and around intramyocardial coronary arteries has been described in left ventricular hypertrophy secondary to pressure overload in humans. Caspari et al., Cardiovasc. Res.. H.: 554-558 (1977); Schwarz et al., Am. J. Cardiol.. 42: 895-903 (1978); Hess et al., Circulation. 63: 360-371 (1981); Pearlman et al., Lab. Invest.. 46: 158-164 (1982). <br><br> It has also been suggested that paracrine factors produced by non-myocyte supporting cells may additionally be involved in the development of cardiac hypertrophy, and various non-myocyte derived hypertrophic factors, such as, leukocyte inhibitory factor (LIF) and endothelin, have been identified. Metcalf, Growth Factors. 7: 169-173 (1992); Kurzrock etal.. Endocrine Reviews. 12: 208-217 (1991); lnoue et al., Proc. Natl. Acad. Sci. USA. 86; 2863-2867 (1989); Yanagisawa and Masaki, Trends Pharm. Sci.. K): 374-378 (1989); U.S. Patent No. 5,573,762 (issued November 12,1996). Further exemplary factors that have been identified as potential mediators <br><br> of cardiac hypertrophy include cardiotrophin-1 (CT-1) (Pennica et al., Proc. Nat. Acad. Sci. USA. 92: 1142-1146 <br><br> (1995)), catecholamines, adrenocorticosteroids, angiotensin, and prostaglandins. <br><br> At present, the treatment of cardiac hypertrophy varies depending on the underlying cardiac disease. Catecholamines, adrenocorticosteroids, angiotensin, prostaglandins, LIF, endothelin (including endothelin-1, -2, and -3 and big endothelin), and CT-1 are among the factors identified as potential mediators of hypertrophy. For example, beta-adrenergic receptor blocking drugs (beta-blockers, e.g., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, carvedilol, etc.) and verapamil have been used extensively in the treatment of hypertrophic cardiomyopathy. The beneficial effects of beta-blockers on symptoms (e.g., chest pain) and exercise tolerance are largely due to a decrease in the heart rate with a consequent prolongation of diastole and increased passive ventricular filling. Thompson et al., Br. Heart J.. 44:488-98 (1980); Harrison et al., Circulation. 29: 84-98 (1964). Verapamil has been described to improve ventricular filling and probably reducing myocardial ischemia. Bonow et al.. Circulation. 72: 853-64 (1985). <br><br> Nifedipine and diltiazem have also been used occasionally in the treatment of hypertrophic cardiomyopathy. Lorell et al.. Circulation. 65: 499-507 (1982); Betocchi et al., Am. J. Cardiol.. 78: 451-457 <br><br> (1996). However, because of its potent vasodilating properties, nifedipine may be harmful, especially in patients with outflow obstruction. Disopyramide has been used to relieve symptoms by virtue of its negative inotropic properties. Pollick, N.Engl. J. Med.. 307:997-999 (1982). In many patients, however, the initial benefits decrease with time. Wigle et al.. Circulation. 92: 1680-1692(1995). Antihypertensive drug therapy has been reported to have beneficial effects on cardiac hypertrophy associated with elevated blood pressure. Examples of drugs used in antihypertensive therapy, alone or in combination, are calcium antagonists, e.g.. nitrendipine; adrenergic receptor blocking agents, e.g., those listed above; angiotensin converting enzyme (ACE) inhibitors such as quinapril, captopril, enalapril, ramipril. benazepril, fosinopril, and lisinopril; diuretics, e.g., chlorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide,and indapamide; and calcium channel blockers, e.g., diltiazem. nifedipine, verapamil, and nicardipine. <br><br> For example, treatment of hypertension with diltiazem and captopril showed a decrease in left ventricular muscle mass, but the Doppler indices of diastolic function did not normalize. Szlachcic et al., Am. J. Cardiol.. 63: 198-201 (1989); Shahi et al., Lancet. 336: 458-461 (1990). These findings were interpreted to indicate that excessive amounts of interstitial collagen may remain after regression of left ventricular hypertrophy. Rossi et al., Am. Heart J.. 124: 700-709 (1992). Rossi et al., supra, investigated the effect of captopril on the prevention and regression of myocardial cell hypertrophy and interstitial fibrosis in pressure overload cardiac hypertrophy, in experimental rats. <br><br> Agents that increase cardiac contractility directly (iontropic agents) were initially thought to benefit patients with heart failure because they improved cardiac output in the short term. However, all positive inotropic agents except digoxigenin have been found to result in increased long-term mortality, in spite of short-term improvements in cardiac performance. Massie, Curr. Op. in Cardiology. 12:209-217 (1997); Reddy et al., Curr. Opin. Cardiol.. 12: 233-241 (1997). Beta-adrenergic receptor blockers have recently been advocated for use in heart failure. Evidence from clinical trials suggests that improvements in cardiac function can be achieved without <br><br> increased mortality, though documented improvements patient survival have not yet been demonstrated. Seealso, U.S. Pat. Nos. 5,935,924, 5,624,806; 5,661,122; and 5,610,134 and WO 95/28173 regarding the use of cardiotropic 1 or antagonists thereof, or growth hormone and/or insulin-like growth factor-I in the treatment of CHF. Another treatment modality is heart transplantation, but this is limited by the availability of donor hearts. <br><br> Endothelin is a vasoconstricting peptide comprising 21 amino acids, isolated from swine arterial endothelial culture supernatant and structurally determined. Yanagisawa et al., Nature. 332: 411-415 (1988). Endothelin was later found to exhibit various actions, and endothelin antibodies as endothelin antagonists have proven effective in the treatment of myocardial infarction, renal failure, and other diseases. Since endothelin is present in live bodies and exhibits vasoconstricting action, it is expected to be an endogenous factor involved in the regulation of the circulatory system, and may be associated with hypertension, cardiovascular diseases such as myocardial infarction, and renal diseases such as acute renal failure. Endothelin antagonists are described, for example, in U.S. Pat. No. 5,773,414: JP Pat. Publ. 3130299/1991, EP 457,195: EP 460,679; and EP 552,489. A new endothelin B receptor for identifying endothelin receptor antagonists is described in U.S. Pat. No. 5,773,223. <br><br> Current therapy for heart failure is primarily directed to using angiotensin-converting enzyme (ACE) inhibitors, such as captopril, and diuretics. These drugs improve hemodynamic profile and exercise tolerance and reduce the incidence of morbidity and mortality in patients with CHF. Kramer et al., Circulation. 67(4): 807-816 (1983); Captopril Multicenter Research Group, J.A.C.C.. 2(4): 755-763 (1983): The CONSENSUS Trial Study Group, N.Ensl. J. Med.. 316(23): 1429-1435 (1987); The SOLVD Investigators, N.Engl. J. Med.. 325(5): 293-302 (1991). Further, they are useful in treating hypertension, left ventricular dysfunction, atherosclerotic vascular disease, and diabetic nephropathy. Brown and Vaughan, supra. However, despite proven efficacy, response to ACE inhibitors has been limited. For example, while prolonging survival in the setting of heart failure, ACE inhibitors appear to slow the progression towards end-stage heart failure, and substantial numbers of patients on ACE inhibitors have functional class III heart failure. <br><br> Moreover, improvement of functional capacity and exercise time is only small and mortality, although reduced, continues to be high. The CONSENSUS Trial Study Group, N. Engl. J. Med.. 316(23): 1429-1453(1987); The SOLVD Investigators, N. Engl. J. Med.. 325(5): 293-302 (1991); Cohn et al., N. Ensl. J. Med.. 325(5): 303-310 (1991); The Captopril-Digoxin Multicenter Research Group, JAMA. 259(4): 539-544 (1988). Hence, ACE inhibitors consistently appear unable to relieve symptoms in more than 60% of heart failure patients and reduce mortality of heart failure only by approximately 15-20%. For further adverse effects, see Brown and Vaughan, supra. <br><br> An alternative to ACE inhibitors is represented by specific ATI receptor antagonists. Clinical studies are planned to compare the efficacy of these two modalities in the treatment of cardiovascular and renal disease. However, animal model data suggests that the ACE/Ang II pathway, while clearly involved in cardiac hypertrophy, is not the only, or even the primary pathway active in this role. Mouse genetic "knockout" models have been made to test individual components of the pathway. In one such model, the primary cardiac receptor for Ang II, AT sub 1A, has been genetically deleted; these mice do not develop hypertrophy when Ang II is given experimentally (confirming the basic success of the model in eliminating hypertrophy secondary to Ang II). However, when the <br><br> 4 <br><br> aorta is constricted in these animals (a model of hypertensive cardiac stress), the hearts still become hypertrophic. This suggests that alternative signaling pathways, not depending on this receptor (AT sub I A), are activated in hypertension. ACE inhibitors would presumably not be able to inhibit these pathways. See, Harada et al., Circulation. 97: 1952-1959 (1998). See also, Homey, Circulation. 97: 1890-1892 (1998) regarding the enigma associated with the process and mechanism of cardiac hypertrophy. <br><br> About 750,000 patients suffer from acute myocardial infarction (AMI) annually, and approximately one-fourth of all deaths in the United States are due to AMI. In recent years, thrombolytic agents, e.g., streptokinase, urokinase, and in particular tissue plasminogen activator (t-PA) have significantly increased the survival of patients who suffered myocardial infarction. When administered as a continuous intravenous infusion over 1.5 to 4 hours, t-PA produces coronary patency at 90 minutes in 69% to 90% of the treated patients. Topol et a!., Am. J. Cardiol.. 61. 723-728 (1988); Neuhaus et al., J. Am. Coll. Cardiol.. J2: 581-587(1988); Neuhaus et al., J. Am. Coll. Cardiol.. J4: 1566-1569 (1989). The highest patency rates have been reported with high dose or accelerated dosing regimens. Topol, J. Am. Coll. Cardiol.. 15: 922-924 (1990). t-PA may also be administered as a single bolus, although due to its relatively short half-life, it is better suited for infusion therapy. Tebbe et al., Am. J. Cardiol.. 64:448-453 (1989). A t-PA variant, specifically designed to have longer half-life and very high fibrin specificity, TNK t-PA (aT103N, NI17Q, KHRR(296-299)A AAA t-PA variant, Keyt etaL Proc. Natl. Acad. Sci. USA. 91: 3670-3674 (1994)) is particularly suitable for bolus administration. However, despite all these advances, the long-term prognosis of patient survival depends greatly on the post-infarction monitoring and treatment of the patients, which should include monitoring and treatment of cardiac hypertrophy. <br><br> B. Growth Factors <br><br> Various naturally occurring polypeptides reportedly induce the proliferation of endothelial cells. Among those polypeptides are the basic and acidic fibroblast growth factors (FGF) (Burgess and Maciag, Annual Rev. Biochem.. 58:575(1989)), platelet-derived endothelialcell growth factor (PD-ECGF) (Ishikawa et al., Nature. 338: 557(1989)), and vascular endothelial growth factor (VEGF). Leung eta!.. Science. 246: 1306(1989): Ferraraand Henzel, Biochem. Biophvs. Res. Commun.. 161: 851 (1989); Tischer et al, Biochem. Biophvs. Res. Commun.. 165: 1198(1989); EP47K754B granted July 31, 1996. <br><br> Media conditioned by cells transfected with the human VEGF (hVEGF) cDN A promoted the proliferation of capillary endothelial cells, whereas control cells did not. Leung et al., Science. 246: 1306 (1989). Several additional cDNAs were identified in human cDNA libraries that encode 121-, 189-. and 206-amino acid isoforms of hVEGF (also collectively referred to as hVEGF-related proteins). The 121-amino acid protein differs from hVEGF by virtue of the deletion of the 44 amino acids between residues 116 and 159 in hVEGF. The 189-amino acid protein differs from hVEGF by virtue of the insertion of 24 amino acids at residue 116 in hVEGF, and apparently is identical to human vascular permeability factor (hVPF). The 206-amino acid protein differs from hVEGF by virtue ofan insertion of41 amino acids at residue 116inhVEGF. Houck etal.. Mol. Endocrin.. 5:1806 (1991); Ferrara et al., J. Cell. Biochem.. 47: 211 (1991); Ferrara et al., Endocrine Reviews. 13: 18 (1992); Keck et al.. Science. 246: 1309 (1989); Connolly et al., J. Biol. Chem.. 264: 20017 (1989); EP 370,989 published May <br><br> 30, 1990. <br><br> It is now well established that angiogenesis, which involves the formation of new blood vessels from preexisting endothelium, is implicated in the pathogenesis of a variety of disorders. These include solid tumors and metastasis, atherosclerosis, retrolental fibroplasia, hemangiomas, chronic inflammation, intraocular neovascular syndromes such as proliferative retinopathies, e.g., diabetic retinopathy, age-related macular degeneration (AMD), neovascular glaucoma, immune rejection of transplanted corneal tissue and other tissues, rheumatoid arthritis, and psoriasis. Folkman ei at., J. Biol.Chem.. 267:10931 -10934 (1992); Klagsbrun et al, Annu. Rev. Phvsiol.. 53:217-239 (1991); and Garner A., "Vascular diseases", In: Pathobioloev of Ocular Disease. A Dynamic Approach. Garner A., Kiintworth GK, eds., 2nd Edition (Marcel Dekker, NY, 1994), pp 1625-1710. <br><br> In the case of tumor growth, angiogenesis appears to be crucial for the transition from hyperplasia to neoplasia, and for providing nourishment to the growing solid tumor. Folkman et al.. Nature. 339: 58(1989). The neovascularization allows the tumor cells to acquire a growth advantage and proliferative autonomy compared to the normal cells. Accordingly, a correlation has been observed between density of microvessels in tumor sections and patient survival in breast cancer as well as in several other tumors. Weidner et al., N. Engl. J. Med. 324: 1-6 (1991); Horak etal., Lancet. 340: 1120-1124 (1992); Macchiarini et al.. Lancet. 340: 145-146 (1992). <br><br> The search for positive regulators of angiogenesis has yielded many candidates, including aFGF, bFGF, TGF-a, TGF*p, HGF, TNF-a, angiogenin. 1L-8, etc. Folkman et al., J.B.C.. supra, and Klagsbrun et al., supra. The negative regulators so far identified include thrombospondin (Good et al., Proc. Natl. Acad. Sci. USA.. 87:6624-6628 (1990)), the 16-kilodalton N-terminal fragment of prolactin (Clapp et al.. Endocrinology. 133: 1292-1299 (1993)), angiostatin (O'Reilly et al.. Cell. 79:315-328 (1994)), and endostatin. O'Reilly et al., Cell. 88:277-285 (1996). <br><br> Work done over the last several years has established the key role of VEGF. not only in stimulating vascular endothelial cell proliferation, but also in inducing vascular permeability and angiogenesis. Ferrara et al., Endocr. Rev.. 18: 4-25 (1997). The finding that the loss of even a single VEGF allele results in embryonic lethality points to an irreplaceable role played by this factor in the development and differentiation of the vascular system. Furthermore, VEGF has been shown to be a key mediator of neovascularization associated with tumors and intraocular disorders. Ferrara et al.. Endocr. Rev., supra. The VEGF mRNA is overexpressed by the majority of human tumors examined. Berkman et al., J. Clin. Invest.. 91: 153-159(1993); Brown et al.. Human Pathol.. 26: 86-91 (1995); Brown et at.. Cancer Res.. 53:4727-4735 (1993); Mattem et al., Brit. J. Cancer. 73:931 -934 (1996); Dvorak et a/.. Am. J. Pathol.. 146: 1029-1039(1995). <br><br> Also, the concentration levels of VEGF in eye fluids are highly correlated to the presence of active proliferation of blood vessels in patients with diabetic and other ischemia-related retinopathies. Aiello etal., N. Engl. J. Med.. 331: 1480-1487 (199-1). Furthermore, recent studies have demonstrated the localization of VEGF in choroidal neovascular membranes in patients affected by AMD. Lopez et a!., Invest. Ophthalmol. Vis. Sci.. 37: 855-868(1996). <br><br> Anti-VEGF neutralizing antibodies suppress the growth of a variety of human tumor cell lines in nude mice (Kim et al, Nature. 362: 841-844 (1993); Warren etal, J. Clin. Invest.. 95: 1789-1797 (1995); Borgstrom etal., <br><br> Cancer Res.. 56: 4032-4039 (1996): Melnyk et al, Cancer Res.. 56: 921 -924 f1996)) and also inhibit intraocular angiogenesis in models of ischemic retinal disorders. Adamis et al.. Arch. Ophthalmol.. 114: 66-71 (1996). Therefore, anti-VEGF monoclonal antibodies or other inhibitors of VEGF action are promising candidates for the treatment of solid tumors and various intraocular neovascular disorders. Such antibodies are described, for example, in EP 817,648 published January 14, 1998 and in PCT/US 98/06724 filed April 3, 1998. <br><br> There exist several other growth factors and mitogens, including transforming oncogenes, that are capable of rapidly inducing a complex set of genes to be expressed by certain cells. Lau and Nathans, Molecular Aspects of Cellular Regulation. 6: 165-202 (1991). These genes, which have been named immediate-early- or early-response genes, are transcriptionally activated within minutes after contact with a growth factor or mitogen, independent of de novo protein synthesis. A group of these intermediate-early genes encodes secreted, extracellular proteins that are needed for coordination of complex biological processes such as differentiation and proliferation, regeneration, and wound healing. Ryseck et al., Cell Growth Differ.. 2: 235-233 (1991). <br><br> Highly-related proteins that belong to this group include cef 10 (Simmons et al.. Proc. Natl. Acad. Sci. USA. 86: 1178-1182 (1989)), cyr 61, which is rapidly activated by serum- or platelet-derived growth factor (PDGF) (O'Brien et al., Mol. Cell Biol.. 10: 3569-3577 (1990), human connective tissue growth factor (CTGF) (Bradham el al., J. Cell. Biol.. 114: 1285-1294 (1991)), which is secreted by human vascular endothelial cells in high levels after activation with transforming growth factor beta (TGF-P), exhibits PDGF-like biological and immunological activities, and competes with PDGF for a particular cell surface receptor, fisp-12 (Ryseck et al., Cell Growth Differ.. 2: 235-233 (1991)). human vascular IBP-like growth factor (V1GF) (WO 96/17931), and nov, normally arrested in adult kidney cells, which was found to be overexpressed in myeloblastosis-associated-virus-type-1 -induced nephroblastomas. Joloit etal.. Mol. Cell. Biol.. 12: 10-21 (1992). <br><br> The expression of these immediate-early genes acts as "third messengers" in the cascade of events triggered by growth factors. It is also thought that they are needed to integrate and coordinate complex biological processes, such as differentiation and wound healing in which cell proliferation is a common event. <br><br> As additional mitogens, insulin-like growth factor binding proteins (lGFBPs) have been shown, in complex with insulin-like growth factor (IGF), to stimulate increased binding of IGF to fibroblast and smooth muscle cell surface receptors. Clemmons et al.. J. Clin. Invest.. 77: 1548(1986). Inhibitory effects of 1GFBP on various IGF actions in vitro include stimulation of glucose transport by adipocytes, sulfate incorporation by chondrocytes, and thymidine incorporation in fibroblast. Zapf et al.. J. Clin. Invest., 63: 1077 (1979). In addition, inhibitory effects of IGFBPs on growth factor-mediated mitogen activity in normal cells have been shown. <br><br> C. Need for Further Treatments <br><br> In view of the role of vascular endothelial cell growth and angiogenesis in many diseases and disorders, it is desirable to have a means of reducing or inhibiting one or more of the biological effects causing these processes. It is also desirable to have a means of assaying for the presence of pathogenic polypeptides in normal and diseased conditions, and especially cancer. Further, in a specific aspect, as there is no generally applicable therapy for the treatment of cardiac hypertrophy, the identification of factors that can prevent or reduce cardiac myocyte <br><br> hypertrophy is of primary importance in the development of new therapeutic strategies to inhibit pathophysiological cardiac growth. While there are several treatment modalities for various cardiovascular and oncologic disorders, there is still a need for additional therapeutic approaches. <br><br> Summary of the Invention <br><br> A. <br><br> Embodiments <br><br> Accordingly, the present invention concerns compositions for promoting or inhibiting angiogenesis and/or cardiovascularization in mammals. Corresponding methods are described. <br><br> The present invention is based on the identification of proteins that test positive in various cardiovascular assays that test promotion or inhibition of certain biological activities. Accordingly, the proteins are believed to be useful drugs for the diagnosis and/or treatment (including prevention) of disorders where such effects are desired, such as the promotion or inhibition of angiogenesis, inhibition or stimulation of vascular endothelial cell growth, stimulation of growth or proliferation of vascular endothelial cells, inhibition of tumor growth, inhibition of angiogenesis-dependent tissue growth, stimulation of angiogenesis-dependent tissue growth, inhibition of cardiac hypertrophy and stimulation of cardiac hypertrophy, e.g., for the treatment of congestive heart failure. <br><br> In one embodiment, the present invention provides a pharmaceutical composition comprising a PRO 1313 polypeptide having at least 80% sequence identity to an amino acid shown in Figure 88 (SEQ ID NO: 216) and a pharmaceuticaliy acceptable carrier, diluent or excipient. In one aspect, <br><br> the composition comprises a therapeutically effective amount of the polypeptide. In another aspect, the composition comprises a further active ingredient, namely, a cardiovascular, endothelial or angiogenic agent or an angiostatic agent, preferably an angiogenic or angiostatic agent. Preferably, the composition is sterile. The PRO 1313 polypeptide may be administered in the form of a liquid pharmaceutical formulation, which may be preserved to achieve extended storage stability. Preserved liquid pharmaceutical formulations might contain multiple doses of PRO polypeptide, and might, therefore, be suitable for repeated use. <br><br> In a further embodiment, the present invention provides a method for preparing such a composition useful for the treatment of a cardiovascular, endothelial or angiogenic disorder comprising admixing a PRO 1313 polypeptide of the invention with a pharmaceuticaliy acceptable carrier. In one aspect, the polypeptide is present in a therapeutically effective amount. <br><br> Also described is a composition comprising an agonist or antagonist of the PRO 1313 polypeptide of the invention in admixture with a pharmaceuticaliy acceptable carrier. In one aspect, the composition <br><br> 8 <br><br> (followed by page 8a) <br><br> Intellectual property office of N.Z. <br><br> 1 5 JUN 2006 <br><br> 660135 l.DOC <br><br> comprises a therapeutically effective amount of the agonist or antagonist. In another aspect, the composition comprises a further active ingredient, namely, a cardiovascular, endothelial or angiogenic agent or an angiostatic agent, preferably an angiogenic or angiostatic agent. Preferably, the composition is sterile. The PRO polypeptide agonist or antagonist may be administered in the form of a liquid pharmaceutical formulation, which may be preserved lo achieve extended storage stability. Preserved liquid pharmaceutical formulations might contain multiple doses of a PRO polypeptide agonist or antagonist, and might, therefore, be suitable for repeated use. <br><br> In a further embodiment, the present invention provides a method for preparing such a composition useful for the treatment of a cardiovascular, endothelial or angiogenic disorder comprising admixing ^-therapeutically -effective amount of a PRO polypeptide agonist or antagonist with a pharmaceuticaliy acceptable carrier. <br><br> 8a <br><br> (followed by page 9) <br><br> intellectual property office of ni <br><br> 1 5 JUN 2006 <br><br> RECEIVED <br><br> 660135_1.DOC <br><br> In yet another embodiment, the present invention concerns a composition comprising an anti-PRO 1313 antibody in admixture with a pharmaceuticaliy acceptable carrier. In one aspect, the composition comprises a therapeutically effective amount of the antibody. In another aspect, the composition comprises a further active ingredient, namely, a cardiovascular, endothelial or angiogenic agent or an angiostatic agent, preferably an angiogenic or angiostatic agent. Preferably, the composition is sterile. The composition may be administered in the form of a liquid pharmaceutical formulation, which may be preserved to achieve extended storage stability. Preserved liquid pharmaceutical formulations might contain multiple doses of the anti-PRO antibody, and might, therefore, be suitable for repeated use. In preferred embodiments, the antibody is a monoclonal antibody, an antibody fragment, a humanized antibody, or a single-chain antibody. <br><br> In a further embodiment, the present invention provides a method for preparing such a composition useful for the treatment of a cardiovascular, endothelial or angiogenic disorder comprising admixing a therapeutically effective amount of an anti-PRO 1313 antibody with a pharmaceuticaliy acceptable carrier. <br><br> In a still further aspect, the present invention provides an article of manufacture comprising: <br><br> (1) a polypeptide having at least 80% sequence identity to an amino acid sequence shown in Figure 88 (SEQ ID NO: 216), or an antibody which specifically binds to said polypeptide in admixture with a pharmaceuticaliy acceptable carrier; <br><br> (2) a container containing said composition; and <br><br> (3) a label affixed to said container, or a package insert included in said container referring to the use of said composition in the treatment of a cardiovascular, endothelial, and angiogenic <br><br> The composition may comprise a therapeutically effective amount of the PRO 1313 polypeptide or the antibody thereto. Compositions with other PRO polypeptide agonists and antagonists are described but not claimed. <br><br> 1313 polypeptide of the invention comprising: <br><br> (a) contacting cells ex vivo and a test compound to be screened under conditions suitable for the induction of a cellular response normally induced by said polypeptide; and <br><br> (b) determining the induction of said cellular response to determine if the test compound is an effective agonist, vherein the induction of said cellular response is indicative of said test compound being an effective <br><br> 1313 polypeptide 'of the invention comprising: <br><br> (a) contacting cells ex vivo and a test compound to be screened under conditions suitable for the stimulation of cell proliferation by said polypeptide; and disorder. <br><br> In another embodiment, the present invention provides a method for identifying an agonist of a PRO <br><br> agonist. <br><br> In another embodiment, the present invention provides a method for identifying an agonist of a PRO <br><br> 9 <br><br> (followed by page 9a) <br><br> 660135 l.DOC <br><br> (b) measuring the proliferation of said ceils to determine if the test compound is an effective agonist, wherein the stimulation of cell proliferation is indicative of said test compound being an effective agonist. <br><br> In another embodiment, the invention provides a method for identifying a compoundthatinhibits the activity of a PRO 1313 polypeptide of the invention comprising contacting a test compound with said polypeptide ex vivo under conditions and for a time sufficient to allow the test compound and polypeptide to interact and determining whether the activity of the PRO polypeptide is inhibited. In a specific preferred aspect, either the test compound or the PRO polypeptide is <br><br> immobilized on. a solid support. In another preferred aspect, the noit-immobilized component carries a detectable label. In a preferred aspect,this method comprises the steps of: <br><br> (a) contacting cells ex vivo and a test compound to be screened in the presence of said polypeptide under conditions suitable for induction of a cellular response induced by: said polypeptide; and <br><br> (b) determining the induction of said cellular response to determine if the test compound is an effective <br><br> In another preferred aspect, this process comprises the steps of: <br><br> (a) contacting cells and a test compound to be screened in the presence of a PRO polypeptide under conditions suitable for the stimulation of cell proliferation by a PRO polypeptide; and <br><br> (b) measuring the proliferation of the cells to determine if the test compound is an effective antagonist. <br><br> In another embodiment, the invention provides a method for identifying a compound that inhibits the expression of a polypeptide of the invention in cells that normally expresses the polypeptide, wherein the method comprises contacting the cells ex vivo with a test compound under conditions suitable for allowing expression of said polypeptide and determining whether the expression of said polypeptide is inhibited. In a preferred aspect, the method comprises the steps of: <br><br> (a) contacting cells ex vivo and a test compound to be screened under conditions suitable for allowing expression of the PRO polypeptide; and <br><br> (b) determining the inhibition of expression of .said pplypepti,de. <br><br> Also described is a compound that inhibits the expression of a PRO polypeptide, such as a compound that is identified by the methods set forth above. <br><br> Also described is an agonist or an antagonist of a PRO polypeptide which may optionally be identified by the methods described above. <br><br> One type of antagonist of a PRO polypeptide that inhibits one or more of the functions or activities ofthe PRO polypeptide is an antibody. Hence, in another aspect, the invention provides an isolated antibody that binds a PRO <br><br> 1313 polypeptide of the invention. In a preferred aspect, the antibody is a monoclonal antibody, <br><br> which preferably has non-human compleme'ntarity-determining-region (CDR) residues and human framework-region (FR) residues. The antibody may be labeled and may be immobilized on a solid support. In a further aspect, the antibody is an antibody fragment, a single-chain antibody, or a humanized antibody. Preferably, the antibody specifically binds to the PRO 1313 polypeptide. <br><br> In a still further aspect, the present invention provides a method for diagnosing a disease or susceptibility to a disease which is related to a mutation in a nucleic acid sequence encoding a polypeptide of the invention comprising determining the presence or absence of said mutation in the PRO polypeptide nucleic acid sequence, wherein the presence or absence of said mutation is indicative of the presence of said disease or susceptibility to said disease. In a still further aspect, the invention provides a method of diagnosing a cardiovascular, endothelial or antagonist <br><br> (followed by page 1 Oa) <br><br> 10 <br><br> angiogenic disorder in a mammal which comprises analyzing the level of expression of a gene encoding a PRO 1313 polypeptide of the invention (a) in a test sample of tissue cells obtained from said mammal, and (b) in a control sample of known normal tissue cells of the same cell type, wherein a higher or lower expression level in the test sample as compared to the control sample is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in said <br><br> 10a <br><br> (followed by page 11) <br><br> | intellectual property <br><br> OFFICE <br><br> Of NX <br><br> 1 <br><br> 5 }UH 2006 <br><br> 660135 l.DOC. <br><br> RECEIVED <br><br> mammal. The expression of a gene encoding the PRO polypeptide may optionally be accomplished by measuring the level of mRNA or the polypeptide in the test sample as compared to the control sample. <br><br> In a still further aspect, the present invention provides a method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal which comprises detecting the presence or absence of a PRO 1313 polypeptide of the invention in a test sample of tissue cells obtained from said mammal, wherein the presence or absence of said PRO polypeptide in said test sample is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in said mammal. <br><br> In a still fUrther embodiment, the invention provides a method of diagnosing £ cardiovascular, endothelial or angiogenic disorder in a mammal comprising (a) contacting an antibody to a polypeptide of the invention with a test sample of tissue cells obtained from the mammal, and (b) detecting the formation of a complex between the antibody and the PRO polypeptide in the test sample, wherein the formation of said complex is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in the mammal. The detection may be qualitative or quantitative, and may be performed in comparison with monitoring the complex formation in a control sample of known normal tissue cells of the same cell type. A larger or smaller quantity of complexes formed in the test sample indicates the presence of a cardiovascular, endothelial or angiogenic dysfunction in the mammal from which the test tissue cells were obtained. The antibody preferably carries a detectable label. Complex formation can be monitored, for example, by light microscopy, flow cytometry, fluorimetry, or other techniques known in the art. The test sample is usually obtained from an individual suspected to have a cardiovascular, endothelial or angiogenic disorder. <br><br> In another embodiment, the invention provides a method for determining the presence of a PRO polypeptide of the invention in a sample comprising exposing a sample suspected of containing the <br><br> PRO polypeptide with an antibody thereto and detenn ining binding of said antibody to a component of said sample. In a specific aspect, the sample comprises a cell suspected of containing the PRO polypeptide and the antibody binds to the cell. The antibody is preferably detectably labeled and/or bound to a solid support <br><br> In further aspects, the inventior provides acardiovascular, endothelial or angiogenic disorder diagnostic kit comprising an antibody to a polypeptide of the invention and a carrier in suitable packaging. <br><br> Preferably, such kit further comprises instructions for using said antibody to detect the presence of the PRO polypeptide. Preferably, the carrier is a buffer, for example. Preferably, the cardiovascular, endothelial or angiogenic disorder4s cancer. <br><br> Described but not claimed is a method for treating a cardiovascular, endothelial <br><br> 11 <br><br> (followed by page 11a) <br><br> intellectual property office of n.z. <br><br> 1 5 J \M 2006 <br><br> 660135 J.DOC <br><br> or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of a PRO polypeptide. Preferably, the disorder is cardiac hypertrophy, trauma such as wounds or bums, or a type of cancer. In a further aspect, the mammal is further exposed to angioplasty or a drug that treats cardiovascular, endothelial or angiogenic disorders such as ACE inhibitors or chemotherapeutic agents if the cardiovascular, endothelial or angiogenic disorder is a type of cancer. Preferably, the mammal is human, preferably one who is at risk of developing cardiac hypertrophy and more preferably has suffered myocardial infarction. <br><br> In another preferred aspect, the cardiac hypertrophy is characterized by the presence of an elevated level of PGF2((. Alternatively, the cardiac hypertrophy may be induced by myocardial infarction, wherein preferably the administration of the PRO polypeptide is initiated within 48 hours, more preferably within 24 hours, following <br><br> 11a <br><br> (followed by page 12) <br><br> 660135 l.DOC <br><br> intellectual property office of n.z. <br><br> 1 5 JUM 2006 RECEIVED <br><br> WO 00/32221 <br><br> PCT7US99y28313 - <br><br> myocardial infarction. <br><br> In anotherpreferredembodimenuhe cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy and said PRO polypeptide is administered together with a cardiovascular, endothelial or angiogenic agent. The preferred cardiovascular, endothelial or angiogenic agent for this purpose is selected from the group consisting of an antihypertensive drug, an ACE inhibitor, an endothelin receptor antagonist and a thrombolytic agent. If a thrombolytic agent is administered, preferably the PRO polypeptide is administered following administration of such agent. More preferably, the thrombolytic agent is recombinant human tissue plasminogen activator. <br><br> In another preferred aspect, the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy and the PRO polypeptide is administered following primary angioplasty for the treatment of acute myocardial infarction, preferably wherein the mammal is further exposed to angioplasty or a cardiovascular, endothelial, or angiogenic agent. <br><br> In another preferred embodiment, the cardiovascular, endothelial or angiogenic disorder is a cancer and the PRO polypeptide is administered in combination with a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent. <br><br> Also described is ' a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of an agonist of a PRO polypeptide. Preferably, the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy, <br><br> trauma, a cancer, or age-related macular degeneration. Also preferred is where the mammal is human, and where an effective amount of an angiogenic or angiostatic agent is administered in conjunction with the agonist. <br><br> Also described is | a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of an antagonist of a PRO polypeptide. Preferably, the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy, <br><br> trauma, a cancer, or age-related macular degeneration. Also preferred is where the mammal is human, and where an effective amount of an angiogenic or angiostatic agent is administered in conjunction with the antagonist. <br><br> Also described is ■ a method for treating a cardiovascular, endothelial or angiogenic disorder in a mammal comprising administering to the mammal an effective amount of an anti-PRO antibody. Preferably, the cardiovascular, endothelial or angiogenic disorder is cardiac hypertrophy, trauma, a cancer, or age-related macular degeneration. Also preferred is where the mammal is human, and where an effective amount of an angiogenic or angiostatic agent is administered in conjunction with the antibody. <br><br> Also described is a method for treating a cardiovascular, endothelial or f <br><br> angiogenic disorder in a mammal that suffers therefrom comprising administering to the mammal a nucleic acid molecule that codes for either (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide or (c) an antagonist of a PRO polypeptide, wherein said agonist or antagonist may be an anti-PRO antibody. In a preferred embodiment, the mammal is human. In another preferred embodiment, the gene is administered via ex vivo gene therapy. In a further preferred embodiment, the gene is comprised within a vector, more preferably an adenoviral, adeno-associated viral, lentiviral, or retroviral vector. <br><br> In yet another aspect, the invention provides. <br><br> [intellectual property office <br><br> 12 0h N'Z' <br><br> 1 5 m 2006 <br><br> RECEIVED <br><br> 10 <br><br> WO 00/322 PCT/US99/28313 - <br><br> a recombinant retroviral particle comprising a retroviral vector consisting essentially of (1) a promoter, (2) nucleic acid encoding a polypeptide of the invention, an antibody thereto, and (3) a signal sequence for cellular secretion of the polypeptide, wherein the retroviral vector is in association with retroviral structure proteins. Preferably, the signal sequence is from a mammal, such as from native PRO polypeptide. <br><br> In a still further embodiment, the invention supplies an ex vivo producer cell comprising a nucleic acid construct that expresses retroviral structural proteins and also comprises a retroviral vector consisting essentially of (1) a promoter, (2) nucleic acid encoding a polypeptide of the invention, or an antibody thereto, and (3) a signal sequence for cellular secretion of the polypeptide, wherein said producer cell packages the retroviral vector in association with the structural proteins to produce recombinant retroviral particles. <br><br> Also described is a method for inhibiting endothelial cell growth in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, <br><br> or (c) an antagonist of a PRO polypeptide, wherein endothelial cell growth in said mammal is inhibited, and wherein said agonist or antagonist may be an anti-PRO antibody. Preferably, the mammal is human and the 15 endothelial cell growth is associated with a tumor or a retinal disorder. <br><br> Also described is a method for stimulating endothelial cell growth in a mammal comprising administering to the mammal (4) a PRO polypeptide, (b) an agonist of a PRO polypeptide, <br><br> or (c) an antagonist of a PRO polypeptide, wherein endothelial cell growth in said mammal is stimulated, and wherein said agonist or antagonist may be an anti-PRO antibody. Preferably, the mammal is human. 20 Also described is a method for inhibiting cardiac hypertrophy in a mammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or (c) an antagonist of a PRO polypeptide, wherein cardiac hypertrophy in said mammal is inhibited, and wherein said agonist or antagonist may be an anti-PRO antibody. Preferably, the mammal is human and the cardiac hypertrophy has been induced by myocardial infarction. <br><br> 25 Also described is amethodforstimulatingcardiachypertrophyinamammal comprising administering to the mammal (a) a PRO polypeptide, (b) an agonist of a PRO polypeptide, or (c) an antagonist of a PRO polypeptide, wherein cardiac hypertrophy in said mammal is stimulated, and wherein said agonist or antagonist may be an anti-PRO antibody. Preferably, the mammal is human who suffers from congestive heart failure. <br><br> 30 Also described is a method for inhibiting angiogenesis induced by a PRO <br><br> polypeptide in a mammal comprising administering a therapeutically effective amount of an anti-PRO antibody to the mammal. Preferably, the mammal is a human, and more preferably the mammal has a tumor or a retinal disorder. <br><br> Also described is ja method for stimulating angiogenesis induced by a PRO <br><br> 35 polypeptide in a mammal comprising administering a therapeutically effective amount of a PRO polypeptide to the mammal. Preferably, the mammal is a human, and more preferably angiogeneisis would promote tissue regeneration or wound healirtg. intellectual property office of !\l2. <br><br> 13 15 JUN 2006 <br><br> (followed by page 13a) _ <br><br> RECEIVED <br><br> The invention also provides an oligonucleotide anti-sense to a nucleic acid molecule encoding a polypeptide of the invention, which inhibits the expression of said polypeptide in a mammalian cell which expresses said polypeptide. <br><br> The invention also relates to a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide, in the manufacture of a medicament for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> In a further aspect, the invention relates to a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide; and a cardiovascular, endothelial or angiogenic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> Also provided by the invention is a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide, and a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> In a further aspect, the invention relates to a use of a nucleic acid molecule that encodes a polypeptide of the invention or an antibody thereto in the manufacture of a medicament for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> The invention also relates to a use of a polypeptide of the invention or an antibody thereto, in the manufacture of a medicament for inhibiting endothelial cell growth in a mammal. <br><br> The invention further relates to a use of an antibody which specifically binds a polypeptide of the invention, in the manufacture of a medicament for stimulating endothelial cell growth in a mammal. <br><br> 13a <br><br> (followed by page 14) <br><br> intellectual property office of n.z. <br><br> 1 5 JUN 2006 <br><br> 660135 l.DOC <br><br> B. <br><br> Additional Embodiments <br><br> In other embodiments of the present invention, the invention provides an isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence that encodes a polypeptide comprising an amino acid sequence shown in Figure 88 (SEQ ID NO:216). <br><br> In one aspect, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80% <br><br> sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at-least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91 % sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA molecule encoding a PRO 1313 polypeptide having a full-length amino acid sequence as disclosed herein, an amino acid sequence lacking the signal peptide as disclosed herein, an extracellu lar domain of a transmembrane protein, with or without the signal peptide, as disclosed herein or any other specifically defined fragment of the full-length amino acid sequence as disclosed herein, or (b) the complement of the DNA molecule of (a). <br><br> In another aspect, the invention provides an isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence shown in Figure 87 (SEQ ID NO:215). <br><br> The invention also provides an isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence comprising the full length coding sequence of the nucleotide sequence shown in Figure 87 (SEQ ID NO:215). <br><br> In other aspects, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91 % sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA molecule comprising the coding <br><br> 14 <br><br> (followed by page 14a) <br><br> 660135 l.DOC <br><br> sequence of a full-length PRO 1313 polypeptide cDNA as disclosed herein, the coding sequence of the PRO polypeptide lacking the signal peptide as disclosed herein, the coding sequence of an extracellular domain of a transmembrane PRO polypeptide, with or without the sign&amp;l peptide, as disclosed herein or the coding sequence of any other \specifically defined fragment of the full-length amino acid sequence as disclosed herein, or (b) the complement of the DNA molecule of (a). <br><br> The invention also provides an isolated nucleic acid having at least 80% nucleic acid sequence identity to the full-length coding sequence of the DNA deposited under ATCC accession number 203575. <br><br> In a further aspect, the invention concerns an isolated nucleic acid molecule comprising a nucleotide sequence haying at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at lea^t about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably <br><br> 14a <br><br> (followed by page 15) <br><br> 660135_1.DOC <br><br> intellectual PROPERTY OFFICE Or Mi <br><br> 1 5 JIM 2006 <br><br> i RECEIVED I <br><br> at least about 84% sequence Identity', yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA <br><br> molecule that encodes the same mature polypeptide encoded by any of the human protein 1313 cDNAs deposited with the ATCC under accession number ATCC 203575 as disclosed herein, or (b) the complement of the DNA molecule of (a). <br><br> The invention also provides an isolated nucleic acid having a nucleotide sequence that encodes a polypeptide comprising the amino acid sequence shown in Figure 88 (SEQ ID NO: 216). <br><br> Another aspect the invention provides^an isolated nucleic acid molecule comprising a nucleotide sequence encoding a PRO 1313 polypeptide which is either transmembrane domain-deleted or transmembrane domain-inactivated, <br><br> or is complementary to such encoding nucleotide sequence, wherein the transmembrane domain(s) of such polypeptide are disclosed herein. Therefore, soluble extracellular domains of the herein described PRO polypeptides are contemplated. <br><br> The invention also provides an isolated nucleic acid molecule consisting of an at least 20 nucleotides fragment of the nucleic acid sequence of SEQ ID NO:215, or a complement thereof, that hybridizes under stringent conditions to: <br><br> (a) the nucleic acid sequence of SEQ ID NO:215 or a complement thereof; <br><br> (b) the full length coding sequence of the cDNA deposited under ATCC accession number 203575 or a complement thereof; and <br><br> (c) wherein, said stringent conditions use 50% formamide, 5 x SSC, 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5x Denhardt's solution, <br><br> sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC and 50% formamide at 55°C, followed by a wash comprising of 0.1 x SSC containing EDTA at 55°C. <br><br> Another embodiment is directed to fragments of a PRO 1313 polypeptide coding sequence, or the complement <br><br> 660135_I.DOC <br><br> thereof, that may find use as, for example, hybridization probes, for encoding fragments of a PRO polypeptide that may optionally encode a polypeptide comprising a binding site for an anti-PRO antibody or as antisense oligonucleotide probes. Such nucleic acid fragments are usually at least about 20 nucleotides in length, preferably at least about 30 nucleotides in length, more preferably at least about 40 nucleotides in length, yet more preferably at least about SO nucleotides in length, yet more preferably at least about 60 nucleotides in length, yet more preferably at least about 70 nucleotides in length, yet more preferably at least about 80 nucleotides in length, yet more preferably at least about 90 nucleotides in length, yet more preferably at least about 100 nucleotides in length, yet more preferably at least about 110 nucleotides in length, yet more preferably at least about 120 nucleotides in length, yet more preferably at least about 130 nucleotides in length, yet more preferably at least about 140 nucleotides in length, yet more preferably at least about 150 nucleotides in length, yet more preferably at least about 160 nucleotides in length, yet more preferably at least about 170 nucleotides in length, yet more preferably at least about 180 nucleotides in length, yet more preferably at least about 190 nucleotides in length, yet more preferably at least about 200 nucleotides in length, yet more preferably at least about 250 nucleotides in length, yet more preferably at least about 300 nucleotides in length, yet more preferably at least about 350 nucleotides in length, yet more preferably at least about 400 nucleotides in length, yet more preferably at least about 450 nucleotides in length, yet more preferably at least about '500 nucleotides in length, yet more preferably at least about 600 nucleotides in length, yet more preferably at least about 700 nucleotides in length, yet more preferably at least about 800 nucleotides in length, yet more preferably at least about 900 nucleotides in length and yet more preferably at least about 1000 nucleotides in length, wherein in this context the term "about" means the referenced nucleotide sequence length plus or minus 10% of that referenced length. It is noted that novel fragments of a PRO polypeptide-encoding nucleotide sequence may be determined in a routine manner by aligning the PRO <br><br> 15a <br><br> (followed by page 16) <br><br> intellectual property office OF N.2. <br><br> 1 5 JUH 2008 <br><br> polypeptide-encoding nucleotide sequence with other known nucleotide Sequences using any of a number of well Known sequence alignment programs and determining which PRO polypeptide-encoding nucleotide sequence fragments) are novel. All of such PRO polypeptide-encoding nucleotide sequences are contemplated herein. Also contemplated are the PRO polypeptide fragments encoded by these nucleotidemolecule fragments, preferably those <br><br> PRO 1313 polypeptide fragments that comprise a binding site of an anti-PRO antibody <br><br> The invention also provides isolated nucleic acid having at least 80% nucleic acid sequence identity to: <br><br> (a) a nucleotide sequence encoding the polypeptide shown in Figure 88 (SEQ ID NO:216) lacking its associated signal peptide; <br><br> (b) a nucleotide sequence encoding an extraceullalar domain of the polypeptide shown in Figure 88 (SEQ ID NO:216), with its associated signal peptide; or <br><br> (c) a nucleotide sequence encoding an extraceullular domain of the polypeptide shown in Figure 88 (SEQ ID NO:216) lacking its associated signal peptide. <br><br> In another embodiment, the invention provides an isolated polypeptide having at least 80% amino acid sequence identity to an amino acid sequence shown in Figure 88 (SEQ ID NO:216). <br><br> In a certain aspect, the invention concerns an isolated PRO 1313 polypeptide, comprising an amino acid sequence having at least about 80% sequence identity, preferably at lea$t about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to a PRO 1313 polypeptide having a full-length amino acid sequence as disclosed herein, an amino acid sequence lacking the signal peptide as disclosed herein, an extracellular domain of a transmembrane protein, with or without the signal peptide, as disclosed herein or any other specifically defined fragment of the f\i 1 l-Iength amino ac jdjsequence as disclosed herein. <br><br> In one aspect, the invention provides an isolated polypeptide comprising a sequence having the amino acid sequence shown in figure 88 (SEQ ID NO: 216). <br><br> The invention also provides an isolated polypeptide having at least 80% amino acid sequence identity to an amino acid sequence encoded by the full-length coding sequence of the DNA deposited under ATCC accession number 203575. <br><br> 16 <br><br> (followed by page 16a) | INTELLECTUAL <br><br> ^ 5 JUH 2G06 <br><br> 660135 l.DOC <br><br> In a further aspect, the invention concerns an isolated PRO polypeptide comprising an amino acid sequence having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to an amino acid sequence encoded by any of the human protein 1313 cDNAs deposited with the ATCC as disclosed <br><br> In one aspect, the invention provides an isolated polypeptide comprising a sequence having the amino acid sequence encoded by the nucleotide deposited under ATCC accession number <br><br> In a further aspect, the invention provides an isolated polypeptide scoring at least 80% positives when compared to an amino acid sequence consisting of the amino acid sequence shown in Figure 88 (SEQ ID NO:216). <br><br> scoring at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82% positives, yet more preferably at least about 83% positives, yet more preferably at least about 84% positives, yet herein. <br><br> 203575. <br><br> In a funher aspect, the invention concerns an isolated PRO polypeptide comprising an amino acid sequence <br><br> 16a <br><br> (followed by page 17) <br><br> 660135_1.DOC <br><br> more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89% positives, yet more preferably at least about 90% positives, yet more preferably at least about 91 % positives, yet more preferably at least about 92% positives, yet more preferably at least about 93% positives, yet more preferably at least about 94% positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives and yet more preferably at least about 99% positives when compared with the amino acid sequence of a PRO 1313 polypeptide having a full-length amino acid sequence as disclosed herein, an amino acid sequence lacking the signal peptide as disclosed herein, an extracellular domain of a transmembrane protein, with or without the signal peptide, as disclosed herein or any other specifically defined fragment of the full-length amino acid sequence as disclosed herein. <br><br> In a specific aspect, the invention provides an isolated PRO 1313 polypeptide without the sequence as hereinbefore described. More particularly, the invention provides an isolated polypeptide having at least 80% amino acid sequence identity to: <br><br> (a) the polypeptide shown in <br><br> Figure 88 (SEQ ID NO:216) lacking its associated signal peptide; <br><br> (b) an extracellular domain of the polypeptide shown in Figure 88 (SEQ ID NO:216) with its associated signal peptide; or <br><br> (c) an extraceullular domain of the polypeptide shown in <br><br> Figure 88 (SEQ ID NO:216) lacking its associated signal peptide. <br><br> Process for producing the same are also herein described, wherein those processes comprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression of the PRO polypeptide and recovering the PRO polypeptide from the cell culture. <br><br> Also described is an isolated PRO 1313 polypeptide which is either transmembrane domain-deleted or transmembrane domain-inactivated. Processes for producing the same are also herein described, wherein those processes comprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression of the PRO polypeptide and recovering the PRO polypeptide from the cell culture. v <br><br> In yet another embodiment, the invention concerns agonists and antagonists of a native PRO 1313 polypeptide as <br><br> N-terminal signal sequence and/or th&lt; <br><br> »d/w &lt;he initiating methionine and is -co"" *'»1nuclebtide sequencefarencode,:such &lt;anaminoacid <br><br> 660135 l.DOC <br><br> 17 <br><br> (followed by page 17a) <br><br> defined herein. In a particular embodiment, the agonist or antagonist is an anti-PRO antibody or a small molecule. <br><br> In a further embodiment, the invention concerns a method of identifying agonists or antagonists to a PRO <br><br> 1313 polypeptide which comprise contacting the PRO polypeptide with a candidate molecule and monitoring a biological activity mediated by said PRO polypeptide. Preferably, the PRO 1313 polypeptide is a native PRO 1313 polypeptide. <br><br> More particularly, the invention provides a method for identifying an agonist of a polypeptide ■ of the invention comprising: <br><br> (a) contacting cells ex vivo and a test compound to be screened under conditions suitable for the induction of a cellular response normally induced by said polypeptide; and <br><br> (b) determining the induction of said cellular response to determine if the test compound is an effective agonist, wherein the induction of said cellular response is indicative of said test compound being an effective agonist. <br><br> Also provided is a method for identifying a compound that inhibits an activity of a polypeptide as defined in the invention comprising contacting a test compound with said polypeptide ex vivo under conditions and for a time sufficient to allow the test compound and polypeptide to interact and determining whether the activity of said polypeptide is inhibited. <br><br> The invention also provides a method for identifying a compound that inhibits an activity of a polypeptide as defined in the invention comprising the steps of: <br><br> (a) contacting cells ex vivo and a test compound to be screened in the presence of said polypeptide under conditions suitable for the induction of a cellular response normally induced by said polypeptide; and <br><br> (b) determining the induction of said cellular response to determine if the test compound is an effective antagonist. <br><br> Also provided is a method for identifying a compound that inhibits the expression of a polypeptide as defined in the invention in cells that normally expresses the polypeptide, wherein the method comprises contacting the cells ex vivo with a test compound under conditions suitable for allowing expression of said polypeptide and determining whether the expression of said polypeptide is inhibited. <br><br> In a still further embodiment, the invention concerns a composition of matter comprising a PRO 1313 polypeptide, or an agonist or antagonist of a PRO 1313 polypeptide as herein described, or an anti-PRO 1313 antibody, in combination with a carrier. Optionally, the carrier is a pharmaceuticaliy acceptable carrier. <br><br> Also described herein is the use of a PRO polypeptide, or an agonist or antagonist thereof as hereinbefore described, or an anti-PRO antibody, for the preparation of a medicament useful in the treatment of a condition which is responsive to the PRO polypeptide, an agonist or antagonist thereof or an anti-PRO <br><br> antibody. <br><br> 17a <br><br> 660135 l.DOC <br><br> (followed by page 17b) <br><br> More particularly, the invention provides a pharmaceutical composition comprising a polypeptide of the invention and a pharmaceuticaliy acceptable carrier, excipient or stabiliser. <br><br> Also provided is a composition comprising an antibody according to the invention in admixture with a pharmaceuticaliy acceptable carrier. <br><br> The invention provides a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide, in the manufacture of a medicament for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> The invention also relates to a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide; and a cardiovascular, endothelial or angiogenic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> Further, the invention relates to a use of a polypeptide of the invention or an antibody which specifically binds to said polypeptide, and a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal. <br><br> The invention also relates to a use of a nucleic acid molecule that encodes a polypeptide of the invention or an antibody thereto in the manufacture of a medicament for treating a cardiovascular, <br><br> endothelial or angiogenic disorder in a mammal. <br><br> In a further embodiment the invention provides a use of a polypeptide of the invention or an antibody thereto, in the manufacture of a medicament for inhibiting endothelial cell growth in a mammal. <br><br> In a still further embodiment, the invention provides a use of an antibody which specifically binds a polypeptide of the invention in the manufacture of a medicament for stimulating endothelial cell growth in a mammal. <br><br> In additional embodiments of the present invention, the invention provides vectors comprising DNA encoding the polypeptides of the invention. Host cells comprising any such vector are also provided. By way of example, the host cells may be CHO cells. E. coli, yeast, or Baculovirus-infected insect cells. A process for <br><br> 660135 l.DOC <br><br> producing any of the herein described polypeptides is further provided and comprises culturing host cells under conditions suitable for expression of the polypeptide and recovering the polypeptide from the cell culture. <br><br> In other embodiments, the invention provides chimeric molecules comprising any of the herein described 5 polypeptides fused to a heterologous polypeptide or amino acid sequence. Example of such chimeric molecules comprise any of the herein described polypeptides fused to an epitope tag sequence or a Fc region of an immunoglobulin. <br><br> In yet another embodiment, the invention provides an antibody which specifically binds to; &lt; the polypeptide of the invention. Optionally, the antibody is a monoclonal antibody, <br><br> humanized antibody, antibody fragment or single-chain antibody. <br><br> In yet other embodiments, the invention provides oligonucleotide probes useful for isolating genomic and cDNA nucleotide sequences or as antisense probes, wherein those probes may be derived from any of the above or below described nucleotide sequences. <br><br> 15 Brief Description of the Drawings <br><br> Figure 1 shows a nucleotide sequence (SEQ ID NO:3) of a native sequence PR0172 cDNA, wherein SEQ IDNO:3 isaclone designated herein as "DNA35916-1161". <br><br> Figure 2 shows the amino acid sequence (SEQ ID NO:4) derived from the coding sequence of SEQ IDNO:3 shown in Figure 1. <br><br> 20 Figure 3 shows a nucleotide sequence (SEQ ID NO:8) of a native sequence PRO 178 cDNA, wherein SEQ <br><br> ID NO:8 is a clone designated herein as "DNA23339-1130". <br><br> Figure 4 shows the amino acid sequence (SEQ ID NO:9) derived from the coding sequence of SEQ ID NO:8 shown in Figure 3. <br><br> Figure 5 shows a nucleotide sequence (SEQ ID NO: 13) of a native sequence PROl 79 cDNA, wherein SEQ <br><br> 25 IDNO:13 is a clone designated herein as "DNA16451-1388". <br><br> Figure 6 shows the amino acid sequence (SEQ ID NO: 14) derived from the coding sequence of SEQ ID NO:13 shown in Figure 5. <br><br> Figure 7 shows a nucleotide sequence (SEQ ID NO: 15) of a native sequence PROl 82 cDNA, wherein SEQ ID NO: 15 is a clone designated herein as "DNA27865-1091". <br><br> 30 Figure 8 shows the amino acid sequence (SEQ ID NO: 16) derived from the coding sequence of SEQ ID <br><br> NO: 15 shown in Figure 7. <br><br> Figure 9 shows a nucleotide sequence (SEQ ID N0:20) of a native sequence PRO 187 cDN A, wherein SEQ ID N0:20 is a clone designated herein as "DNA27864-1155". <br><br> Figure 10 shows the amino acid sequence (SEQ ID NO:21) derived from the coding sequence of SEQ ID <br><br> 35 N0:20 shown in Figure 9. <br><br> Figure 11 shows a nucleotide sequence (SEQ ID NO:25)ofa native sequence PRO 188 cDNA, wherein SEQ <br><br> ID NO:25 is a clone designated herein as "DNA28497-1130". <br><br> rr^tci i egtuj <br><br> Of <br><br> 18 <br><br> 1 5 jUH ® <br><br> Figure 12 shows the amino acid sequence (SEQ ID NO:26) derived from the coding sequence of SEQ ID NO:25 shown in Figure 11. <br><br> Figure 13 shows a nucleotide sequence (SEQ ID N0:30) of a native sequence PRO 195 cDN A, wherein SEQ ID N0:30 is a clone designated herein as "DNA26847-1395". <br><br> Figure 14 shows the amino acid sequence (SEQ ID NO:31) derived from the coding sequence of SEQ ID N0:30 shown in Figure 13. <br><br> Figure 15 shows a nucleotide sequence (SEQ ID NO:35) of a native sequence PR0212 cDNA, wherein SEQ ID NO:35 is a clone designated herein as "DNA30942-1134". <br><br> Figure 16 shows the amino acid sequence (SEQ ID NO:36) derived from the coding sequence of SEQ ID NO:35 shown in Figure 15. <br><br> Figure 17 shows a nucleotide sequence (SEQ ID N0:40) of a native sequence PR0214 cDNA, wherein SEQ ID N0:40 is a clone designated herein as "DNA32286-1191". <br><br> Figure 18 shows the amino acid sequence (SEQ ID NO:41) derived from the coding sequence of SEQ ID N0:40 shown in Figure 17. <br><br> Figure 19 shows a nucleotide sequence (SEQ ID NO:45) of a native sequence PR0217 cDNA, wherein SEQ ID NO:45 is a clone designated herein as "DNA33094-1131 <br><br> Figure 20 shows the amino acid sequence (SEQ ID NO:46) derived from the coding sequence of SEQ ID NO:45 shown in Figure 19. <br><br> Figure 21 shows a nucleotide sequence (SEQ ID N0:50) of a native sequence PR0224 cDNA, wherein SEQ ID N0:50 is a clone designated herein as "DNA33221-1133". <br><br> Figure 22 shows the amino acid sequence (SEQ ID NO:5l) derived from the coding sequence of SEQ ID N0:50 shown in Figure 21. <br><br> Figure 23 shows a nucleotide sequence (SEQ ID NO:55) of a native sequence PR0231 cDNA, wherein SEQ ID NO:55 is a clone designated herein as "DNA34434-1139". <br><br> Figure 24 shows the amino acid sequence (SEQ ID NO:56) derived from the coding sequence of SEQ ID NO:55 shown in Figure 23. <br><br> Figure 25 shows a nucleotide sequence (SEQ ID NO:61) of a native sequence PR0235 cDNA, wherein SEQ ID NO:61 is a clone designated herein as "DNA35558-1167". <br><br> Figure 26 shows the amino acid sequence (SEQ ID NO:62) derived from the coding sequence of SEQ ID NO:61 shown in Figure 25. <br><br> Figure 27 shows a nucleotide sequence (SEQ ID NO:66) of a native sequence PR0245 cDNA, wherein SEQ IDNO:66 is a clone designated herein as "DNA35638-1141". <br><br> Figure 28 shows the amino acid sequence (SEQ ID NO:67) derived from the coding sequence of SEQ ID N0.66 shown in Figure 27. <br><br> Figure 29 shows a nucleotide sequence (SEQ ID NO:71) of a native sequence PR0261 cDNA, wherein SEQ ID NO:71 is a clone designated herein as "DNA33473-1176". <br><br> Figure 30 shows the amino acid sequence (SEQ ID NO:72) derived from the coding sequence of SEQ ID <br><br> N0:71 shown in Figure 29. <br><br> Figure 31 shows a nucleotide sequence (SEQ ID NO:76) of a native sequence PR0269 cDNA, wherein SEQ ID NO:76 is a clone designated herein as "DNA38260-1180". <br><br> Figure 32 shows the amino acid sequence (SEQ ID NO:77) derived from the coding sequence of SEQ ID NO:76 shown in Figure 31. <br><br> Figure 33 shows a nucleotide sequence (SEQ ID NO:84) of a native sequence PR0287 cDNA, wherein SEQ ID NO:84 is a clone designated herein as "DNA39969-1185". <br><br> Figure 34 shows the amino acid sequence (SEQ ID NO:85) derived from the coding sequence of SEQ ID NO:84 shown in Figure 33. <br><br> Figure 35 shows a nucleotide sequence (SEQ ID NO:89) of a native sequence PR0301 cDNA, wherein SEQ ID NO:89 is a clone designated herein as "DNA40628-1216". <br><br> Figure 36 shows the amino acid sequence (SEQ ID N0:90) derived from the coding sequence of SEQ ID NO:89 shown in Figure 35. <br><br> Figure 37 shows a nucleotide sequence (SEQ ID NO:97) of a native sequence PR0323 cDNA, wherein SEQ ID NO:97 is a clone designated herein as "DNA35595-1228". <br><br> Figure 38 shows the amino acid sequence (SEQ ID NO:98) derived from the coding sequence of SEQ ID NO:97 shown in Figure 37. <br><br> Figure 39 shows a nucleotide sequence (SEQ ID NO: 106) of a native sequence PR0331 cDNA, wherein SEQ IDNO:106 is a clone designated herein as "DNA40981-1234". <br><br> Figure 40 shows the amino acid sequence (SEQ ID NO: 107) derived from the coding sequence of SEQ ID NO: 106 shown in Figure 39. <br><br> Figure41 shows a nucleotide sequence (SEQ ID NO: 111) of a native sequence PR03 56 cDN A, wherein SEQ ID NO:l 11 is a clone designated herein as "DNA47470-1130-P1". <br><br> Figure 42 shows the amino acid sequence (SEQ ID NO:l 12) derived from the coding sequence of SEQ ID NO: 111 shown in Figure 41. <br><br> Figure 43 shows a nucleotide sequence (SEQ ID NO: 116) of a native sequence PR0364 cDNA, wherein SEQ ID NO:l 16 is a clone designated herein as "DNA47365-1206". <br><br> Figure 44 shows the amino acid sequence (SEQ ID NO: 117) derived from the coding sequence of SEQ ID NO: 116 shown in Figure 43. <br><br> Figure 45 shows a nucleotide sequence (SEQ ID NO: 126) of a native sequence PR0526 cDN A, wherein SEQ ID NO: 126 is a clone designated herein as "DNA44184-I319". <br><br> Figure 46 shows the amino acid sequence (SEQ ID NO: 127) derived from the coding sequence of SEQ ID NO: 126 shown in Figure 45. <br><br> Figure 47 shows a nucleotide sequence (SEQ ID NO: 131) of a native sequence PR0538 cDN A, wherein SEQ IDNO:131 is a clone designated herein as "DNA48613-1268". <br><br> Figure 48 shows the amino acid sequence (SEQ ID NO: 132) derived from the coding sequence of SEQ ID NO: 131 shown in Figure 47. <br><br> Figure 49 shows a nucleotide sequence (SEQ ID NO: 136) of a native sequence PR07 13 cDN A, wherein SEQ ID NO:l36 is a clone designated herein as "DNA29101-1122". <br><br> Figure 50 shows the amino acid sequence (SEQ ID NO: 137) derived from the coding sequence of SEQ ID NO: 136 shown in Figure 49. <br><br> Figure 51 shows a nucleotide sequence (SEQ ID NO: 142) of a native sequence PR0719 cDNA, wherein SEQ ID NO: 142 is a clone designated herein as "DNA49646-1327". <br><br> Figure 52 shows the amino acid sequence (SEQ ID NO: 143) derived from the coding sequence of SEQ ID NO: 142 shown in Figure 51. <br><br> Figure 53 shows a nucleotide sequence (SEQ ID NO: 147) of a native sequence PR0771 cDNA, wherein SEQ ID NO: 147 is a clone designated herein as "DNA49829-1346". <br><br> Figure 54 shows the amino acid sequence (SEQ ID NO: 148) derived from the coding sequence of SEQ ID NO: 147 shown in Figure 53. <br><br> Figure 55 shows a nucleotide sequence (SEQ ID NO: 152) of a native sequence PR0788 cDN A, wherein SEQ ID NO: 152 is a clone designated herein as "DNA56405-1357". <br><br> Figure 56 shows the amino acid sequence (SEQ ID NO: 153) derived from the coding sequence of SEQ ID NO: 152 shown in Figure 55. <br><br> Figure 57 shows a nucleotide sequence (SEQ ID NO: 154) of a native sequence PR0792 cDN A, wherein SEQ ID NO: 154 is a clone designated herein as "DNA56352-I358". <br><br> Figure 58 shows the amino acid sequence (SEQ ID NO: 155) derived from the coding sequence of SEQ ID NO: 154 shown in Figure 57. <br><br> Figure 59 shows a nucleotide sequence (SEQ ID NO: 159) of a native sequence PR0812 cDN A, wherein SEQ ID NO: 159 is a clone designated herein as "DNA59205-1421 <br><br> Figure 60 shows the amino acid sequence (SEQ ID NO: 160) derived from the coding sequence of SEQ ID NO: 159 shown in Figure 59. <br><br> Figure 61 shows a nucleotide sequence (SEQ ID NO: 161) of a native sequence PR0865 cDN A, wherein SEQ lDNO:161 is a clone designated herein as "DNA53974-1401". <br><br> Figure 62 shows the amino acid sequence (SEQ ID NO:162) derived from the coding sequence of SEQ ID N0.161 shown in Figure 61. <br><br> Figure 63 shows a nucleotide sequence (SEQ ID NO.169) of a native sequence PR01075 cDNA, wherein SEQ ID NO: 169 is a clone designated herein as "DNA57689-1385". <br><br> Figure 64 shows the amino acid sequence (SEQ ID N0:170) derived from the coding sequence of SEQ ID NO: 169 shown in Figure 63. <br><br> Figure 65 shows a nucleotide sequence (SEQ ID NO: 180) of a native sequence PROl 126 cDNA, wherein SEQ ID NO: 180 is a clone designated herein as "DNA60615-1483". <br><br> Figure 66 shows the amino acid sequence (SEQ ID NO: 181) derived from the coding sequence of SEQ ID NO: 180 shown in Figure 65. <br><br> Figure 67 shows a nucleotide sequence (SEQ 1DN0:182) ofa native sequence PROl 130 cDNA, wherein <br><br> 21 <br><br> SEQ ID NO: 182 is a clone designated herein as "DNA59814-1486". <br><br> Figure 68 shows the amino acid sequence (SEQ ID NO: 183) derived from the coding sequence of SEQ ID NO: 182 shown in Figure 67. <br><br> Figure 69 shows a nucleotide sequence (SEQ ID NO: 190) of a native sequence PROl 154 cDNA, wherein SEQ ID N0:190 is a clone designated herein as "DNA59846-1503". <br><br> Figure 70 shows the amino acid sequence (SEQ ID NO: 191) derived from the coding sequence of SEQ ID NO: 190 shown in Figure 69. <br><br> Figure 71 shows a nucleotide sequence (SEQ ID NO: 192) of a native sequence PR01244 cDNA, wherein SEQ ID NO: 192 is a clone designated herein as "DNA64883-1526". <br><br> Figure 72 shows the amino acid sequence (SEQ ID NO: 193) derived from the coding sequence of SEQ ID NO: 192 shown in Figure 71. <br><br> Figure 73 shows a nucleotide sequence (SEQ ID NO: 194) of a native sequence PRO 1246 cDNA, wherein SEQ ID NO: 194 is a clone designated herein as "DNA64885-1529". <br><br> Figure 74 shows the amino acid sequence (SEQ ID NO: 195) derived from the coding sequence of SEQ ID NO: 194 shown in Figure 73. <br><br> Figure 75 shows a nucleotide sequence (SEQ ID NO:196) of a native sequence PR01274 cDNA, wherein SEQ ID NO: 196 is a clone designated herein as "DNA64889-I541". <br><br> Figure 76 shows the amino acid sequence (SEQ ID NO: 197) derived from the coding sequence of SEQ ID NO: 196 shown in Figure 75. <br><br> Figure 77 shows a nucleotide sequence (SEQ ID NO: 198) of a native sequence PR01286 cDNA, wherein SEQ ID NO: 198 is a clone designated herein as "DNA64903-1553". <br><br> Figure 78 shows the amino acid sequence (SEQ ID NO: 199) derived from the coding sequence of SEQ ID NO: 198 shown in Figure 77. <br><br> Figure 79 shows a nucleotide sequence (SEQ ID N0:200) of a native sequence PRO 1294 cDNA, wherein SEQ ID N0:200 is a clone designated herein as "DNA64905-1558". <br><br> Figure 80 shows the amino acid sequence (SEQ ID N0:201) derived from the coding sequence of SEQ ID N0:200 shown in Figure 79. <br><br> Figure 81 shows a nucleotide sequence (SEQ ID N0.202) of a native sequence PRO 1303 cDNA, wherein SEQ ID N0:202 is a clone designated herein as "DNA65409-1566". <br><br> Figure 82 shows the amino acid sequence (SEQ ID N0:203) derived from the coding sequence of SEQ ID N0:202 shown in Figure 81. <br><br> Figure 83 shows a nucleotide sequence (SEQ ID N0:204) of a native sequence PRO 1304 cDNA, wherein SEQ ID N0:204 is a clone designated herein as "DNA65406-1567". <br><br> Figure 84 shows the amino acid sequence (SEQ ID N0:205) derived from the coding sequence of SEQ ID N0:204 shown in Figure 83. <br><br> Figure 85 shows a nucleotide sequence (SEQ ID NO:213) of a native sequence PRO 1312 cDNA, wherein SEQ ID NO:213 is a clone designated herein as "DNA61873-1574". <br><br> Figure 86 shows the amino acid sequence (SEQ ID NO:214) derived from the coding sequence of SEQ ID <br><br> NO:213 shown in Figure 85. <br><br> Figure 87 shows a nucleotide sequence (SEQ ID NO:215) of a native sequence PRO 1313 cDNA, wherein SEQ ID NO:215 is a clone designated herein as "DNA64966-1575". <br><br> Figure 88 shows the amino acid sequence (SEQ ID NO:216) derived from the coding sequence of SEQ ID NO:215 shown in Figure 87. <br><br> Figure 89 shows a nucleotide sequence (SEQ ID NO:217) of a native sequence PRO 1376 cDNA, wherein . SEQ1DN0:217 is a clone designated herein as "DNA67300-1605". <br><br> Figure 90 shows the amino acid sequence (SEQ ID N0:218) derived from the coding sequence of SEQ ID NO:217 shown in Figure 89. <br><br> Figure 91 shows a nucleotide sequence (SEQ ID NO:219) of a native sequence PR01387 cDNA, wherein SEQ ID N0:219 is a clone designated herein as "DNA68872-I620". <br><br> Figure 92 shows the amino acid sequence (SEQ ID N0:220) derived from the coding sequence of SEQ ID NO:219 shown in Figure 91. <br><br> Figure 93 shows a nucleotide sequence (SEQ ID NO:221) of a native sequence PR01561 cDNA, wherein SEQ ID NO:22l is a clone designated herein as "DNA76538-I670". <br><br> Figure 94 shows the amino acid sequence (SEQ ID NO:222) derived from the coding sequence of SEQ ID NO:221 shown in Figure 93. <br><br> Figure 95 shows a nucleotide sequence (SEQ ID NO:226)ofa native sequence PR0216 cDNA, wherein SEQ ID NO:226 is a clone designated herein as "DNA33087". <br><br> Figure 96 shows the amino acid sequence (SEQ ID NO:227) derived from the coding sequence of SEQ ID NO:226 shown in Figure 95. <br><br> Certain statements that appear below are broader than what appears in the statements of invention above. These statements are provided in the interests of providing the reader with a better understanding of the invention and its practice. <br><br> Detailed* Description of the Invention <br><br> I. Definitions <br><br> The phrases "cardiovascular, endothelial and angiogenic disorder", "cardiovascular, endothelial and angiogenic dysfunction", "cardiovascular, endothelial or angiogenic disorder" and "cardiovascular, endothelial or angiogenic disfunction" are used interchangeably and refer in part to systemic disorders that affect vessels, such as diabetes mellitus, as well as diseases of the vessels themselves, such as of the arteries, capillaries, veins, and/or lymphatics. This would include indications that stimulate angiogenesis and/or cardiovascularization, and those that inhibit angiogenesis and/or cardiovascularization. Such disorders include, for example, arterial disease, such as atherosclerosis, hypertension, inflammatory vasculitides, Reynaud's disease and Reynaud's phenomenon, aneurysms, and arterial restenosis: venous afnd lymphatic disorders such as thrombophlebitis, lymphangitis, and lymphedema: and other vascular disorders such as peripheral vascular disease, cancer such as vascular tumors, e.g.. hemangioma (capillary and cavernous), glomus tumors, telangiectasia, bacillary angiomatosis, hemangioendothelioma, angiosarcoma, haemangiopericytoma, Kaposi's sarcoma, lymphangioma, and lymphangiosarcoma, tumor angiogenesis, trauma such as wounds, burns, and other injured tissue, implant fixation, scarring, ischemia reperfusion injury, rheumatoid arthritis, cerebrovascular disease, renal diseases such as acute <br><br> infarctions, cardiac hypertrophy, and heart failure such as CHF. <br><br> "Hypertrophy", as used herein, is defined as an increase in mass of an organ or structure independent of natural growth that does not involve tumor formation. Hypertrophy of an organ or tissue is due either to an increase in the mass of the individual cells (true hypertrophy), or to an increase in the number of cells making up the tissue (hyperplasia), or both. Certain organs, such as the heart, lose the ability to divide shortly after birth. Accordingly, "cardiac hypertrophy" is defined as an increase in mass of the heart, which, in adults, is characterized by an increase in myocyte cell size and contractile protein content without concomitant cell division. The character of the stress responsible for inciting the hypertrophy, (e.g., increased preload, increased afterload, loss of myocytes, as in myocardial infarction, or primary depression of contractility), appears to play a critical role in determining the nature of the response. The early stage of cardiac hypertrophy is usually characterized morphologically by increases in the size of myofibrils and mitochondria, as well as by enlargement of mitochondria and nuclei. At this stage, while muscle cells are larger than normal, cellular organization is largely preserved. At a more advanced stage of cardiac hypertrophy, there are preferential increases in the size or number of specific organelles, such as mitochondria, and new contractile elements are added in localized areas of the cells, in an irregular manner. Cells subjected to long-standing hypertrophy show more obv ious disruptions in eel lu lar organ izat ion, inc lud ing markedly enlarged nuclei with highly lobulated membranes, which displace adjacent myofibrils and cause breakdown of normal Z-band registration. The phrase "cardiac hypertrophy" is used to include all stages of the progression of this condition, characterized by various degrees of structural damage of the heart muscle, regardless of the underlying cardiac disorder. Hence, the term also includes physiological conditions instrumental in the development of cardiac hypertrophy, such as elevated blood pressure, aortic stenosis, or myocardial infarction. <br><br> "Heart failure" refers to an abnormality of cardiac function where the heart does not pump blood at the rate needed for the requirements of metabolizing tissues. The heart failure can be caused by a number of factors, including ischemic, congenital, rheumatic, or idiopathic forms. <br><br> "Congestive heart failure" (CHF) is a progressive pathologic state where the heart is increasingly unable to supply adequate cardiac output (the volume of blood pumped by the heart over time) to deliver the oxygenated blood to peripheral tissues. As CHF progresses, structural and hemodynamic damages occur. While these damages have a variety of manifestations, one characteristic symptom is ventricular hypertrophy. CHF is a common end result of a number of various cardiac disorders. <br><br> "Myocardial infarction" generally results from atherosclerosis of the coronary arteries, often with superimposed coronary thrombosis. It may be divided into two major types: transmural infarcts, in which myocardial necrosis involves the full thickness of the ventricular wall, and subendocardial (nontransmural) infarcts, in which the necrosis involves the subendocardium, the intramural myocardium, or both, without extending all the way through the ventricular wall to the epicardium. Myocardial infarction is known to cause both a change in hemodynamic effects and an alteration in structure in the damaged and healthy zones of the heart. Thus, for example, myocardial infarction reduces the maximum cardiac output and the stroke volume of the heart. Also associated with myocardial infarction is a stimulation of the DNA synthesis occurring in the interstice as well as an increase in the formation of collagen in the areas of the heart not affected. <br><br> As a result of the increased stress or strain placed on the heart in prolonged hypertension due, for example, <br><br> WO 00/32221 <br><br> PCT/US99/28313 - <br><br> to the increased total peripheral resistance, cardiac hypertrophy has long been associated with "hypertension". A characteristic of the ventricle that becomes hypertrophic as a result of chronic pressure overload is an impaired diastolic performance. Fouadetal.. J. Am.Coll. Cardiol.. 4:1500-1506(1984); Smith etal., J. Am. Coll. Cardiol.. 5:869-874 (1985). A prolonged left ventricular relaxation has been detected in early essential hypertension, in spite 5 of normal or supranormal systolic function. Hartford et a!.. Hypertension. 6: 329-338 (1984V However, there is no close parallelism between blood pressure levels and cardiac hypertrophy. Although improvement in left ventricu lar function in response to antihypertensive therapy has been reported in humans, patients variously treated with a diuretic (hydrochlorothiazide), a (J-blocker (propranolol), or a calcium channel blocker (diltiazem), have shown reversal of left ventricular hypertrophy, without improvement in diastolic function. Inouye et al., Am. J. 10 Cardiol.. £3: 1583-7 (1984). <br><br> Another complex cardiac disease associated with cardiac hypertrophy is "hypertrophic cardiomyopathy". <br><br> This condition is characterized by a great diversity of morphologic, functional, and clinical features (Maron et al., N. Engl. J. Med.. 316:780-789 (1987); Spirito el al., N. Engl. J. Med.. 320: 749-755 (1989); Louie and Edwards, <br><br> Prog. Cardiovasc. Pis.. 36:275-308 (1994); Wigle et al, Circulation. 92: 1680-1692 (1995)), the heterogeneity of 15 which is accentuated by the fact that it afflicts patients of all ages. Spirito et al, N. Enel. J. Med.. 336: 775-785 (1997). The causative factors of hypertrophic cardiomyopathy are also diverse and little understood. In general, mutations in genes encoding sarcomeric proteins are associated with hypertrophic cardiomyopathy. Recent data suggest that P-myosin heavy chain mutations may account for approximately 30 to 40 percent of cases of familial hypertrophic cardiomyopathy. Watkins e/a/., N. Ensl. J. Med.. 326:1108-1114(1992): Schwartz et al Circulation. 20 91:532-540 H995&gt;: Marian and Roberts. Circulation. 92: 1336-1347 (1995); Thierfeldere/ al, CeH, Th 701-712 (1994); Watkins et al, Nat. Gen.. 11:434-437 (1995). Besides P-myosin heavy chain, other locations of genetic mutations include cardiac troponin T. alpha topomyosin, cardiac myosin binding protein C. essential myosin light chain, and regulatory myosin light chain. See, Malik and Watkins. Curr. Qpin. Cardiol.. 12.: 295-302 (1997). <br><br> Supravalvu lar "aortic stenosis" is an inherited vascular disorder characterized by narrowing of the ascending 25 aorta, but other arteries, including the pulmonary arteries, may also be affected. Untreated aortic stenosis may lead to increased intracardiac pressure resulting in myocardial hypertrophy and eventually heart failure and death. The pathogenesis of this disorder is not fully understood, but hypertrophy and possibly hyperplasia of medial smooth muscle are prominent features of this disorder. It has been reported that molecular variants of the elastin gene are involved in the development and pathogenesis of aortic stenosis. U.S. Patent No. 5,650,282 issued July 22,1997. 30 "Valvular regurgitation" occurs as a result of heart diseases resulting in disorders of the cardiac valves. <br><br> Various diseases, like rheumatic fever, can cause the shrinking or pulling apart of the valve orifice, while other diseases mayresult in endocarditis, an inflammation ofthe endocardium or lining membrane ofthe atrioventricular orifices and operation of the heart. Defects such as the narrowing of the valve stenosis or the defective closing of the valve result in an accumulation of blood in the heart cavity or regurgitation of blood past the valve. If 35 uncorrected, prolonged valvular stenosis or insufficiency may result in cardiac hypertrophy and associated damage to the heart muscle, which may eventually necessitate valve replacement. <br><br> The treatment of all these, and other cardiovascular, endothelial and angiogenic disorders, which mav or mav . .. not be accompanied by cardiac hypertrophy, is described herein. |~lNTELLECTUA^_^y <br><br> 15 JUS) 2006 <br><br> 25 <br><br> REG <br><br> The terms "cancer", "cancerous", and "malignant" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, carcinoma including adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer. Hodgkin's and non-Hodgkin's lymphoma, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer such as hepatic carcinoma and hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer such as renal cell carcinoma and Wilms' tumors, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, and various types of head and neck cancer. The preferred cancers for treatment herein are breast, colon, lung, melanoma, ovarian, and others involving vascular tumors as noted above. <br><br> The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes (e.g., ml, 1251, 90 Y, and ,86Re), chemotherapeutic agents, and toxins such as enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof. <br><br> A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents, folic acid antagonists, anti-metabolites of nucleic acid metabolism, antibiotics, pyrimidine analogs, 5-fluorouraciI, cisplatin, purine nucleosides, amines, amino acids, triazol nucleosides, or corticosteroids. Specific examples include Adriamycin, Doxorubicin, 5-Fluorouracil, Cytosine arabinoside ("Ara-C"), Cyclophosphamide, Thiotepa, Busulfan, Cytoxin, Taxol, Toxotere, Methotrexate, Cisplatin, Melphalan, Vinblastine. Bleomycin, Etoposide, Ifosfamide, Mitomycin C, Mitoxantrone, Vincreistine, Vinorelbine, Carboplatin. Teniposide, Daunomycin, Carminomycin, Aminopterin, Dactinomycin, Mitomycins, Esperamicins (see U.S. Pat. No. 4.675,187), Melphalan, and other related nitrogen mustards. Also included in this definition are hormonal agents that act to regulate or inhibit hormone action on tumors, such as tamoxifen and onapristone. <br><br> A "growth-inhibitory agent" when used herein refers to a compound or composition that inhibits growth of a cell, such as an Wnt-overexpressing cancer cell, either in vitro or in vivo. Thus, the growth-inhibitory agent is one which significantly reduces the percentage of malignant cells in S phase. Examples of growth-inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest. Classical M-phase blockers include the vincas (vincristine and vinblastine), taxol, and topo II inhibitors such as doxorubicin, daunorubicin, etoposide, and bleomycin. Those agents that arrest G1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin. methotrexate,15-fluorouracil, and ara-C. Further information can be found in The Molecular Basis of Cancer. Mendelsohn and Israel, eds., Chapter I, entitled "Cell cycle regulation, oncogenes, and antineoplasticdrugs" by Murakami et al. (WB Saunders: Philadelphia, 1995), especially p. 13. Additional examples include tumor necrosis factor (TNF). an antibody capable of inhibiting or neutralizing the angiogenic activity of acidic or basic FGF or hepatocyte growth factor (HGF), an antibody capable of inhibiting or neutralizing the coagulant activities of tissue factor, protein C, or protein S (see, WO 91/01753, published 21 February 1991), or an antibody capable of binding to HER2 receptor (WO 89/06692), such as the 4D5 antibody (and functional <br><br> equivalents thereof) (e.g., WO 92/22653). <br><br> "Treatment" is an. intervention performed with the intention of preventing the development or altering the pathology of a cardiovascular, endothelial, and angiogenic disorder. The concept of treatment is used in the broadest sense, and specifically includes the prevention (prophylaxis), moderation, reduction, and curing of cardiovascular, endothelial, and angiogenic disorders of any stage. Accordingly, "treatment" refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) a cardiovascular, endothelial, and angiogenic disorder such as hypertrophy. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. The disorder may result from any cause, including idiopathic, cardiotrophic, or myotrophic causes, or ischemia or ischemic insults, such as myocardial infarction. <br><br> "Chronic" administration refers to administration ofthe agent(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial effect, such as an anti-hypertrophic effect, for an extended period of time. <br><br> "Mammal" for purposes oftreatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, sheep, pigs, etc. Preferably, the mammal is human. <br><br> Administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order. <br><br> The phrase "cardiovascular, endothelial or angiogenic agents" refers generically to any drug that acts in treating cardiovascular, endothelial, and angiogenic disorders. Examples of cardiovascular agents are those that promote vascular homeostasis by modulating blood pressure, heart rate, heart contractility, and endothelial and smooth muscle biology, all of which factors have a role in cardiovascular disease. Specific examples of these include angiotensin-!! receptor antagonists; endothelin receptor antagonists such as, for example, BOSENTAN™ and MOXONODIN™; interferon-gamma (IFN-y); des-aspartate-angiotensin 1; thrombolytic agents, e.g., streptokinase, urokinase, t-PA, and a t-PA variant specifically designed to have longer half-life and very high fibrin specificity, TNK t-PA (a T103N, N117Q, KHRR(296-299)AAAA t-PA variant, Keyt et al, Proc. Natl. Acad. Sci. USA 91. 3670-3674 (1994)); inotropic or hypertensive agents such as digoxigenin and p-adrenergic receptor blocking agents, e.g., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, and carvedilol: angiotensin converting enzyme (ACE) inhibitors, e.g., quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, and lisinopril; diuretics, e.g., chlorothiazide, hydrochlorothiazide, hydroflumethazide, methvlchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, and indapamide; and calcium channel blockers. e.g., diltiazem, nifedipine, verapamil, nicardipine. One preferred category of this type is a therapeutic agent used for the treatment of cardiac hypertrophy or of a physiological condition instrumental in the development of cardiac hypertrophy, such as elevated blood pressure, aortic stenosis, or myocardial infarction. The term 'comprising' as used in this specification and claims means 'consisting at least in part of, that is to say when interpreting statements in this specification and claims which include that term, the features, prefaced by that term in each statement, all need to be present but other features can also be present. Angiogenic agents" and "endothelial agents" are active agents that promote angiogenesis and/or endothelial tell growth, or. if applicable, vasculogenesis. This would include factors that accelerate wound healing, such as growth hormone, insulin-like growth factor-1 (IGF-1), VEGF, V1GF, PDGF, epidermal growth factor (EGF), CTGF and members of its family. FGF, and TGF-a and TGF-p. <br><br> "Angiostatic agents" are active agents that inhibit angiogenesis or vasculogenesis or otherwise inhibit or <br><br> 27 I <br><br> OF N2 <br><br> t 5 JUH 20' <br><br> 660135 1.DOC \ n r- <br><br> H t <br><br> WO 00/32221 PCT/US99/28313 <br><br> prevent growth of cancer cells. Examples include antibodies or other antagonists to angiogenic agents as defined above, such as antibodies to VEGF. They additionally include cytotherapeutic agents such as cytotoxic agents, chemotherapeutic agents, growth-inhibitory agents, apoptotic agents, and other agents to treat cancer, such as anti-HER-2, anti-CD20, and other bioactive and organic chemical agents. <br><br> In a pharmacological sense, in the context of the present invention, a "therapeutically effective amount" of an active agent such as a PRO polypeptide or agonist or antagonist thereto or an anti-PRO antibody, refers to an amount effective in the treatment of a cardiovascular, endothelial or angiogenic disorder in a mammal and can be determined empirically. <br><br> As used herein, an "effective amount" of an active agent such as a PRO polypeptide or agonist or antagonist thereto or an anti-PRO antibody, refers to an amount effective for carrying out a stated purpose, wherein such amounts may be determined empirically for the desired effect. <br><br> The terms "PRO polypeptide" and "PRO" as used herein and when immediately followed by a numerical designation refer to various polypeptides, wherein the complete designation (i.e., PRO/number) refers to specific polypeptide sequences as described herein. The terms "PRO/number polypeptide" and "PRO/number" wherein the term "number" is provided as an actual numerical designation as used herein encompass native sequence polypeptides and polypeptide variants (which are further defined herein). The PRO polypeptides described herein may be isolated from a variety of sources, such as from human tissue types or from another source, or prepared by recombinant or synthetic methods. <br><br> A "native sequence PRO polypeptide" comprises a polypeptide having the same amino acid sequence as the corresponding PRO polypeptide derived from nature. Such native sequence PRO polypeptides can be isolated from nature or can be produced by recombinant or synthetic means. The term "native sequence PRO polypeptide" specifically encompasses naturally-occurring truncated or secreted forms of the specific PRO polypeptide (e.g., an extracellular domain sequence), naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the polypeptide. In various embodiments of the invention, the native sequence PRO polypeptides disclosed herein are mature or full-length native sequence polypeptides comprising the full-length amino acids sequences shown in the accompanying figures. Start and stop codons are shown in bold font and underlined in the figures. However, while the PRO polypeptide disclosed in the accompanying figures are shown to begin with methionine residues designated herein as amino acid position I in the figures, it is conceivable and possible that other methionine residues located either upstream or downstream from the amino acid position 1 in the figures may be employed as the starting amino acid residue for the PRO polypeptides. <br><br> The PRO polypeptide "extracellular domain" or "ECD" refers to a form of the PRO polypeptide which is essentially free of the transmembrane and cytoplasmic domains. Ordinarily, a PRO polypeptide ECD will have less than 1% of such transmembrane and/or cytoplasmic domains and preferably, will have less than 0.5% of such domains. It will be understood that any transmembrane domains identified for the PRO polypeptides of the present invention are identified pursuant to criteria routinely employed in the art for identifying that type of hydrophobic domain. The exact boundaries of a transmembrane domain may vary but most likely by no more than about 5 amino acids at either end of the domain as initially identified herein. Optionally, therefore, an extracellular domain of a PRO polypeptide may contain from about 5 or fewer amino acids on either side of the transmembrane <br><br> 28 <br><br> domain/extracellular domain boundary as identified in the Examples or specification and such polypeptides, with or without the associated signal peptide, and nucleic acid encoding them, are contemplated by the present invention. <br><br> The approximate location of the "signal peptides" of the^arious PRO polypeptides disclosed herein are shown in the present specification and/or the accompanying figures. It is noted, however, that the C-terminal boundary of a signal peptide may vary, but most likely by no more than about 5 amino acids on either side of the signal peptide C-terminal boundary as initially identified herein, wherein the C-terminal boundary of the signal peptide may be identified pursuant to criteria routinely employed in the art for identifying that type of amino acid sequence element (e.g., Nielsen el al., Prot. Eng.. 10:1-6 (1997) and von Heinje et al.. Nucl. Acids Res.. 14:4683-4690 (1986)). Moreover, it is also recognized that, in some cases, cleavage of a signal sequence from a secreted polypeptide is not entirely uniform, resulting in more than one secreted speqjes. These mature polypeptides, where the signal peptide is cleaved within no more than about 5 amino acids on either side of the C-terminal boundary of the signal peptide as identified herein, and the polynucleotides encoding them, are contemplated by the present invention. <br><br> "PRO polypeptide variant" means an active PRO polypeptide as defined above or below having at least about 80% amino acid sequence identity with a full-length native sequence PRO polypeptide sequence as disclosed herein, a PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal peptide, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein. Such PRO polypeptide variants include, for instance, PRO polypeptides wherein one or more amino acid residues are added, or deleted, at the N- or C-terminus of the full-length native amino acid sequence. Ordinarily, a PRO polypeptide variant will have at least about 80% amino acid sequence identity, preferably at least about 81 % amino acid sequence identity, more preferably at least about 82% amino acid sequence identity, more preferably at least about 83% amino acid sequence identity, more preferably at least about 84% amino acid sequence identity, more preferably at least about 85% amino acid sequence identity, more preferably at least about 86% amino acid sequence identity, more preferably at least about 87% amino acid sequence identity, more preferably at least about 88% amino acid sequence identity, more preferably at least about 89% amino acid sequence identity, more preferably at least about 90% amino acid sequence identity, more preferably at least about 91% amino acid sequence identity, more preferably at least about 92% amino acid sequence identity, more preferably at least about 93% amino acid sequence identity, more preferably at least about 94% amino acid sequence identity, more preferably at least about 95% amino acid sequence identity, more preferably at least about 96% amino acid sequence identity, more preferably at least about 97% amino acid sequence identity, more preferably at least about 98% amino acid sequence identity and most preferably at least about 99% amino acid sequence identity with a full-length native sequence PRO polypeptide sequence as disclosed herein, a PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal peptide, as disclosed herein or any other specifically defined fragment of a full-length PRO polypeptide sequence as disclosed herein. Ordinarily, PRO variant polypeptides are at least about 10 amino acids in length, often at least about 20 amino acids in length, more often at least about 30 amino acids in length, more often at least about 40 amino acids in length, more often at least about 50 amino acids in <br><br> 29 <br><br> p f length, more often at least about 60 amino acids in length, more often at least aibout 70 amino acids in length, more often at least about 80 amino acids in length, more often at least about 90 amino acids in length, more often at least about 100 amino acids in length, more often at least about 150 amino acids in length, more often at least about 200 amino acids in length, more often at least about 300 amino acids in length, or more. <br><br> As shown below, Table 1 provides the complete source code forthe ALIGN-2 sequence comparison computer program. This source code may be routinely compiled for use on a UNIX operating system to provide the ALIGN-2 sequence comparison computer program. <br><br> In addition, Tables 2A-2D show hypothetical exemplifications for using the below described method to determine % amino acid sequence identity (Tables 2A-2B) and % nucleic acid sequence identity (Tables 2C-2D) using the ALIGN-2 sequence comparison computer program, wherein "PRO" represents the amino acid sequence of a hypothetical PRO polypeptide of interest, "Comparison Protein" represents the amino acid sequence of a polypeptide against which the "PRO" polypeptide of interest is being compared, "PRO-DNA" represents a hypothetical PRO-encoding nucleic acid sequence of interest, "Comparison DNA" represents the nucleotide sequence of a nucleic acid molecule against which the "PRO-DNA" nucleic acid molecule of interest is being compared, "X"," Y", and "Z" each represent different hypothetical amino acid residues and "N", "L" and "V" each represent different hypothetical nucleotides. <br><br> 30 <br><br> Table 1 <br><br> /* <br><br> * <br><br> * C-C increased from 12 to 15 <br><br> * Z is average of EQ <br><br> * B is average of ND <br><br> * match with stop is M; stop-stop = 0; J (joker) match = 0 */ <br><br> ^define _M -8 /* value of a match with a stop *1 _day[26)[26] = { <br><br> ABCDEFGHIJKLMNOPQRSTUVWXYZ*/ <br><br> ; 2. 0.-2. 0, 0,-4, 1,-1,-1. 0,-1,-2,-1, 0,_M, 1, 0,-2, 1, 1, 0, 0,-6, 0,-3, 0}, <br><br> 0. 3,-4. 3, 2.-5, 0, 1,-2. 0. 0,-3,-2, 2,_M,-1, 1. 0, 0, 0, 0,-2,-5, 0,-3, 1). <br><br> -2,-4.15,-5,-5,-4,-3,-3,-2. 0,-5,-6,-5,-4,_M,-3,-5,-4, 0,-2, 0,-2,-8, 0, 0,-5), <br><br> 0, 3.-5. 4, 3,-6, 1, 1,-2, 0, 0,-4,-3, 2,_M,-1, 2,-1, 0, 0, 0,-2,-7, 0,-4, 2&gt;, <br><br> 0, 2.-5, 3, 4,-5, 0, 1,-2, 0, 0,-3,-2, 1,_M,-1, 2,-1, 0, 0, 0,-2,-7. 0,-4, 3}, <br><br> -4,-5.-4,-6,-5, 9,-5,-2. 1. 0,-5, 2, 0,-4,_M,-5,-5,-4,-3,-3, 0,-1, 0, 0, 7,-5}, <br><br> 1, 0.-3, 1. 0.-5, 5,-2,-3. 0,-2,-4,-3, 0,_M,-l,-l,-3, 1. 0, 0,-1,-7. 0,-5, 0}, <br><br> -1. 1.-3. 1, 1,-2,-2, 6.-2. 0, 0,-2,-2, 2,_M, 0, 3, 2,-1,-1, 0,-2,-3. 0, 0, 2}, <br><br> -1,-2.-2.-2,-2, 1,-3,-2. 5. 0,-2, 2, 2,-2._M,-2,-2,-2,-1, 0, 0, 4,-5, 0,-1,-2}, <br><br> 0. 0. 0. 0, 0, 0, 0, 0, 0. 0. 0, 0. 0, 0,_M, 0, 0, 0, 0, 0, 0, 0, 0, 0. 0, 0}. <br><br> -1. 0.-5, 0. 0.-5,-2, 0.-2. 0, 5,-3, 0, 1,_M,-1, 1, 3, 0, 0, 0,-2,-3, 0,-4, 0}, <br><br> -2.-3.-6.-4,-3, 2,-4,-2, 2. 0,-3, 6, 4,-3,_M,-3,-2,-3,-3,-1,0, 2,-2, 0,-1,-2}, <br><br> -1.-2.-5,-3,-2, 0,-3,-2. 2. 0, 0, 4, 6,-2,_M,-2,-1, 0,-2.-1, 0, 2,-4, 0,-2.-1}, <br><br> 0. 2,-4. 2. 1,-4. 0, 2,-2. 0, 1,-3,-2. 2,_M,-1, 1, 0, 1. 0, 0,-2,-4. 0,-2, 1}, <br><br> M,_M,_M,_M, M, 0,_M,_M, M,_M}, <br><br> 1.-1.-3,-1,-1.-5,-1,0.-2. 0,-1,-3,-2,-1~,_M, 6, 0,0, 1,0, 0,-1,-6, 0,-5?0}7 <br><br> 0. 1.-5,2,2,-5,-1,3.-2.0, 1,-2,-1, 1,_M. 0,4, 1,-1,-1, 0,-2,-5, 0,-4, 3}, <br><br> -2, 0.-4.-1,-1,-4,-3, 2,-2. 0, 3,-3, 0, 0,_M, 0, 1, 6, 0,-1, 0,-2, 2, 0,-4, 0}, <br><br> 1, 0. 0. 0, 0,-3, 1,-1,-1. 0, 0,-3,-2, l._M. 1,-1, 0, 2, 1, 0,-1,-2. 0,-3, 0J, <br><br> 1. 0.-2, 0, 0,-3, 0,-1, 0, 0, 0,-1,-1, 0,_M, 0,-1,-1, 1, 3, 0, 0,-5, 0,-3, 0}, <br><br> 0. 0. 0, 0, 0, 0, 0, 0, 0. 0. 0, 0, 0, 0, M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0}. <br><br> 0.-2.-2,-2,-2.-1,-1,-2, 4. 0,-2, 2, 2,-27_M,-1,-2,-2,-1, 0, 0, 4,-6, 0,-2,-2}, <br><br> -6.-5,-8,-7.-7. 0,-7,-3.-5. 0,-3,-2,-4,-4,_M,-6,-5, 2,-2,-5. 0,-6,17, 0, 0,-6}. <br><br> 0, 0. 0, 0, 0. 0. 0, 0. 0. 0. 0, 0, 0, 0, M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0}. <br><br> -3.-3. 0,-4,-4, 7,-5, 0,-1. 0,-4,-l,-2,-27_M,-5,-4,-4,-3,-3, 0,-2, 0, 0,10,-4}, <br><br> 0. 1.-5. 2, 3.-5, 0, 2.-2. 0. 0,-2,-1, 1,_M, 0, 3, 0, 0, 0, 0,-2,-6, 0,-4, 4} <br><br> Page 1 of day.h <br><br> 31 <br><br> I* <br><br> *1 <br><br> ^include &lt;stdio.h&gt; <br><br> ^include <br><br> &lt;ctype.h&gt; <br><br> ^define <br><br> MAXJMP <br><br> 16 <br><br> /* max jumps in a diag */ <br><br> ^define <br><br> MAXGAP <br><br> 24 <br><br> /* don't continue to penalize gaps larger than this */ <br><br> ^define <br><br> JMPS <br><br> 1024 <br><br> /* max jmps in an path */ <br><br> ^define <br><br> MX <br><br> 4 <br><br> 1* save if there's at least MX-1 bases since last jmp */ <br><br> ^define <br><br> DMAT <br><br> 3 <br><br> /* value of matching bases */ <br><br> ^define <br><br> DMIS <br><br> 0 <br><br> I* penalty for mismatched bases *1 <br><br> ^define <br><br> DINSO <br><br> 8 <br><br> 1* penalty for a gap */ <br><br> #define <br><br> DINS1 <br><br> 1 <br><br> /* penalty per base *1 <br><br> ^define <br><br> PINSO <br><br> 8 <br><br> 1* penalty for a gap */ <br><br> ^define <br><br> PINS1 <br><br> 4 <br><br> /* penalty per residue */ <br><br> struct jmp { <br><br> short unsigned short <br><br> }; <br><br> struct diag { <br><br> int long short struct jmp <br><br> }; <br><br> struct path { int spc; short int <br><br> }: <br><br> score; <br><br> jp; <br><br> n[MAXJMP]; I* size of jmp (neg for dely) */ x[MAXJMP]; /* base no. of jmp in seq x */ /* limits seq to 2*16 -1 */ <br><br> I* score at last jmp *1 offset; /* offset of prev block */ <br><br> ijmp; /* current jmp index */ <br><br> /* list of jmps */ <br><br> /* number of leading spaces */ <br><br> n[JMPS]; I* size of jmp (gap) */ <br><br> x[JMPS]; I* loc of jmp (last elem before gap) */ <br><br> char char char char int int int int int int int int int long struct struct char char <br><br> *ofile; <br><br> *namex{2J; <br><br> ♦prog; <br><br> *seqx[2]; <br><br> dmax; <br><br> dmaxO; <br><br> dna; <br><br> endgaps; <br><br> gapx, gapy; lenO,lenl; ngapx, ngapy; smax; <br><br> *xbm: <br><br> offset; <br><br> diag *dx; path pp[2]; <br><br> I* output file name */ /* seq names: getseqs() */ <br><br> /* prog name for err msgs */ /* seqs: getseqs() */ /* best diag: nw() */ <br><br> /* final diag */ <br><br> /* set if dna: main() */ <br><br> /* set if penalizing end gaps */ /* total gaps in seqs */ <br><br> /* seq lens */ <br><br> /* total size of gaps *1 /* max score: nw() */ <br><br> /* bitmap for matching */ /* current offset in jmp file */ /* holds diagonals */ (* holds path for seqs */ <br><br> *calioc(), *malloc(), *index(). *strcpyO; *getseq(), *g_calloc(); <br><br> Page 1 of nw.h <br><br> 32 <br><br> /* Needleman-Wunsch alignment program * <br><br> * usage: progs filel file2 <br><br> * where filel and file2 are two dna or two protein sequences. <br><br> * The sequences can be in upper- or lower-case an may contain ambiguity <br><br> * Any lines beginning with ' &gt;' or' &lt;' are ignored <br><br> * Max file length is 65535 (limited by unsigned short x in the jmp struct) <br><br> * A sequence with 1/3 or more of its elements ACGTU is assumed to be DNA <br><br> * Output is in the File "align.out" <br><br> * <br><br> * The program may create a tmp file in /tmp to hold info about traceback. <br><br> * Original version developed under BSD 4.3 on a vax 8650 */ <br><br> ^include "nw.h" <br><br> ^include "day.h" <br><br> static dbval[26] = { <br><br> 1,14~2,13,0,0,4,11,0,0,12,0.3.15.0,0,0,5,6,8,8,7,9,0,10,0 <br><br> }: <br><br> static _pbval[26] = { <br><br> 1, 2[(1 &lt; &lt;('D'-'A'))|(1 &lt; &lt;CN'-'A')), 4, 8, 16, 32, 64, <br><br> 128, 256, OxFFFFFFF, I &lt; &lt; 10, I &lt; &lt; 11, 1 &lt; &lt; 12, 1 &lt; &lt; 13, 1 &lt; &lt; 14, <br><br> 1 &lt; &lt; 15, 1 &lt; &lt; 16, 1 &lt; &lt; 17. 1 &lt; &lt; 18, 1 &lt; &lt; 19, 1 &lt; &lt;20. 1 &lt; &lt;21, I &lt; &lt;22, <br><br> 1 &lt; &lt;23, 1&lt; &lt;24, 1 &lt; &lt;25|(1 &lt; &lt;('E'-'A'))](1 &lt; &lt;('Q'-'A')) <br><br> }; <br><br> main(ac, av) main int ac; <br><br> char *av[]; <br><br> { <br><br> prog = av(0]; <br><br> if (ac != 3) { <br><br> fprintf(stderr,"usage: %s filel file2\n", prog); <br><br> fprintf(stderr,"where filel and file2 are two dna or two protein sequencesAn"); fprintf(stderr, "The sequences can be in upper- or lower-case\n"); fprintf(stderr,"Any lines beginning with or ' &lt;' are ignored\n"); fprintf(stderr,"Output is in the file \"align.out\"\n"); <br><br> exit(l); <br><br> } <br><br> namex[0] = av[l]; <br><br> namexll] = avl2); <br><br> seqxfO] = getseq(namex[0], &amp;len0); <br><br> seqxfl] = getseq(namex[l], &amp;lenl); <br><br> xbm = (dna)? _dbval : _pbval: <br><br> endgaps = 0; /* 1 to penalize endgaps */ <br><br> ofile = "align.out"; * output file */ <br><br> nw(); /* fill in the matrix, get the possible jmps *1 <br><br> readjmps(); /* get the actual jmps */ <br><br> print(); I* print stats, alignment */ <br><br> cleanup(O); I* unlink any tmp files */ <br><br> Page 1 of nw.c <br><br> 33 <br><br> /* do the alignment, return best score: mainO <br><br> * dna: values in Fitch and Smith, PNAS, 80, 1382-1386, 1983 <br><br> * pro: PAM 250 values <br><br> * When scores are equal, we prefer mismatches to any gap, prefer <br><br> * a new gap to extending an ongoing gap, and prefer a gap in seqx <br><br> * to a gap in seq y. <br><br> */ <br><br> nw() IW <br><br> char <br><br> *px, *py <br><br> ; /* seqs and ptrs */ <br><br> int <br><br> ♦ndely, *dely; <br><br> /* keep track of dely */ <br><br> int ndelx, delx; <br><br> /* keep track of delx */ <br><br> int <br><br> ♦tmp; <br><br> f* for swapping rowO, rowl */ <br><br> int mis; <br><br> /* score for each type */ <br><br> int insO, insl; <br><br> l* insertion penalties *1 <br><br> register id; <br><br> /* diagonal index ♦/ <br><br> register <br><br> •j; <br><br> /* jmp index */ <br><br> register <br><br> *co!0, *coll; /* score for curr. last row */ <br><br> register xx, yy; <br><br> /* index into seqs */ <br><br> dx = (struct diag *)g_calloc("to get diags". lenO+lenl + 1, sizeof(struct diag)); <br><br> ndely = (int *)g_calloc("to get ndely", lenl + 1, sizeof(int)); <br><br> dely = (int *)g_calloc("to get delv", lenl + 1, sizeof(int)); <br><br> colO = (int *)g_calloc("to get colO". lenl + 1, sizeof(int)); <br><br> coll = (int *)g_calloc("to get coll", lenl + 1, sizeof(int)); <br><br> insO = (dna)? DINSO : PINSO: <br><br> insl = (dna)'? DINS1 : PINS1: <br><br> smax = -10000; <br><br> if (endgaps) { <br><br> for (col0[0] - dely[0] = -insO. yy = 1; yy &lt; = lenl; yy++) { <br><br> col0[yy] = dely[yy] = col0[yy-l] - insl; <br><br> ndelyjyy] = yy; <br><br> } <br><br> co!0[0] = 0; /* Waterman Bull Math Biol 84 */ <br><br> } <br><br> else for (yy = 1; yy &lt; = lenl: yy+ +) <br><br> delyfyy] = -insO: <br><br> /* fill in match matrix *1 <br><br> for (px = seqx[0], xx = 1; xx &lt; = lenO; px+ +, xx+ + ) { <br><br> /* initialize first entry in col *1 <br><br> if (endgaps) { <br><br> if (xx = = 1) <br><br> col 1 [0} = delx = -(insO+insl); <br><br> else coll[0] = delx = col0[0] - insl; <br><br> ndelx = xx: <br><br> } <br><br> else { <br><br> collfO] = 0; <br><br> delx = -insO; <br><br> ndelx = 0: <br><br> &gt; <br><br> Page 2 of nw.c <br><br> 34 <br><br> ...nw for (py = seqxfl], yy = 1; yy &lt; = lenl; py + + , yy++) { <br><br> mis = colO[yy-l]; <br><br> if (dna) <br><br> mis + = (xbm[*px-'A']&amp;xbm[*py-'A'])? DMAT : DMIS; <br><br> else mis + = _day[*px-'A'][*py-'A']; <br><br> I* update penalty for del in x seq; <br><br> * favor new del over ongong del <br><br> * ignore MAXGAP if weighting endgaps */ <br><br> if (endgaps 11 ndely[yy] &lt; MAXGAP) { <br><br> if (col0[yyj - insO &gt; = dely[yyj) { <br><br> dely[yy] = colO(yy] - (insO+insl); <br><br> ndelylyy] = 1; <br><br> } else { <br><br> dely[yy] -= insl; <br><br> ndely [yyj+ +; <br><br> } <br><br> } else { <br><br> if (colOtyyJ - (insO+insl) &gt; = dely[yy]) { <br><br> delylyy] = coI0[yy] - (insO+insl); <br><br> ndelylyy] — 1; <br><br> } else ndely(yy] + +; <br><br> } <br><br> /* update penalty for del in y seq; <br><br> * favor new del over ongong del */ <br><br> if (endgaps 11 ndelx &lt; MAXGAP) { <br><br> if (coll[yy-l] - insO &gt; = delx) { <br><br> delx = coll[yy-l] - (insO+insl); <br><br> ndelx = 1; <br><br> } else { <br><br> delx -= insl; <br><br> ndelx + +; <br><br> } <br><br> } else { <br><br> if (col 1 (yy-1 ] - (insO+insl) &gt; = delx) { <br><br> delx = colllyy-1] - (insO+insl); <br><br> ndelx =1; <br><br> } else ndelx + + ; <br><br> } <br><br> /* pick the maximum score; we're favoring <br><br> * mis over any del and delx over dely */ <br><br> Page 3 of nw.c <br><br> 35 <br><br> ...nw id = xx - yy + lenl - 1; <br><br> if (mis &gt; = delx &amp;&amp; mis &gt; = delyfyy)) <br><br> col 1 [yyl = mis; <br><br> else if (delx &gt; = delyfyy]) { <br><br> col 1 [yy] = delx; <br><br> ij = dx[id].ijmp; <br><br> if (dx[id].jp.n[0] &amp;&amp; (!dna 11 (ndelx &gt; = MAXJMP &amp;&amp; xx &gt; dx[id].jp.x[ij]+MX) || mis &gt; dx[id].score+DINSO)) { <br><br> dx[id].ijmp+ + ; <br><br> if (+ +ij &gt;= MAXJMP) { <br><br> wriiejmps(id); <br><br> ij = dx[id].ijmp = 0; <br><br> dx[id].offset = offset; <br><br> offset + = sizeof(struct jmp) + sizeof(offset); <br><br> } <br><br> } <br><br> dxlid].jp.n[ij] = ndelx; <br><br> dx[id].jp.x[ij] = xx; <br><br> dx[id].score = delx; <br><br> } <br><br> else { <br><br> coll[yy] = dely[yy]; <br><br> ij = dx[id].ijmp; <br><br> if (dx[id].jp.n[0] &amp;&amp; (!dna 11 (ndely[yyj &gt; = MAXJMP <br><br> &amp;&amp; xx &gt; dxfidj.jp.x[ij]+MX) || mis &gt; dx(id].score+DINSO)) { <br><br> dx[id].ijmp+ + ; <br><br> if ( + +ij &gt; = MAXJMP) { <br><br> writejmps(id); <br><br> ij = dx[id].ijmp = 0; <br><br> dx[id], offset = offset; <br><br> offset + = sizeof(struct jmp) + sizeof(offset); <br><br> } <br><br> &gt; <br><br> dx[id] jp.nfij] = -nde|y[yy]; <br><br> dx[id]jp.x[ij] = xx; <br><br> dx[id).score = dely[yy]; <br><br> &gt; <br><br> if (xx == Ien0&amp;&amp; yy &lt; lenl) { <br><br> /* last col */ <br><br> if (endgaps) <br><br> col Hyy] -= insO+insl*(lenl-yy); <br><br> if (coll [jy] &gt; smax) { <br><br> smax = coll(yy]; <br><br> dmax = id; <br><br> } <br><br> } <br><br> } <br><br> if (endgaps &amp;&amp; xx &lt; lenO) <br><br> collfyy-1]-= insO+insl*(lenO-xx); <br><br> if (col 1 Lyy-1J &gt; smax) { <br><br> smax = coIl[yy-l]: <br><br> dmax = id; <br><br> &gt; <br><br> tmp = colO; colO = coll; coll = tmp; <br><br> } <br><br> (void) free((char *)ndely); <br><br> (void) free((char *)dely): <br><br> (void) free((char *)col0); <br><br> (void) free((char *)coll); <br><br> Page 4 of nw.c <br><br> 36 <br><br> t* <br><br> * <br><br> * print() — only routine visible outside this module <br><br> * <br><br> * static: <br><br> * getmat() - trace back best path, count matches: print() <br><br> * pr_align() — print alignment of described in array p[]: print() <br><br> * dumpblockO - dump a block of lines with numbers, stars: pr_align() <br><br> * nums() - put out a number line: dumpblockO <br><br> * putlineO — put out a line (name, [num], seq, [num]): dumpblockO <br><br> * stars() - -put a line of stars: dumpblockO <br><br> * stripname() — strip any path and prefix from a seqname */ , <br><br> ^include "nw.h" <br><br> ^define SPC 3 ^define PLINE 256 ^define P SPC 3 <br><br> extern _day[26][26J; <br><br> int olen; <br><br> FILE *fx; <br><br> /* maximum output line */ <br><br> I* space between name or num and seq *1 <br><br> I* set output line length */ <br><br> I* output file */ <br><br> printO print <br><br> { <br><br> int Ix, ly, firstgap, lastgap; '* overlap */ <br><br> If ((fx = fopen(ofile, "w")) = = 0) { <br><br> fprintf(stderr,"%s: can't write %s\n", prog, ofile); <br><br> cleanup(l); <br><br> } <br><br> fprintf(fx, "&lt; first sequence: %s (length = %d)\n", namex[0], lenO); <br><br> fprintf(fx, "&lt;second sequence: Tcs (length = %d)\n", namex[l], lenl); <br><br> olen = 60; <br><br> Ix = lenO; <br><br> ly = lenl; <br><br> firstgap = lastgap = 0: <br><br> if (dmax &lt; lenl - I) { * leading gap in x */ <br><br> pp[0].spc = firstgap = lenl - dmax - 1; <br><br> ly -= pp[0].spc; <br><br> &gt; <br><br> else if (dmax &gt; lenl -1) { '* leading gap in y *1 pp[l].spc = firstgap = dmax - (lenl - 1); <br><br> Ix -= pp[l].spc; <br><br> } <br><br> if (dmaxO &lt; lenO -1) { * trailing gap in x */ <br><br> lastgap = lenO - dmaxO -1: <br><br> Ix -= lastgap; <br><br> } <br><br> else if (dmaxO &gt; lenO - 1) { * trailing gap in y */ <br><br> lastgap = dmaxO - OenO - 1); <br><br> ly -= lastgap; <br><br> } <br><br> getmat(lx, ly, firstgap, lastgap): <br><br> pr_align(); <br><br> Page 1 of nwprint.c <br><br> 37 <br><br> /* <br><br> * trace back the best path, count matches *t static getmat(lx, ly, firstgap. lastgap) int Ix, ly; <br><br> int firstgap, lastgap; <br><br> { <br><br> getmat <br><br> /* "core" (minus endgaps) */ I* leading trailing overlap */ <br><br> int char double register register char nm, iO, il, sizO, sizl; <br><br> outx[32]; pet; nO, nl; <br><br> pO, *pl; <br><br> I* get total matches, score *1 <br><br> iO = i 1 - sizO = sizl = 0; <br><br> pO = seqxfO] + pp[l].spc; <br><br> pi = seqx[ 1 ] + pp[0].spc; <br><br> nO = pp[l].spc + I; <br><br> nl = pp(0].spc +- 1: <br><br> nm = 0; <br><br> while ( *p0 &amp;&amp; *pl ) { <br><br> if (sizO) { <br><br> pl + + ; <br><br> nl + +: <br><br> sizO--; <br><br> &gt; <br><br> else if (sizl) { <br><br> p0+ + ; <br><br> n0+ + : <br><br> sizl—; <br><br> } <br><br> else { <br><br> if (xbmI*pO-'A']&amp;xbmI*pl-'A']) <br><br> nm+ + ; <br><br> if (n0+ + = = pp(0].x[i0]) <br><br> sizO = pp[0].n[i0++]; if (nl + + == pp[l].x[il]) <br><br> sizl = pp[l].n[il + +l; <br><br> p0+ + ; <br><br> pl + +; <br><br> /* pet homology: <br><br> * if penalizing endgaps, base is the shorier seq <br><br> * else, knock off overhangs and take shorier core *1 <br><br> if (endgaps) <br><br> lx = (lenO &lt; lenl)? lenO : lenl; <br><br> else lx = (lx &lt; ly)? Ix : ly; <br><br> pet = 100.*(double;nm/(double)lx; <br><br> fprintf(fx. "\n"); <br><br> fprintf(fx, " &lt; %d maich%s in an overlap of %d: %.2f percent similarity\n", nm. (nm == 1)? "" : "es". h. pet); <br><br> } <br><br> } <br><br> Page 2 of nwprint.c <br><br> 38 <br><br> fprintf(fx, " &lt;gaps in first sequence: %d", gapx); if (gapx) { <br><br> (void) sprintf(outx, " (%d %s%s)", <br><br> ngapx, (dna)? "baseVresidue", (ngapx = fprintf(fx,"%s", outx); <br><br> ...getmat <br><br> = !)? "":"s"); <br><br> fprintf(fx, ", gaps in second sequence: %d", gapy); <br><br> if (gapy) { <br><br> (void) sprintf(outx, " (%d %s%s)", <br><br> ngapy, (dna)? "baseVresidue", (ngapy == 1)? "":"s"); <br><br> fprintf(fx,"%s", outx); <br><br> } <br><br> if (dna) <br><br> fprintf(fx, <br><br> "\n&lt; score: %d (match = %d, mismatch = %d, gap penalty = %d + %d per base)\n" smax. DMAT, DMIS, DINSO, DINS1); <br><br> else fprintf(fx, <br><br> "\n&lt; score: %d (Dayhoff PAM 250 matrix, gap penalty = %d + %d per residue)\n", smax, PINSO. PINS1); <br><br> if (endgaps) <br><br> fprintf(fx, <br><br> "&lt;endgaps penalized, left endgap: %d %s%s, right endgap: %d %s%s\n". <br><br> firstgap, (dna)? "base" : "residue", (firstgap == 1)? "" : "s", <br><br> lastgap, (dna)? "base" : "residue", (lastgap == 1)? "" : "s"); <br><br> else fprintf(fx, "&lt; endgaps not penalizedNn"); <br><br> static nm; <br><br> I* matches in core — for checking *1 <br><br> static lmax: <br><br> /* lengths of stripped file names */ <br><br> static <br><br> UP]: <br><br> /* jmp index for a path ♦/ <br><br> static nc|2]; <br><br> 1* number at start of current line *1 <br><br> static ni[2]; <br><br> 1* current elem number - for gapping */ <br><br> static siz[2]; <br><br> static char <br><br> *ps[2]; <br><br> /* ptr to current element */ <br><br> static char <br><br> *po[2]: <br><br> /* ptr to next output char slot */ <br><br> static char out[2][P LINE]: <br><br> I* output line */ <br><br> static char star[P LINE]; <br><br> /* set by starsO *1 <br><br> * print alignment of described in struct path pp[] */ <br><br> static pr_alignO { <br><br> int nn; /* char count */ <br><br> int more; <br><br> register i; <br><br> pralign for (i = 0. lmax = 0: i &lt; 2: i+ +) { : nn = stripname(namexlij); <br><br> if (nn &gt; lmax) <br><br> lmax = nn; <br><br> ncli) = 1; ni[i] = 1; <br><br> siz[i] = ij[i] = 0: ps[i] = seqxfi]; po[i] = out[i]; <br><br> Page 3 of nwprint.c <br><br> 39 <br><br> for (nn = nm = 0, move = 1; more;) { ...pi"_align for (i = more = 0; i &lt; 2: i+ +) { <br><br> /* <br><br> * do we have more of this sequence? <br><br> *1 <br><br> if (!*ps[ij) <br><br> continue; <br><br> more++; <br><br> •f (pp[i]-spc) { /* leading space */ <br><br> *po[i]++ =* ' '; <br><br> pplij.spc--; <br><br> } <br><br> else if (siz[i]) { /* in a gap */ <br><br> *po[i] + + = <br><br> sizfi]—; <br><br> &gt; <br><br> else { /* we're putting a seq element <br><br> */ <br><br> *po[i) = *ps{i]; <br><br> if (islower(*ps[i])) <br><br> *ps[i] = toupper(*ps[i]); <br><br> po[i] + + : <br><br> psl&gt;]++: <br><br> /* <br><br> * are we at next gap for this seq? <br><br> */ <br><br> if (ni[i] == pp[i].x[ij[i]]&gt; { <br><br> /* <br><br> * we need to merge all gaps <br><br> * at this location <br><br> */ <br><br> sizfi] = ppfi].n[ij[i]+ +]; <br><br> while (nifi] == ppfi].x[ij[i]]) <br><br> sizfi] += ppfi].nfij[i]+ +]; <br><br> &gt; <br><br> nili]+ + : <br><br> } <br><br> } <br><br> if (+ +nn = = olen 11 Imore &amp;&amp; nn) { <br><br> dumpblockO; <br><br> for (i = 0; i &lt; 2; i+ +) <br><br> pofi] = outfi]; <br><br> nn = 0; <br><br> } <br><br> } <br><br> } <br><br> /* <br><br> * dump a block of lines, including numbers, stars: pr alignO */ -static dumpblockO dumpblock <br><br> { <br><br> register i; <br><br> for (i = 0; i &lt; 2; i++) <br><br> *pofi]-- = '\0'; <br><br> Page 4 of nwprint.c <br><br> 40 <br><br> (void) putc('\n\ fx): <br><br> for (i = 0; i &lt; 2; i+ +) { <br><br> if (*out[i] &amp;&amp; (*out[i] != ' 1 11 *(pofi]) ! = ' ')) { if(i ==0) <br><br> nums(i): <br><br> if (i = = 0&amp;&amp; *ou((l]) <br><br> siars(); <br><br> putline(i); <br><br> if (i = = 0 &amp;&amp; *out[l]) <br><br> fprintf(fx. star); <br><br> if (i == 1) <br><br> nums(i): <br><br> } <br><br> } <br><br> } <br><br> /* <br><br> * put out a number line: dumpblockO */ <br><br> static nums(ix) <br><br> int ix; I* index in out[] holding seq line */ <br><br> { <br><br> char nline[P_LINE]; <br><br> register i, j; <br><br> register char *pn, *px. *py; <br><br> for (pn = nline, i = 0: i &lt; lmax+P_SPC; i + + , pn++) *pn = " <br><br> for (i = ncfix], py = out[ix]; *pv: py+ +, pn+ +) { <br><br> &gt;f (*py == ' ' 11 *py == <br><br> *pn = ' ': <br><br> else { <br><br> if (i% 10 == 0 |j (i == 1 &amp;&amp; ncfix] != 1)) { j = (i &lt; 0)7 -i : i; <br><br> for (px = pn; j; j /= 10, px—) *px = j%10 + '0'; <br><br> if (i &lt; 0) <br><br> *px - <br><br> &gt; <br><br> else <br><br> *pn = ' <br><br> i+ + ; <br><br> } <br><br> &gt; <br><br> *pn = '\0'; <br><br> nc[ix] = i; <br><br> for (pn = nline; *pn: pn+ +) <br><br> (void) putc(*pn, fx): <br><br> (void) putc('\n', fx): <br><br> &gt; <br><br> I* <br><br> * put out a line (name, |num]. seq, Inum]): dumpblockO */ <br><br> static putline(ix) <br><br> int ix; <br><br> { <br><br> ...dumpblock nums putline <br><br> Page 5 of nwprint.c <br><br> 41 <br><br> ...putline int i; <br><br> register char *px; <br><br> for (px = namex[ix), i = 0; *px &amp;&amp; *px ! = px+ +, i+ +) <br><br> (void) putc(*px, fx); <br><br> for (; i &lt; lmax+P_SPC; i++) <br><br> (void) putc(" \ fx); <br><br> I* these count from 1: <br><br> * ni{] is current element (from I) <br><br> * nc[] is number at start of current line <br><br> */ <br><br> for (px = out[ix); *px: px+ +) <br><br> (void) putc(*px&amp;0x7F, fx); <br><br> (void) putc('\n\ fx); <br><br> /* <br><br> * put a line of stars (seqs always in out[0]. outfl]): dumpblockO *1 <br><br> static starso stars <br><br> { <br><br> int i; <br><br> register char *p0, *pl. cx, *px; <br><br> if (!*out[0J ! | (*out(0] =='•&amp;&amp; *(po[0]) = = ' ') 11 !*out|l] 11 (*out[l] = =■'&amp;&amp; *(pof 1 J) = ="')) <br><br> return: <br><br> px = star: <br><br> for (i = lmax + P_SPC; i; i~) <br><br> *px++ = 1 <br><br> for (pO = out[0], pi = out(l]; *p0 &amp;&amp; *pl; p0++, pl + +) { <br><br> if (isalpha(*pO) &amp;&amp; isa!pha(*pl)) { <br><br> if (xbm(*pO-'A']&amp;xbm[*pl-'A']) { <br><br> cx = <br><br> nm+ + : <br><br> } <br><br> else if (!dna &amp;&amp; ^dav^pO-'A'Jppl-'A'J &gt; 0) <br><br> cx = V: <br><br> else cx = * <br><br> &gt; <br><br> else cx = ' <br><br> *px+ + = cx: <br><br> } <br><br> *px++ = '\n'; <br><br> *px = '\0': <br><br> } <br><br> Page 6 of nwprint.c <br><br> 42 <br><br> I* <br><br> * strip path or prefix from pn, return len: pralignO «/ <br><br> static stripname(pn) Stripname char *pn; /* file name (may be path) *i <br><br> { <br><br> register char *px, *py; <br><br> py = 0; <br><br> for (px = pn; *px; px++) <br><br> if (*px - = '/') <br><br> py = px + 1; <br><br> if (py) <br><br> (void) strcpy(pn, py); <br><br> return(strlen(pn)); <br><br> } <br><br> 43 <br><br> Page 7 of nwprint.c <br><br> /* <br><br> * cleanupO -- cleanup any tmp file <br><br> * getseqO — read in seq. set dna. len. maxlen <br><br> * g_calloc() -- cailocO with error checkin <br><br> * readjmpsO - get the good jmps. from tmp file if necessary <br><br> * writejmps() ~ write a filled array of jmps to a tmp file: nw() <br><br> */ <br><br> ^include "nw.h" <br><br> ^include &lt;sys/file.h&gt; <br><br> char *jname = "/tmp/homgXXXXXX"; /* tmp file for jmps */ <br><br> FILE *fj; <br><br> int cleanupO; /* cleanup tmp file */ <br><br> long IseekO; <br><br> I* <br><br> * remove any tmp file if we blow */ <br><br> cieanup(i) cleanup int i; <br><br> { <br><br> if(fj) <br><br> (void) unlink(jname); <br><br> exit(i); <br><br> } <br><br> /* <br><br> * read, return ptr to seq, set dna. len. maxlen <br><br> * skip lines starting with or1 &gt; ' <br><br> * seq in upper or lower case */ <br><br> char * <br><br> getseq(fi)e, len) getseq char *file: /* file name */ <br><br> int *len; /* seq len */ <br><br> { <br><br> char line[1024). *pseq; <br><br> register char *px, *py: <br><br> int natgc, tlen: <br><br> FILE *fp; <br><br> if ((fp = fopen(file,"r")) == 0) { <br><br> fprintf(stderr,"%s: can't read %s\n", prog, file); <br><br> exit(l); <br><br> } <br><br> tlen = natgc = 0; <br><br> while (fgets(line, 1024, fp)) { <br><br> if (*line = = ';' | j *line = = ' &lt;' 11 *line = = ' &gt;') <br><br> continue; <br><br> for (px = line; *px != '\n': px++) <br><br> if (isupper(*px) 11 islower(*px)) <br><br> tlen+ +: <br><br> &gt; <br><br> if((pseq = malloc((unsigned)(tlen+6))) == 0) { <br><br> fprintf(stderr,"%s: mallocO failed to get %d bytes for %s\n", prog, tlen+6, file); exit(l); <br><br> } <br><br> pseq[0] = pseqfl] = pseq(2J = pseq[3] = '\0"; <br><br> Page 1 of nwsubr.c <br><br> 44 <br><br> } <br><br> ...getseq py = pseq + 4; <br><br> *len = tlen; <br><br> rewind(fp); <br><br> while (fgets(line, 1024, fp)) { <br><br> if (*line = = '11 *line = = ' &lt;' 11 *line = = ' &gt;') <br><br> continue; <br><br> for(px = line; *px != '\n'; px++) { <br><br> if (isupper(*px)) <br><br> *py + + = *px; <br><br> else if (islower(*px)) <br><br> *py + + = toupper(*px); <br><br> if (index("ATGCU",*(py-l))) <br><br> natgc + + : <br><br> } <br><br> &gt; <br><br> *py + + = *\0'; <br><br> *py = '\0'; <br><br> (void) fclose(fp); <br><br> dna = natgc &gt; (tlen/3); <br><br> return (pseq+4); <br><br> char * <br><br> g_calloc(msg, nx, sz) g_Call0C <br><br> char *msg; /* program, calling routine */ <br><br> int nx, sz; I* number and size of elements */ <br><br> { <br><br> char *px. *calloc(); <br><br> if ((px = ca!loc((unsigned)nx. (unsigned)sz)) = = 0) { <br><br> if (*msg) { <br><br> fprintf(stderr, "7cs: g_calloc() failed %s (n = %d, sz=%d)\n", prog, msg, nx, sz); exit(l); <br><br> } <br><br> } <br><br> return(px); <br><br> * get final jmps from dx[] or tmp file, set pp[]&gt; reset dmax: main() <br><br> »/ <br><br> readjmpso readjmps <br><br> { <br><br> int fd = -1; <br><br> int siz, iO. il: <br><br> register i, j, xx; <br><br> if(fi){ <br><br> (void) fclose(fj): <br><br> if ((fd = open(jname, O RDONLY, 0)) &lt; 0) { <br><br> fprintf(stderr, "Sirs: can't open() %s\n". prog, jname); <br><br> cleanup(l): <br><br> } <br><br> &gt; <br><br> for (i = iO = il = 0. dmaxO = dmax. xx = lenO; ; i++) { <br><br> while (1) { <br><br> for (j = dx[dmax].ijmp: j &gt; = 0 &amp;&amp; dx(dmax].jp.x(j] &gt; = xx; j--) <br><br> Page 2 of nwsubr.c <br><br> 45 <br><br> ...readjmps if (j &lt; 0 &amp;&amp; dxfdmax]. offset &amp;&amp; 0) { <br><br> (void) lseek(fd, dxfdmax],offset, 0); <br><br> (void) read(fd, (char *)&amp;dx[dmax].jp, sizeof(struct jmp)); <br><br> (void) read(fd, (char *)&amp;dx[dmaxj.offset, sizeof(dx[dmax].offset)); <br><br> dx[dmax].ijmp = MAXJMP-I; <br><br> &gt; <br><br> else break; <br><br> } <br><br> if (i &gt; = JMPS) { <br><br> fprimf(stderr, "%s: too many gaps in alignmeni\n", prog); <br><br> cleanupO); <br><br> } <br><br> if (j &gt; = 0) { <br><br> siz = dx[dmax].jp.n[j]; <br><br> xx = dx[dmax].jp.x[j]; <br><br> dmax += siz; <br><br> if (siz &lt; 0) { /* gap in second seq */ <br><br> pp[lj.n[il] = -siz; <br><br> xx + = siz; <br><br> /* id = xx - yy + lenl - 1 */ <br><br> pp[l].x[il) = xx - dmax + lenl - 1; <br><br> gapy++; <br><br> ngapy -= siz; <br><br> /* ignore MAXGAP when doing endgaps */ <br><br> siz = (-siz &lt; MAXGAP 11 endgaps)? -siz : MAXGAP; <br><br> il + + ; <br><br> &gt; <br><br> else if (siz &gt; 0) { I* gap in first seq */ <br><br> pp[0].n[i0J = siz; <br><br> pplOj.xiiO) = xx; <br><br> gapx++; <br><br> ngapx + = siz; <br><br> /* ignore MAXGAP when doing endgaps */ <br><br> siz = (siz &lt; MAXGAP 11 endgaps)? siz : MAXGAP; <br><br> i0+ + : <br><br> &gt; <br><br> } <br><br> else break; <br><br> } <br><br> /* reverse the order of jmps *1 <br><br> for (j = 0. i0~; j &lt; iO; j + +. iO-) { <br><br> i = pplPl.nlj]; pplOl.nfo] = pplOJ.nliO]; ppl0].n[i0] = i; <br><br> i = pp(OJ.xU); pp[0].x[j] = pp[0].x[i0]; pp[0].xfi0] = i; <br><br> } <br><br> for (j = 0. il--; j &lt; il; j+ + , il--) { <br><br> i = pp(l].n[j); pp[l].n[j] = ppllj.nfilj; pp(l].n[il] = i; <br><br> i = ppll].x(j]; pp[l].x[j] = pp|l].x[ilj; pp[l].x[il] = i; <br><br> } <br><br> if (fd &gt; = 0) <br><br> (void) close(fd): <br><br> if(fj){ <br><br> (void) unlink(jname); <br><br> fj = o.offset = 0;}} Page 3 of nwsubr.c <br><br> 46 <br><br> /* <br><br> * write a filled jmp struct offset of the prev one (if any): nw() <br><br> */ <br><br> writejmps(ix) writejmps int ix; <br><br> { <br><br> char *mktempO; <br><br> if(!fj){ <br><br> if (mktemp(jname) &lt; 0) { <br><br> fprintf(stderr, "%s: can't mktempO %s\n", prog, jname); <br><br> cleanup(l); <br><br> } <br><br> if ((fj = fopen(jname, "w")) == 0) { <br><br> fprintf(stderr, "%s: can't write %s\n", prog, jname); <br><br> exit(l): <br><br> } <br><br> } <br><br> (void) fwrite((char *)&amp;dx[ix].jp. sizeof(struct jmp), I, fj); <br><br> (void) fwrite((char *)&amp;dx[ix].offset, sizeof(dx[ix].offset), 1, fj); <br><br> } <br><br> 47 <br><br> Page 4 of nwsubr.c <br><br> Table 2A <br><br> PRO XXXXXXXXXXXXXXX (Length = 15 amino acids) <br><br> Comparison Protein XXXXXYYYYYYY (Length = 12 amino acids) <br><br> % amino acid sequence identity = <br><br> (the number of identically matching amino acid residues between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of amino acid residues of the PRO polypeptide) = <br><br> 5 divided by 15 = 33.3% <br><br> 48 <br><br> Table 2B <br><br> PRO XXXXXXXXXX (Length = 10 amino acids) <br><br> Comparison Protein XXXXXYYYYYYZZYZ (Length = 15 amino acids) <br><br> % amino acid sequence identity = <br><br> (the number of identically matching amino acid residues between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of amino acid residues of the PRO polypeptide) = <br><br> 5 divided by 10 = 50% <br><br> 49 <br><br> Table 2C <br><br> PRO-DNA NNNNNNNNNNNNNN (Length = 14 nucleotides) <br><br> Comparison DNA NNNNNNLLLLLLLLLL (Length = 16 nucleotides) <br><br> % nucleic acid sequence identity = <br><br> (the number of identically matching nucleotides between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the PRO-DNA nucleic acid sequence) = <br><br> 6 divided by 14 = 42.9% <br><br> 50 <br><br> Table 2D <br><br> PRO-DNA NNNNNNNNNNNN (Length = 12 nucleotides) <br><br> Comparison DNA NNNNLLLVV (Length = 9 nucleotides) <br><br> % nucleic acid sequence identity = <br><br> (the number of identically matching nucleotides between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the PRG-DNA nucleic acid sequence) = <br><br> 4 divided by 12 = 33.3% <br><br> 51 <br><br> "Percent (%) amino acid sequence identity" with respect to the PRO polypeptide sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a PRO sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence 5 identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. For purposes herein, however, % amino acid sequence 10 identity values are obtained as described below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Table 1. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code shown in Table 1 has been filed with user documentation in the U.S. Copyright Office. Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available through Genentech, 15 Inc., South San Francisco. California or may be compiled from the source code provided in Table I. The ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. <br><br> For purposes herein, the % amino acid sequence identity of a given am ino ac id sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or 20 comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: <br><br> 100 times the fraction X/Y <br><br> where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program s alignment of A and B, and where Y is the total number of amino acid residues in B. 25 It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. As examples of % amino acid sequence identity calculations, Tables 2A-2B demonstrate how to calculate the % amino acid sequence identity of the amino acid sequence designated "Comparison Protein" to the amino acid sequence designated "PRO". <br><br> 30 Unless specifically stated otherwise, al) % amino acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program. However, % amino acid sequence identity may also be detenu ined using the sequence comparison program NCB1-BLAST2 (Altschul etal., Nucleic Acids Res.. 25:3389-3402 (1997)). The NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih.gov. NCBI-BLAST2 uses several search parameters, wherein all of those search 35 parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, <br><br> 52 <br><br> minimum low complexity length = 1515. multi-pass e-value = 0.01, constant for multi-pass = 25, dropoff for final gapped alignment = 25 and scoring matrix = BLOSUM62. <br><br> In situations where NCBI-BLAST2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can 5 alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: <br><br> 100 times the fraction X/Y <br><br> where X is the number of amino acid residues scored as identical matches by the sequence alignment program NCB1-BLAST2 in that program's alignment of A and B, and where Y is the total number of amino acid residues 10 in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. <br><br> In addition, % amino acid sequence identity may also be determined using the WU-BLAST-2 computer program (Altschul et al. Methods in Enzvmologv. 266:460-480 (1996)). Most of the WU-BLAST-2 search 15 parameters are set to the default values. Those not set to default values, i.e., the adjustable parameters, are set with the following values: overlap span = I, overlap fraction = 0.125, word threshold (T) =11, and scoring matrix = BLOSUM62. For purposes herein, a % amino acid sequence identity value is determined by dividing (a) the number of matching identical amino acids residues between the amino acid sequence of the PRO polypeptide of interest having a sequence derived from the native PRO polypeptide and the comparison amino acid sequence of 20 interest (i.e., the sequence against which the PRO polypeptide of interest is being compared which may be a PRO variant polypeptide) as determined by WU-BLAST-2 by (b) the total number of amino acid residues of the PRO polypeptide of interest. For example, in the statement "a polypeptide comprising an amino acid sequence A which has or having at least 80% amino acid sequence identity to the amino acid sequence B", the amino acid sequence A is the comparison amino acid sequence of interest and the amino acid sequence B is the amino acid sequence of 25 the PRO polypeptide of interest. <br><br> "PRO variant polynucleotide" or "PRO variant nucleic acid sequence" means a nucleic acid molecule which encodes an active PRO polypeptide as defined below and which has at least about 80% nucleic acid sequence identity with a nucleic acid sequence encoding a full-length native sequence PRO polypeptide sequence as disclosed herein, a full-length native sequence PRO polypeptide sequence lacking the signal peptide as disclosed herein, an 30 extracellular domain of a PRO polypeptide^ with or without the signal peptide, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein. Ordinarily, a PRO variant polynucleotide will have at least about 80% nucleic acid sequence identity, more preferably at least about 81% nucleic acid sequence identity, more preferably at least about 82% nucleic acid sequence identity, more preferably at least about 83% nucleic acid sequence identity, more preferably at least about 84% nucleic acid sequence identity, more 35 preferably at least about 85% nucleic acid sequence identity, more preferably at least about 86% nucleic acid <br><br> 53 <br><br> sequence identity, more preferably at least about 87% nucleic acid sequence identity, more preferably at least about 88% nucleic acid sequence identity, more preferably at least about 89% nucleic acid sequence identity, more preferably at least about 90% nucleic acid sequence identity, more preferably at least about 91% nucleic acid sequence identity, more preferably at least about 92% nucleic acid sequence identity, more preferably at least about 5 93% nucleic acid sequence identity, more preferably at least about 94% nucleic acid sequence identity, more preferably at least about 95% nucleic acid sequence identity, more preferably at least about 96% nucleic acid sequence identity, more preferably at least about 97% nucleic acid sequence identity, more preferably at least about 98% nucleic acid sequence identity and yet more preferably at least about 99% nucleic acid sequence identity with a nucleic acid sequence encoding a full-length native sequence PRO polypeptide sequence as disclosed herein, a 10 full-length native sequence PRO polypeptide sequence lacking the signal peptide as disclosed herein, an extracellular domain of a PRO polypeptide, with or without the signal sequence, as disclosed herein or any other fragment of a full-length PRO polypeptide sequence as disclosed herein. Variants do not encompass the native nucleotide sequence. <br><br> Ordinarily, PRO variant polynucleotides are at least about 30 nucleotides in length, often at least about 60 15 nucleotides in length, more often at least about 90 nucleotides in length, more often at least about 120 nucleotides in length, more often at least about 150 nucleotides in length, more often at least about 180 nucleotides in length, more often at least about 210 nucleotides in length, more often at least about 240 nucleotides in length, more often at least about 270 nucleotides in length, more often at least about 300 nucleotides in length, more often at least about 450 nucleotides in length, more often at least about 600 nucleotides in length, more often at least about 900 20 nucleotides in length, or more. <br><br> "Percent (%) nucleic acid sequence identity" with respect to the PRO polypeptide-encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in a PRO polypeptide-encoding nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of 25 determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. For purposes herein, however, % nucleic acid sequence identity values are obtained as described 30 below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Table I. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code shown in Table I has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, 35 California or may be compiled from the source code provided in Table 1. The ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary. <br><br> 54 <br><br> For purposes herein, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows: <br><br> 5 100 times the fraction W/Z <br><br> where W is the number of nucleotides scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment ofC and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. As examples of % 10 nucleic acid sequence identity calculations, Tables 2C-2D demonstrate how to calculate the % nucleic acid sequence identity of the nucleic acid sequence designated "Comparison DNA" to the nucleic acid sequence designated "PRO-DNA". <br><br> Unless specifically stated otherwise, all % nucleic acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program. However, % nucleic acid sequence 15 identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et at.. Nucleic Acids Res.. 25:3389-3402(1997)). The NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih.gov. NCB1-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, minimum low complexity length = 15/5, multi-pass e-value = 0.01, constant for multi-pass = 25, dropoff for final 20 gapped alignment = 25 and scoring matrix = BLOSUM62. <br><br> In situations where NCBI-BLAST2 is employed for sequence comparisons, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows: <br><br> 25 100 times the fraction W/Z <br><br> where W is the number of nucleotides scored as identical matches by the sequence alignment program NCBI-BLAST2 in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. 30 In addition, % nucleic acid sequence identity values may also be generated using the WU-BLAST-2 <br><br> computer program (Altschul et at.. Methods in Enzvmology. 266:460-480 (1996)). Most of the WU-BLAST-2 search parameters are set to the default values. Those not set to default values, i.e., the adjustable parameters, are set with the following values: overlap span = 1, overlap fraction = 0.125, word threshold (T) = II, and scoring <br><br> 55 <br><br> WO 00/32221 PCT/US99/28313 - <br><br> matrix = BLOSUM62. For purposes herein, a % nucleic acid sequence identity value is determined by dividing (a) the number of matching identical nucleotides between the nucleic acid sequence of the PRO polypeptide-encoding nucleic acid molecule of interest having a sequence derived from the native sequence PRO polypeptide-encoding nucleic acid and the comparison nucleic acid molecule of interest (i.e., the sequence against which the 5 PRO polypeptide-encoding nucleic acid molecule of interest is being compared which may be a variant PRO polynucleotide) as determined by WU-BLAST-2 by (b) the total number of nucleotides of the PRO polypeptide-encoding nucleic acid molecule of interest. For example, in the statement "an isolated nucleic acid molecule comprising a nucleic acid sequence A which has or having at least 80% nucleic acid sequence identity to the nucleic acid sequence B", the nucleic acid sequence A is the comparison nucleic acid molecule of interest and the nucleic 10 acid sequence B is the nucleic acid sequence of the PRO polypeptide-encoding nucleic acid molecule of interest. <br><br> In other embodiments, PRO variant polynucleotides are nucleic acid molecules that encode an active PRO polypeptide and which are capable of hybridizing, preferably under stringent hybridization and wash conditions, to nucleotide sequences encoding the full-length PRO polypeptide shown in Figure 2 (SEQ ID NO:4), Figure 4 (SEQ ID NO:9), Figure 6 (SEQ ID NO: 14), Figure 8 (SEQ ID NO: 16), Figure 10 (SEQ ID NO:21), Figure 12 15 (SEQ IDNO:26), Figure 14 (SEQ IDNO:31), Figure 16 (SEQ IDNO:36), Figure 18 (SEQ IDNO:41), Figure20 (SEQ ID NO:46), Figure 22 (SEQ ID NO:51), Figure 24 (SEQ ID NO:56), Figure 26 (SEQ ID NO:62), Figure 28 (SEQ ID NO:67), Figure 30 (SEQ ID NO:72), Figure 32 (SEQ ID NO:77), Figure 34 (SEQ ID NO:85), Figure 36 (SEQ ID N0:90), Figure 38 (SEQ ID NO:98), Figure 40 (SEQ ID NO: 107), Figure 42 (SEQ ID NO: 112), Figure 44 (SEQ ID NO: 117), Figure 46 (SEQ ID NO: 127), Figure 48 (SEQ ID NO: 132), Figure 50 (SEQ ID NO: 137), 20 Figure 52 (SEQ ID NO: 143), Figure 54 (SEQ ID NO: 148), Figure 56 (SEQ ID NO: 153), Figure 58 (SEQ ID NO: 155), Figure 60 (SEQ ID NO: 160), Figure 62 (SEQ ID NO: 162), Figure 64 (SEQ ID NO: 170), Figure 66 (SEQ ID NO: 181), Figure 68 (SEQ ID NO: 183), Figure 70 (SEQ ID NO: 191), Figure 72 (SEQ ID NO: 193), Figure 74 (SEQ ID NO: 195), Figure 76 (SEQ ID NO: 197), Figure 78 (SEQ ID NO: 199), Figure 80 (SEQ ID N0:201), Figure 82 (SEQ ID N0:203), Figure 84 (SEQ ID N0:205), Figure 86 (SEQ ID NO:214), Figure 88 (SEQ ID NO:216), 25 Figure 90 (SEQ ID NO:218), Figure 92 (SEQ ID N0:220), Figure 94 (SEQ ID NO:222),and Figure 96 (SEQ ID NO:227), respectively. PRO variant polypeptides may be those that are encoded by a PRO variant polynucleotide. <br><br> The term "positives", in the context of the am ino acid sequence identity comparisons performed as described above, includes amino acid residues in the sequences compared that are not only identical, but also those that have similar properties. Amino acid residues that score a positive value to an amino acid residue of interest are those 30 that are either identical to the amino acid residue of interest or are a preferred substitution (as defined in Table 3 below) of the amino acid residue of interest. <br><br> For purposes herein, the % value of positives of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % positives to, with, or against a given amino acid sequence B) is calculated as follows: <br><br> 35 100 times the fraction X/Y <br><br> 56 <br><br> where X is the number of amino acid residues scoring a positive value by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % positives of A to B will not equal the % positives of B to A. <br><br> 5 "Isolated", when used to describe the various polypeptides disclosed herein, means a polypeptide that has been identified and separated and/or recovered from a component of its natural environment. Preferably, the isolated polypeptide is free of association with all components with which it is naturally associated. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous 10 solutes. In preferred embodiments, the polypeptide will be purified (1) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or, preferably, silver stain. Isolated polypeptide includes polypeptide in situ within recombinant cells, since at least one component of the PRO natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one 15 purification step. <br><br> An "isolated" nucleic acid molecule encoding a PRO polypeptide or an "isolated" nucleic acid molecule encoding an anti-PRO antibody is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source ofthe PRO-encoding nucleic acid or the natural source of the anti-PRO-encoding nucleic acid. Preferably, the isolated nucleic acid is 20 free of association with all components with which it is naturally associated. An isolated PRO-encoding nucleic acid molecule or an isolated anti-PRO-encoding nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the PRO-encoding nucleic acid molecule or from the anti-PRO-encoding nucleic acid molecule as it exists in natural cells. However, an isolated nucleic acid molecule encoding a PRO polypeptide or an isolated nucleic acid molecule encoding an anti-25 PRO antibody includes PRO-nucleic acid molecules or anti-PRO-nucleic acid molecules contained in cells that ordinarily express PRO polypeptides or anti-PRO antibodies where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells. <br><br> The term "control sequences" refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, 30 include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenvlation signals, and enhancers. <br><br> Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example. DNA for a presequence or secretory leader is operably linked to DNA for a PRO polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or 35 enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, <br><br> 57 <br><br> contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice. <br><br> "Stringency" ofhybridization reactions is readily determinable by one of ordinary skill in the art, and generally 5 is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows 10 that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency ofhybridization reactions, see, Ausubel etal., Current Protocols in Molecular Biology (Wiley Interscience Publishers, 1995). <br><br> "Stringent conditions" or "high-stringency conditions", as defined herein, may be identified by those that: (1) employ low ionic strength and high temperature for washing, for example, 0.015 M sodium chloride/0.0015 M 15 sodium citrate/0. l%sodium dodecyl sulfate at 50°C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at42°C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Den hard t's solution, sonicated salmon sperm DNA (50 20 ^ig/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 55 °C, followed by a high-stringency wash consisting of 0.1 x SSC containing EDTA at 55 °C. <br><br> "Moderately-stringent conditions" may be identified as described by Sambrook el a!., Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Press, 1989), and include the use of washing solution and 25 hybridization conditions (e.g., temperature, ionic strength, and % SDS) less stringent than those described above. An example of moderately stringent conditions is overnight incubation at 37 °C in a solution comprising: 20% formamide. 5 x SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/ml denatured sheared salmon sperm DNA, followed by washing the filters in 1 x SSC at about 37-50°C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. 30 as necessary to accommodate factors such as probe length and the like. <br><br> The modifier "epitope-tagged" when used herein refers to a chimeric polypeptide comprising a PRO polypeptide fused to a "tag polypeptide". The tag polypeptide has enough residues to provide an epitope against which an antibody can be made, yet is short enough such that it does not interfere with activity of the polypeptide to which it is fused. The tag polypeptide preferably also is fairly unique so that the antibody does not substantially 35 cross-react with other epitopes. Suitable tag polypeptides generally have at least six amino acid residues and usually between about 8 and 50 amino acid residues (preferably, between about 10 and 20 amino acid residues). <br><br> "Active" or "activity" in the context of PRO variants refers to form(s) of PRO proteins that retain the biologic <br><br> 58 <br><br> WO 00/32221 PCT/US99/28313 - <br><br> and/or immunologic activities of a native or naturally-occurTing PRO polypeptide. <br><br> "Biological activity" in the context of a molecule that antagonizes a PRO polypeptide that can be identified by the screening assays disclosed herein (e.g., an organic or inorganic small molecule, peptide, etc.) is used to refer to the ability of such molecules to bind or complex with the PRO polypeptide identified herein, or otherwise 5 interfere with the interaction of the PRO polypeptides with other cellular proteins or otherwise inhibits the transcription or translation of the PRO polypeptide. Particularly preferred biological activity includes cardiac hypertrophy, activity that acts on systemic disorders that affect vessels, such as diabetes mellitus, as well as diseases of the arteries, capillaries, veins, and/or lymphatics, and cancer. <br><br> The term "antagonist" is used in the broadest sense, and includes any molecule that partially or fully blocks, 10 inhibits, or neutralizes one or more of the biological activities of a native PRO polypeptide disclosed herein, for example, if applicable, its mitogenic or angiogenic activity. Antagonists of a PRO polypeptide may act by interfering with the binding of a PRO polypeptide to a cellular receptor, by incapacitating or killing cells that have been activated by a PRO polypeptide, or by interfering with vascular endothelial cell activation after binding of a PRO polypeptide to a cellular receptor. All such points of intervention by a PRO polypeptide antagonist shall be 15 considered equivalent for purposes of this invention. The antagonists inhibit the mitogenic. angiogenic, or other biological activity of PRO polypeptides, and thus are useful for the treatment of diseases or disorders characterized by undesirable excessive neovascularization, including by way of example tumors, and especially solid malignant tumors, rheumatoid arthritis, psoriasis, atherosclerosis, diabetic and other retinopathies, retrolental fibroplasia, age-related macular degeneration, neovascular glaucoma, hemangiomas, thyroid hyperplasias (including Grave's 20 disease), corneal and other tissue transplantation, and chronic inflammation. The antagonists also are useful for the treatment of diseases or disorders characterized by undesirable excessive vascular permeability, such as edema associated with brain tumors, ascites associated with malignancies, Meigs' syndrome, lung inflammation, nephrotic syndrome, pericardial effusion (such as that associated with pericarditis), and pleural effusion. In a similar manner, the term "agonist" is used in the broadest sense and includes any molecule that mimics a biological activity of a 25 native PRO polypeptide disclosed herein. Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, fragments, or amino acid sequence variants of native PRO polypeptides, peptides, small organic molecules, etc. <br><br> A "small molecule" is defined herein to have a molecular weight below about 500 dahons. <br><br> The term "PRO polypeptide receptor" as used herein refers to a cellular receptor for a PRO polypeptide, 30 ordinarily a cell-surface receptor found on vascular endothelial cells, as well as variants thereof that retain the ability to bind a PRO polypeptide. <br><br> "Antibodies" (Abs) and "immunoglobulins" (Igs) are glycoproteins having the same structural characteristics. While antibodies exhibit binding specificity to a specific antigen, immunoglobulins include both antibodies and other antibody-like molecules that lack antigen specificity. Polypeptides of the latter kind are, for example, 35 produced at low levels by the lymph system and at increased levels by myelomas. The term "antibody" is used in the broadest sense and specifically covers, without limitation, intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies) formed from at least two intact antibodies, and antibody <br><br> 59 <br><br> fragments, so long as they exhibit the desired biological activity. <br><br> "Native antibodies" and "native immunoglobulins" are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the 5 heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed 10 to form an interface between the light- and heavy-chain variable domains. <br><br> The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody to and for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called complementarity-determining regions (CDRs) or hypervariable regions 15 both in the light-chain and the heavy-chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FR). The variable domains of native heavy and light chains each comprise four FR regions, largely adopting a p-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the P-sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the 20 antigen-binding site of antibodies. See. Kabat et al.. NIH Publ. No.91-3242. Vol. I. pages 647-669 (1991). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity. <br><br> "Antibody fragments" comprise a portion of an intact antibody, preferably the antigen-binding or variable region of the intact antibody. Examples of antibody fragments include Fab. Fab', F(ab'),, and Fv fragments; 25 diabodies;linearantibodies(Zapata etal., Protein Eng.. 8(10): 1057-1062 (1995)): single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. <br><br> Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments,each with a single antigen-binding site, and a residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab'), fragment that has two antigen-combining sites and is still capable of cross-linking 30 antigen. <br><br> "Fv" is the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. 35 However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. <br><br> The Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) <br><br> 60 <br><br> of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab'), antibody fragments originally were produced as pairs of Fab' fragments that have hinge cysteines between 5 them. Other chemical couplings of antibody fragments are also known. <br><br> The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (k) and lambda (A), based on the amino acid sequences of their constant domains. <br><br> Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can 10 be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM; and several ofthese may be further divided into subclasses (isotypes), e.g., IgG 1, lgG2, IgG3, IgG4, IgA, and lgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called a, 6, e, Y, and fx, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. <br><br> 15 The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), 20 each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontam inated by other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the 25 present invention may be made by the hybridoma method first described by Kohler et al, Nature. 256:495 (1975), i or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al. Nature. 352: 624-628 (1991)and Marks et al.. J. Mol. Biol.. 222: 581-597 (1991), for example. <br><br> The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a 30 portion of the heavy and 'or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity. U.S. Patent No. 4,816,567; Morrison eta!., Proc. Natl. Acad. Sci. USA. 81: 6851-35 6855(1984). <br><br> "Humanized" forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab', F(ab'): or other antigen-binding subsequences of antibodies) <br><br> 61 <br><br> that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv FR residues ofthe human immunoglobulin are replaced 5 by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and maximize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those 10 of a human immunoglobulin sequence. The humanized antibody preferably also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones etal, Nature. 321: 522-525 (1986); Reichmann et al., Nature. 332: 323-329 (1988); and Presta, Curr. Op. Struct. Biol.. 2: 593-596 (1992). The humanized antibody includes a PRIMATIZED™ antibody wherein the antigen-binding region of the antibody is derived from an antibody produced by immunizing macaque monkeys 15 with the antigen of interest. <br><br> "Single-chain Fv" or "sFv" antibody fragments comprise the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding. For a review of sFv see, Pluckthun in The Pharmacology of Monoclonal Antibodies. Vol. 113. Rosenburg and Moore, 20 eds. (Springer-Verlag: New York, 1994), pp. 269-315. <br><br> The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH - VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two 25 antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA. 90: 6444-6448 (1993). <br><br> An "isolated" antibody is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or 30 nonproteinaceous solutes. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. Isolated antibody includes the antibody in situ within recombinant cells, since at 35 least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step. <br><br> The word "label" when used herein refers to a detectable compound or other composition that is conjugated <br><br> 62 <br><br> directly or indirectly to the antibody so as to generate a "labeled" antibody. The label may be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition that is detectable. Radionuclides that can serve as detectable labels include, for example, 1-131,1-123,1-125, Y-90, Re-188, At-211, Cu-67, Bi-212, and Pd-109. The label may 5 also be a non-detectable entity such as a toxin. <br><br> By "solid phase" is meant a non-aqueous matrix to which an antibody of the present invention can adhere. Examples of solid phases encompassed herein include those formed partially or entirely of glass (e.g., controlled pore glass), polysaccharides (e.g., agarose), polyacry lam ides, polystyrene, polyvinyl alcohol and silicones. In certain embodiments, depending on the context, the solid phase can comprise the well of an assay plate; in others 10 it is a purification column (e.g., an affinity chromatography column). This term also includes a discontinuous solid phase of discrete particles, such as those described in U.S. Patent No. 4.275,149. <br><br> A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactant that is | useful for delivery of a drug (such as the PRO polypeptide or antibodies thereto disclosed herein) to a mammal. <br><br> The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement 15 of biological membranes. <br><br> As used herein, the term "immunoadhesin" designates antibody-like molecules that combine the binding specificity of a heterologous protein (an "adhesin") with the effector functions of immunoglobulin constant domains. Structurally, the immunoadhesins comprise a fusion of an amino acid sequence with the desired binding specificity that is other than the antigen recognition and binding site of an antibody (i.e., is "heterologous"), and 20 an immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as IgG-1, lgG-2, lgG-3, or IgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD, or lgM. <br><br> 11. Compositions and Methods of the Invention ^25 A. PRO Variants <br><br> In addition to the full-length native sequence PRO polypeptides described herein, it is contemplated that PRO variants can be prepared. PRO variants can be prepared by introducing appropriate nucleotide changes into the PRO DNA, and/or by synthesis ofthe desired PRO polypeptide. Those skilled in the art will appreciate that amino acid changes may alter post-translational processes of the PRO polypeptide, such as changing the number or 30 position of glycosylation sites or altering the membrane anchoring characteristics. <br><br> Variations in the native full-length sequence PRO polypeptide or in various domains of the PRO polypeptide described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No. 5,364,934. Variations may be a substitution, deletion or insertion of one or more codons encoding the PRO polypeptide that results in a change in the amino acid 35 sequence of the PRO polypeptide as compared with the native sequence PRO polypeptide. Optionally the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the PRO <br><br> 63 <br><br> 10 <br><br> 15 <br><br> 20 <br><br> 25 <br><br> 30 <br><br> 35 <br><br> polypeptide. Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found by comparing the sequence of the PRO polypeptide with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology. Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, i.e., conservative amino acid replacements. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids. The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full-length or mature native sequence. <br><br> In particular embodiments, conservative substitutions of interest are shown in Table 3 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes, denominated exemplary substitutions in Table 3, or as further described below in reference to amino acid classes, are introduced and the products screened. <br><br> Table 3 <br><br> Original <br><br> Exemplary <br><br> Preferred <br><br> Residue <br><br> Substitutions <br><br> Substitutions <br><br> Ala (A) <br><br> val; leu; ile val <br><br> Arg (R) <br><br> lys; gin; asn lys <br><br> Asn (N) <br><br> gin; his; lys; arg gin <br><br> Asp (D) <br><br> glu glu <br><br> Cys(C) <br><br> ser ser <br><br> Gin (Q) <br><br> asn asn <br><br> Glu (E) <br><br> asp asp <br><br> Gly(G) <br><br> pro; ala ala <br><br> His (H) <br><br> asn; gin; lys; arg arg <br><br> He (I) <br><br> leu: val; met; ala; phe; <br><br> norleucine leu <br><br> Leu (L) <br><br> norleucine; ile; val; <br><br> met; ala; phe ile <br><br> Lys (K) <br><br> arg; gin; asn arg <br><br> Met (M) <br><br> leu; phe; ile leu <br><br> Phe (F) <br><br> leu; val; ile; ala; tyr leu <br><br> Pro (P) <br><br> ala ala <br><br> Ser(S) <br><br> thr thr <br><br> Thr (T) <br><br> ser ser <br><br> Trp(W) <br><br> tyr; phe tyr <br><br> Tyr(Y) <br><br> trp; phe; thr; ser phe <br><br> Val (V) <br><br> ile: leu; met; phe; <br><br> ala; norleucine leu <br><br> 40 Substantial modifications in function or immunological identity of the PRO polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are <br><br> 64 <br><br> divided into groups based on common side-chain properties: <br><br> (1) hydrophobic: norleucine, met, ala, val, leu, ile; <br><br> (2) neutral hydrophilic: cys, ser, thr; <br><br> (3) acidic: asp, glu; <br><br> (4) basic: asn, gin, his, lys, arg; <br><br> (5) residues that influence chain orientation: gly, pro; and <br><br> (6) aromatic: trp, tyr, phe. <br><br> Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Such substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites. <br><br> The variations can be made using methods known in the art such as oligonucleotide-mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis [Carter et al., Nucl. Acids Res.. 13:4331 (1986): Zoller et al., Nucl. Acids Res.. 10:6487 (1987)], cassette mutagenesis [Wells et al., Gene. 34:315 (1985)], restriction selection mutagenesis [Wells et a!., Philos. Trans. R. Soc. London SerA. 317:415 (1986)] or other known techniques can be performed on the cloned DNA to produce the PRO variant DNA. <br><br> Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant [Cunningham and Wells. Science. 244: 1081-1085(1989)]. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions fCreighton. The Proteins. (W.H. Freeman &amp; Co., N.Y.);Chothia, J. Mol. Biol.. 150:1 (1976)]. Ifalanine substitution does not yield adequate amounts of variant, an isoteric amino acid can be used. <br><br> B. Modifications of PRO Polypeptides Covalent modifications of PRO polypeptides are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of a PRO polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the PRO polypeptide. Derivatization with Afunctional agents is useful, for instance, for crosslinking a PRO polypeptide to a water-insoluble support matrix or surface for use in the method for purifying anti-PRO antibodies, and vice-versa. Commonly used crosslinkingagents include, e.g., 1,1 -bis(diazoacety l)-2-pheny lethane, glutaraldehyde, N-hydroxy-succinimide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis(succinimidylpropionate), bifunctional maleimides such as bis-N-maleimido-1.8-octane and agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate. <br><br> Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of servl or threonv I residues, methylation of the a-amino groups of lysine, arginine, and histidine side chains <br><br> 65 <br><br> [T.E. Creighton, Proteins: Structure and Molecular Properties. W.H. Freeman &amp; Co., San Francisco, pp. 79-86 (1983)], acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. <br><br> Another type of covalent modification of the PRO polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of the polypeptide. "Altering the native glycosylation pattern" 5 is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence PRO polypeptides (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence PRO polypeptide, in addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present. 10 Addition of glycosylation sites to the PRO polypeptide may be accomplished by altering the amino acid sequence. The alteration may be made, for example, by the addition of, or substitution by, one or more serine or threonine residues to the native sequence PRO polypeptide (for O-linked glycosylation sites). The PRO amino acid | sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the PRO polypeptide at preselected bases such that codons are generated that will translate into the desired amino 15 acids. <br><br> Another means of increasing the number of carbohydrate moieties on the PRO polypeptide is by chem ical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem.. pp. 259-306 (1981). <br><br> Removal of carbohydrate moieties present on the PRO polypeptide may be accomplished chemically or 20 enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al., Arch. Biochem. Biophvs.. 259:52 (1987) and by Edge et a!., Anal. Biochem.. 118:131(1981). Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al, Meth. EnzvmoL 138:350 (1987). <br><br> 25 Another type of covalent modification of PRO polypeptides comprises linking the PRO polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337. <br><br> The PRO polypeptides of the present invention may also be modified in a way to form a chimeric molecule 30 comprising the PRO polypeptide fused to another, heterologous polypeptide or amino acid sequence. <br><br> In one embodiment, such a chimeric molecule comprises a fusion of the PRO polypeptide with a tag polypeptide which provides an epitope to which an anti-tag antibody can selectively bind. The epitope tag is generally placed at the amino- or carboxyl- terminus of the PRO polypeptide. The presence of such epitope-tagged forms of the PRO polypeptide can be detected using an antibody against the tag polypeptide. Also, provision of 35 the epitope tag enables the PRO polypeptide to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag. Various tag polypeptides and their respective antibodies are well known in the art. Examples include poly-histidine (poly-His) or poly-histidine-glycine (poly- <br><br> 66 <br><br> WO 00/32221 PCT/US99/28313 - <br><br> His-gly) tags; the flu HA tag polypeptide and its antibody I2CA5 [Field et al., Mol. Cell. Biol.. 8:2159-2165 (1988)]; the c-myc tag and the 8F9. 3C7,6E10, G4, B7 and 9E10 antibodies thereto [Evan et al., Molecular and Cellular Biology. 5:3610-3616 (1985)]; and the Herpes Simplex vims glycoprotein D (gD) tag and its antibody [Paborsky etal., Protein Engineering. 3(6):547-553 (1990)]. Other tag polypeptides include the Flag-peptide [Hopp 5 et al., BioTechnologv. 6:1204-1210 (1988)]; the KT3 epitope peptide [Martin et al., Science. 255:192-194 (1992)]; an ((-tubulin epitope peptide [Skinner et al., J. Biol. Chem.. 266:15163-15166 (1991)]; and the T7 gene 10 protein peptide tag [Lutz-Freyermuth etal.. Proc. Natl. Acad. Sci. USA. 82:6393-6397 (1990)]. <br><br> In an alternative embodiment, the chimeric molecule may comprise a fusion of the PRO polypeptide with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also 10 referred to as an "immunoadhesin"). such a fusion could be to the Fc region of an IgG molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a PRO polypeptide in place of at least one variable region within an Ig molecule. In a particularly preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgG 1 molecule. Forthe production of immunoglobulin fusions see also, US Patent No. 5,428,130 issued June 27,1995. <br><br> 15 C. Preparation of the PRO polypeptides <br><br> The present invention provides newly identified and isolated nucleotide sequences encoding a polypeptide referred to in the present application as PRO 1313 (SEQ ID NO: 216), Figure 88. In particular, <br><br> cDNAs encoding identified and isolated, as disclosed in further detail in the Examples below. It is noted that proteins produced in 20 separate expression rounds may be given different PRO numbers but the UNQ number is unique for any given DNA and the encoded protein, and will not be changed. However, for sake of simplicity, in the present specification the protein encoded by DNA359I6-1161, DMA23339-1130, DNA16451-1388, DNA27865-1091, DNA27864-1155, DNA28497-1130. DNA26847-I395, DNA30942-1134, DNA32286-1191, DNA33094-1131, DNA33221-1133, DNA34434-1139. DNA35558-1167, DNA35638-1141, DNA33473-1176, DNA38260-1180, ^ . DNA39969-1185,DNA40628-1216.DN A35595-1228,DN A40981 -1234.DNA47470-1130-P1DNA47365-1206, DNA44I84-13I9, DNA48613-1268. DNA29I01-1122, DNA49646-1327, DNA49829-I346, DNA56405-1357, DNA56352-1358, DNA59205-1421.DNA53974-1401, DNA57689-1385, DNA60615-1483, DNA59814-1486, h DNA59846-1503, DNA64883-I526. DNA64885-1529, DNA64889-1541, DNA64903-1553, DNA64905-1558, DNA65409-I566, DNA65406-1567. DNA61873-1574, DMA64966-1575, DNA67300-I605, DNA68872-1620, 30 DNA76538-1670, or DNA33087 as well as all further native homologues and variants included in the foregoing definition of PRO, will be referred to as "PRO", respectively, regardless of their origin or mode of preparation. ' The description below relates primarily to production of PRO polypeptides by culturing cells transformed or itransfected with a vector containing nucleic acid encoding PRO polypeptides. It is, of course, contemplated that (alternative methods that are well known in the art may be employed to prepare PRO polypeptides. For instance, 35 tthe PRO polypeptide sequence, or portions thereof, may be produced by direct peptide synthesis using solid-phase ^techniques. See, e. p.. Stewart et al.. Solid-Phase Peptide Synthesis (W.H. Freeman Co.: San Francisco, CA, 1969); <br><br> Merrifleld, J. Am. Chem. Soc.. 85:2149-2154 (1963). In vitro protein synthesis may be ^ <br><br> of n.l. <br><br> 67 ',15 JUM 20® <br><br> RECEJ <br><br> techniques or by automation. Automated synthesis may be accomplished, for instance, with an Applied Biosystems Peptide Synthesizer (Foster City. CA) using manufacturer's instructions. Various portions of a PRO polypeptide may be chemically synthesized separately and combined using chemical or enzymatic methods to produce the full-length PRO polypeptide. <br><br> 5 i. Isolation of DNA Encoding PRO Polypeptides <br><br> DNA encoding a PRO polypeptide may be obtained from a cDNA library prepared from tissue believed to possess the.mRNA encoding the PRO polypeptide and to express it at a detectable level. Accordingly, DNAs encoding human PRO polypeptides can be conveniently obtained from cDNA libraries prepared from human tissues, such as described in the Examples. The gene encoding a PRO polypeptide may also be obtained from a 10 genomic library or by oligonucleotide synthesis. <br><br> Libraries can be screened with probes (such as antibodies to the PRO polypeptide or oligonucleotides of at least about 20-80 bases) designed to identify the gene of interest or the protein encoded by it. Screening the cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al., supra. An alternative means to isolate the gene encoding a PRO polypeptide is to use PCR 15 methodology. Sambrook et al., supra; Dieffenbach et al, PCR Primer: A Laboratory Manual /New York: Cold Spring Harbor Laboratory Press, 1995). <br><br> The Examples below describe techniques for screening a cDNA library. The oligonucleotide sequences selected as probes should be of sufficient length and sufficiently unambiguous that false positives are minimized. The oligonucleotide is preferably labeled such that it can be detected upon hybridization to DNA in the library being 20 screened. Methods of labeling are well known in the art, and include the use of radiolabels like 32P-labeled ATP, biotinylation, or enzyme labeling. Hybridization conditions, including moderate stringency and high stringency, are provided in Sambrook et al., supra. <br><br> Sequences identified in such library screening methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private sequence databases. 25 Sequence identity (at either the amino acid or nucleotide level) within defined regions of the molecule or across the full-length sequence can be determined through sequence alignment using computer software programs such as ALIGN, DNAstar, and INHERIT, which employ various algorithms to measure homology. <br><br> Nucleic acid having protein coding sequence may be obtained by screening selected cDNA or genomic libraries using the deduced amino acid sequence disclosed herein for the first time, and, if necessary, using 30 conventional primer extension procedures as described in Sambrook et al., supra, to detect precursors and processing intermediates of mRNA that may not have been reverse-transcribed into cDNA. <br><br> ii. Selection and Transformation of Host Cells Host cells are transfected or transformed with expression or cloning vectors described herein for PRO polypeptide production and cultured in conventional nutrient media modified as appropriatefor inducing promoters, 35 selecting transformants. or amplifying the genes encoding the desired sequences. The culture conditions, such as <br><br> 68 <br><br> media, temperature, pH, and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach. M. Butler, ed. (1RL Press, 1991) and Sambrook etal, supra. <br><br> 5 Methods of transfection are known to the ordinarily skilled artisan, for example, CaPOj treatment and electroporation. Depending on the host cell used, transformation is performed using standard techniques appropriate to such cells. The calcium treatment employing calcium chloride, as described in Sambrook et al, supra, or electroporation is generally used for prokaryotes or other cells that contain substantial cell-wall barriers. Infection with Agrobacteriwn tumefaciens is used for transformation of certain plant cells, as described by Shaw 10 et al., Gene. 23: 315 (1983) and WO 89/05859 published 29 June 1989. For mammalian cells without such cell walls, the calcium phosphate precipitation method of Graham and van der Eb. Virology. 52:456-457 (1978) can be employed. General aspects of mammalian cell host system transformations have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of Van Solingen et al, J. Bact.. 130: 946 (1977) and Hsiao et al, Proc. Natl. Acad. Sci. (USA). 76: 3829 (1979). However, other 15 methods for introducing DNA into cells, such as by nuclear microinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene or polyornithine, may also be used. For various techniques for transforming mammalian cells, see, Keown et al. Methods in Enzvmology. 185:527-537 (1990) and Mansour et al., Nature. 336: 348-352 (1988). <br><br> Suitable host cells for clon ing or expressing the DNA in the vectors herein include prokaryote, yeast, or higher 20 eukaryote cells. Suitable prokaryotes include, but are not limited to, eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli. Various E. coli strains are publicly available, such as E. coli K12 strain MM294 (ATCC 31,446); E. coli X1776 (ATCC 31.537); E. coli strain W3110 (ATCC 27,325); and K5 772 (ATCC 53.635). Other suitable prokaryotic host cells include Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g.. Salmonella typhimurium, 25 Serratia, e.g., Serratia marcescans. and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis4 IP disclosed in DD 266,710 published 12 April 1989), Psendomonas such as P. aeruginosa, And Streptomyces. These examples are illustrative rather than limiting. Strain W3110 is one particularly preferred host or parent host because it is a common host strain for recombinant DNA product fermentations. Preferably, the host cell secretes minimal amounts of proteolytic enzymes. For example, strain W3110 may be modified to effect a 30 genetic mutation in the genes encoding proteins endogenous to the host, with examples of such hosts including E. coli W3110 strain IA2, which has the complete genotype tonA ;E. co//W3l 10 strain 9E4, which has the complete genotype tonA ptr3; E. coli W3110 strain 27C7 (ATCC 55,244), which has the complete genotype tonA ptr3 phoA E15 (argF-lac)169 degP ompTkanr; E. coli W3110 strain 37D6, which has the complete genotype tonA ptr3 phoA EI5 (argF-lac) 169 degP ompT rbs7 HvG karf \ E. coli W3110strain40B4, which is strain 37D6 with a non-35 kanamycin resistant degP deletion mutation; and an E. coli strain having mutant periplasmic protease disclosed in U.S. Patent No. 4,946.783 issued 7 August 1990. Alternatively,, in vitro methods of cloning, e.g., PCR or other nucleic acid polymerase reactions, are suitable. <br><br> 69 <br><br> In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for vectors encoding PRO polypeptides. Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism. Others include Schizosaccharomycespombe (Beach and Nurse, Nature. 290: 140 [1981]; EP 139,383 published 2 May 1985); Klvyveromyces hosts (U.S. Patent No. 4,943,529; Fleer et al., 5 Bio/Technology. 9: 968-975 (1991)) such as, e.g., K. lactis (MW98-8C, CBS683, CBS4574; Louvencourt et at., J. BacterioL 737 [1983]), K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. wa/tii(ATCC 56,500), K. drosophilarum (ATCC 36,906; Van den Berg et al. Bio/Technology. 8: 135 (1990)), AT. thermotolerans, and K. marxiamis; yarrowia (EP 402,226); Pichia pastor is (EP 183,070; Sreekrishna el al., J. Basic Microbiol.. 28: 265-278 [ 1988]); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa 10 (Case et al., Proc. Natl. Acad. Sci. USA. 76: 5259-5263 [1979]); Schwanniomyces such as Schwanniomyces occidental is (EP 394,538 published 31 October 1990); and filamentousfungi such as, e.g., Neurospora, PenicUlium, Tolypocladium (WO 91/00357 published 10 January 1991), and Aspergillus hosts such as A. nidulans (Ballance et al., Biochem. Biophvs. Res. Commun.. 112: 284-289 [1983]; Tilburn et al.. Gene. 26: 205-221 [1983]; Yelton et al., Proc. Natl. Acad. Sci. USA. 81: 1470-1474 [1984]) and A. niger (Kelly and Hynes, EMBO J.. 4: 475-479 15 [1985]). Methylotropic yeasts are suitable herein and include, but are not limited to, yeast capable of growth on methanol selected from the genera consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotornla. A list of specific species that are exemplary of this class of yeasts may be found in C. Anthony, The Biochemistry of Methviotrophs. 269 (1982). <br><br> Suitable host cells for the expression of nucleic acid encoding a glycosylated PR.O polypeptide are derived 20 from multicellular organisms. Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, as well as plant cells. Examples of useful mammalian host cell lines include Chinese hamster ovary (CHO) and COS cells. More specific examples include monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al.. J. Gen Virol.. 36: 59 (1977)); Chinese hamster ovary ceIls/-DHFR (CHO, Urlaub and 25 Chasin, Proc. Natl. Acad. Sci. USA. 77:4216(1980)); mouse Sertoli cells (TM4. Mather, Biol. Reprod.. 23:243-251 (1980)); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); and mouse mammary tumor (MMT 060562, ATCC CCL51). The selection of the appropriate host cell is deemed to be within the skill in the art. <br><br> iii. Selection and Use of a Replicable Vector 30 The nucleic acid (e.g., cDNA or genomic DNA) encoding a PRO polypeptide may be inserted into a replicable vector for cloning (amplification of the DNA) or for expression. Various vectors are publicly available. The vector may, for example, be in the form of a plasmid, cosmid, viral particle, or phage. The appropriate nucleic acid sequence may be inserted into the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art. Vector components generally include, but are 35 not limited to. one or more of a signal sequence if the sequence is to be secreted, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Construction of <br><br> 70 <br><br> suitable vectors containing one or more of these components employs standard ligation techniques that are known to the skilled artisan. <br><br> The PRO polypeptide may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence or other polypeptide having a specific cleavage site 5 at the N-terminus of the mature protein or polypeptide. In general, the signal sequence may be a component ofthe vector, or it may be a part of the DNA encoding the PRO polypeptide that is inserted into the vector. The signal sequence may be a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders. For yeast secretion the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and K/uyveromyces a-factor leaders, the latter 10 described in U.S. Patent No. 5,010.182), or acid phosphatase leader, the C. albicans glucoamylase leader (EP 362,179 published 4 April 1990), or the signal described in WO 90/13646 published 15 November 1990. In mammalian cell expression, mammalian signal sequences may be used to direct secretion of the protein, such as | signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders. <br><br> Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one 15 or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2^ plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV, or BPV) are useful for cloning vectors in mammalian cells. <br><br> Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker. 20 Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g.. the gene encoding D-alanine racemase for Bacilli. <br><br> An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the nucleic acid encoding a PRO polypeptide, such as DHFR or thymidine kinase. An 25 appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared | and propagated as described by Urlaub et al., Proc. Natl. Acad. Sci. USA. 77: 4216 (1980). A suitable selection gene for use in yeast is the trp 1 gene present in the yeast plasmid YRp7. Stinchcomb et a!., Nature. 282:39 (1979); Kingsman et al.. Gene. 7: i41 (1979): Tschemper et al., Gene. 10:157(1980). The trp] gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or 30 PEP4-1. Jones, Genetics. 85: 12 (1977). <br><br> Expression and cloning vectors usually contain a promoter operably linked to the nucleic acid sequence encoding a PRO polypeptide to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the P-lactamase and lactose promoter systems (Chang et at.. Nature. 275: 615 (1978); Goeddel et a!., Nature. 281: 544 (1979)), alkaline phosphatase, a 35 tryptophan (trp) promoter system (Goeddel, Nucleic Acids Res.. 8:4057 (1980): EP 36,776), and hybrid promoters such as the tac promoter. deBoer et al., Proc. Natl. Acad. Sci. USA. 80: 21-25 (1983). Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the PRO <br><br> 71 <br><br> polypeptide. <br><br> Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem.. 255:2073 (1980)) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg.. 7:149 (1968): Holland, Biochemistry. j7:4900 (1978)). such as enolase, glyceraldehyde-5 3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. <br><br> Otheryeast promoters that are inducible promoters having the additional advantage of transcription controlled by growth conditions are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, 10 degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657. <br><br> PRO nucleic acid transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 July 1989), 15 adenovirus (such as Adenovirus 2). bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus, and Simian Virus 40 (SV40); by heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter; and by heat-shock promoters, provided such promoters are compatible with the host cell systems. <br><br> Transcription of a DNA encoding the PRO polypeptide by higher eukaryotes may be increased by inserting 20 an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, a-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and 25 adenovirus enhancers. The enhancer may be spliced into the vector at a position 5' or 3' to the sequence coding for a PRO polypeptide, but is preferably located at a site 5' from the promoter. <br><br> Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated 30 regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the PRO polypeptide. <br><br> Still other methods, vectors, and host cells suitable for adaptation to the synthesis of a PRO polypeptide in recombinant vertebrate cell culture are described in Gething et al., Nature. 293: 620-625 (1981): Mantei et al., Nature. 281: 40-46(1979): EP 117.060; and EP 117,058. <br><br> 35 iv. Detecting Gene Amplification/Expression <br><br> Gene amplification and/or expression may be measured in a sample directly, for example, by conventional <br><br> 72 <br><br> Southern blotting. Northern blotting to quantitate the transcription of mRNA (Thomas. Proc. Natl. Acad. Sci. USA. 27:5201 -5205 (1980)), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes. RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected. <br><br> Gene expression, alternatively, may be measured by immunological methods, such as immunohistochemical staining of cells or tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native-sequence PRO polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to DNA encoding the PRO polypeptide and encoding a specific antibody epitope. <br><br> v. Purification of Polypeptide <br><br> Forms of PRO polypeptides may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., TR1TON-X™ 100) or by enzymatic cleavage. Cells employed in expression of nucleic acid encoding the PRO polypeptide can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell-lysing agents. <br><br> It may be desired to purify the PRO polypeptide from recombinant cell proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms ofthe PRO polypeptide. Various methods of protein purification may be employed and such methods are known in the art and described, for example, in Deutscher, Methods in Enzvmology. 182 (1990); Scopes. Protein Purification: Principles and Practice (Springer-Verlag: New York, 1982). The purification step(s) selected will depend, for example, on the nature of the production process used and the particular PRO polypeptide produced. <br><br> D. Uses of the PRO polypeptides i. Assays for Cardiovascular. Endothelial, and Angiogenic Activity <br><br> Various assays can be used to test the polypeptide herein for cardiovascular, endothelial, and angiogenic activity. Such assays include those provided in the Examples below. <br><br> Assays for testing for endothelin antagonist activity, as disclosed in U.S. Pat. No. 5,773,414, include a rat heart ventricle binding assay where the polypeptide is tested for its ability to inhibit iodinized endothelin-l binding <br><br> 73 <br><br> in a receptor assay, an endothelin receptor binding assay testing for intact cell binding of radiolabeled endothelin-1 using rabbit renal artery vascular smooth muscle cells, an inositol phosphate accumulation assay where functional activity is determined in Rat-1 cells by measuring intra-cellular levels of second messengers, an arachidonic acid release assay that measures the ability of added compounds to reduce endothelin-stimulated arachidonic acid release 5 in cultured vascular smooth muscles, in vitro (isolated vessel) studies using endothelium from male New Zealand rabbits, and in vivo studies using male Sprague-Dawley rats. <br><br> Assays for tissue generation activity include, without limitation, those described in WO 95/16035 (bone, cartilage, tendon); WO 95/05846 (nerve, neuronal), and WO 91/07491 (skin, endothelium). <br><br> Assays for wound-healing activity include, for example, those described in Winter, Epidermal Wound 10 Healing. Maibach, HI and Rovee, DT, eds. (Year Book Medical Publishers, Inc., Chicago), pp. 71-112, as modified by the article of Eaglstein and Mertz, J. Invest. Dermatol.. 71: 382-384 (1978). <br><br> An assay to screen for a test molecule relating to a PRO polypeptide that binds an endothelin B, (ETB,) ^ receptor polypeptide and modulates signal transduction activity involves providing a host cell transformed with a DNA encoding endothelin B, receptor polypeptide, exposing the cells to the test candidate, and measuring 15 endothelin B, receptor signal transduction activity, as described, e.g., in U.S. Pat. No. 5,773,223. <br><br> There are several cardiac hypertrophy assays. In vitro assays include induction of spreading of adult rat cardiac myocytes. In this assay, ventricular myocytes are isolated from a single (male Sprague-Dawley) rat, essentially following a modification ofthe procedure described in detail by Piper et at., "Adult ventricular rat heart muscle cells"' in Cell Culture Techniques in Heart and Vessel Research. H.M. Piper, ed. (Berlin: Springer-Verlag, 20 1990), pp. 36-60. This procedure permits the isolation of adult ventricular myocytes and the long-term culture of these cells in the rod-shaped phenotype. Phenylephrine and Prostaglandin F2(I (PGF2o) have been shown to induce a spreading response in these adult cells. The inhibition of myocyte spreading induced by PGF;&lt;t or PGF,0 analogs (e.g., fluprostenol) and phenylephrine by various potential inhibitors of cardiac hypertrophy is then tested. <br><br> One example of an in vivo assay is a test for inhibiting cardiac hypertrophy induced by fluprostenol in vivo. 25 This pharmacological model tests the ability of the PRO polypeptide to inhibit cardiac hypertrophy induced in rats ^ (e.g., male Wistar or Sprague-Dawley) by subcutaneous injection of fluprostenol (an agonist analog of PGF,B). It is known that rats with pathologic cardiac hypertrophy induced by myocardial infarction have chronically elevated levels of extractable PGF^ in their myocardium. Lai et al.. Am. J. Phvsiol. (Heart Circ. Physiol.). 271: H2J97-H2208 (1996). Accordingly, factors that can inhibit the effects of fluprostenol on myocardial growth in vivo are 30 potentially useful for treating cardiac hypertrophy; The effects of the PRO polypeptide on cardiac hypertrophy are determined by measuring the weight of heart, ventricles, and left ventricle (normalized by body weight) relative to fluprostenol-treated rats not receiving the PRO polypeptide. <br><br> Another example of an in vivo assay is the pressure-overload cardiac hypertrophy assay. For in vivo testing it is common to induce pressure-overload cardiac hypertrophy by constriction of the abdominal aorta of test 35 animals. In a typical protocol, rats (e.g., male Wistar or Sprague-Dawley) are treated under anesthesia, and the abdominal aorta of each rat is narrowed down just below the diaphragm. Beznak M., Can. J. Biochem. Phvsiol.. 33: 985-94 (1955). The aorta is exposed through a surgical incision, and a blunted needle is placed next to the <br><br> 74 <br><br> vessel. The aorta is constricted with a ligature of silk thread around the needle, which is immediately removed and which reduces the lumen of the aorta to the diameter of the needle. This approach is described, for example, in Rossi et al. Am. Heart J.. 124: 700-709 (1992) and O'Rourke and Reibel, P.S.E.M.B.. 200: 95-100 (1992). <br><br> In yet another in vivo assay, the effect on cardiac hypertrophy following experimentally induced myocardial 5 infarction (MI) is measured. Acute MI is induced in rats by left coronary artery ligation and confirmed by electrocardiographic examination. A sham-operated group of animals is also prepared as control animals. Earlier data have shown that cardiac hypertrophy is present in the group of animals with MI, as evidenced by an 18% increase in heart weight-to-body weight ratio. Lai et al. supra. Treatment of these animals with candidate blockers of cardiac hypertrophy, e.g.. a PRO polypeptide, provides valuable information about the therapeutic 10 potential of the candidates tested. One further such assay test for induction of cardiac hypertrophy is disclosed in U.S. Pat. No. 5,773,415, using Sprague-Dawley rats. <br><br> For cancer, a variety of well-known animal models can be used to further understand the role of the genes ^ identified herein in the development and pathogenesis of tumors, and to test the efficacy of candidate therapeutic agents, including antibodies and other antagonists of the native PRO polypeptides, such as small-molecule 15 antagonists. The in vivo nature of such models makes them particularly predictive of responses in human patients. Animal models of tumors and cancers (e.g., breast cancer, colon cancer, prostate cancer, lung cancer, etc.) include both non-recombinant and recombinant (transgenic) animals. Non-recombinant animal models include, for example, rodent, e.g., murine models. Such models can be generated by introducing tumor cells into syngeneic mice using standard techniques, e.g.. subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal 20 implantation, implantation under the renal capsule, or orthopin implantation, e.g., colon cancer cells implanted in colonic tissue. See, e.g., PCT publication No. WO 97/33551, published September 18, 1997. Probably the most often used animal species in oncological studies are immunodeflcient mice and. in particular, nude mice. The observation that the nude mouse with thymic hypo/aplasia could successfully act as a host for human tumor xenografts has lead to its widespread use for this purpose. The autosomal recessive nu gene has been introduced 25- into a very large number of distinct congenic strains of nude mouse, including, for example, ASW, A/He, AKR, ^ BALB/c, BIO.LP, CI7, C3H, C57BL. C57, CBA, DBA, DDD, I/st,NC, NFR, NFS, NFS/N, NZB, NZC, NZW, P, RIH, and SJL. In addition, a wide variety of other animals with inherited immunological defects other than the nude mouse have been bred and used as recipients of tumor xenografts. For further details see, e.g., The Nude Mouse in Oncology Research. E. Boven and B. Winograd. eds. (CRC Press, Inc., 1991). <br><br> 30 The cells introduced into such animals can be derived from known tumor/cancer cell lines, such as any of the above-listed tumor cell lines, and. for example, the B104-1-1 cell line (stable NIH-3T3 cell line transfected with the neu protooncogene); ras-transfected NIH-3T3 cells; Caco-2 (ATCC HTB-37); or a moderately well-differentiated grade II human colon adenocarcinoma cell line, HT-29 (ATCC HTB-38); or from tumors and cancers. Samples of tumor or cancer cells can be obtained from patients undergoing surgery, using standard conditions 35 involving freezing and storing in liquid nitrogen. Karmali et al, Br. J. Cancer. 48: 689-696 (1983). <br><br> Tumor cells can be introduced into animals such as nude mice by a variety of procedures. The subcutaneous (s.c.) space in mice is very suitable for tumor implantation. Tumors can be transplanted s.c. as solid blocks, as <br><br> 75 <br><br> needle biopsies by use of a trochar. or as cell suspensions. For solid-block or trochar implantation, tumor tissue fragments of suitable size are introduced into the s.c. space. Cell suspensions are freshly prepared from primary tumors or stable tumor cell lines, and injected subcutaneously. Tumor cells can also be injected as subdermal implants. In this location, the inoculum is deposited between the lower part of the dermal connective tissue and 5 the s.c. tissue. <br><br> Animal models of breast cancer can be generated, for example, by implanting rat neuroblastoma cells (from which the neu oncogene was initially isolated), or /^/-transformed N1H-3T3 cells into nude mice, essentially as described by Drebin et al. Proc. Nat. Acad. Sci. USA. 83: 9129-9133 (1986). <br><br> Similarly, animal models of colon cancer can be generated by passaging colon cancer cells in animals, e.g., 10 nude mice, leading to the appearance of tumors in these animals. An orthotopic transplant model of human colon cancer in nude mice has been described, for example, by Wang et al., Cancer Research. 54:4726-4728 (1994) and Too et al.. Cancer Research. 55: 681-684 (1995). This model is based on the so-called "METAMOUSE"™ sold by Anticancer, Inc., (San Diego, California). <br><br> Tumors that arise in animals can be removed and cultured in vitro. Cells from the in vitro cultures can then 15 be passaged to animals. Such tumors can serve as targets for further testing or drug screening. Alternatively, the tumors resulting from the passage can be isolated and RNA from pre-passage cells and cells isolated after one or more rounds of passage analyzed for differential expression of genes of interest. Such passaging techniques can be performed with any known tumor or cancer cell lines. <br><br> For example, Meth A. CMS4, CMS5, CMS21, and WEH1-164 are chemically induced fibrosarcomas of 20 BALB/c female mice (DeLeo et al.. J. Exp. Med.. 146: 720 (1977)), which provide a highly controllable model system for studying the anti-tumor activities of various agents. Palladino et al., J. Immunol.. 138: 4023-4032 (1987). Briefly, tumor cells are propagated in vitro in cell culture. Prior to injection into the animals, the cell lines are washed and suspended in buffer, at a cell density of about 10xl06 to lOxlO7 cells/ml. The animals are then infected subcutaneously with 10 to 100 /ul of the cell suspension, allowing one to three weeks for a tumor to appear. 25 In addition, the Lewis lung (3LL) carcinoma of mice, which is one of the most thoroughly studied experimental tumors, can be used as an investigational tumor model. Efficacy in this tumor model has been correlated with beneficial effects in the treatment of human patients diagnosed with small-cell carcinoma of the lung (SCCL). This tumor can be introduced in normal mice upon injection of tumor fragments from an affected mouse or of cells maintained in culture. Zupi et al., Br. J. Cancer. 41: suppl. 4,30 (1980). Evidence indicates that tumors 30 can be started from injection of even a single cell and that a very high proportion of infected tumor cells survive. For further information about this tumor model see, Zacharski, Haemostasis. 16.: 300-320 (1986). <br><br> One way of evaluating the efficacy of a test compound in an animal model with an implanted tumor is to measure the size of the tumor before and after treatment. Traditionally, the size of implanted tumors has been measured with a slide caliper in two or three dimensions. The measure limited to two dimensions does not 35 accurately reflect the size of the tumor: therefore, it is usually converted into the corresponding volume by using a mathematical formula. However, the measurement of tumor size is very inaccurate. The therapeutic effects of a drug candidate can be better described as treatment-induced growth delay and specific growth delay. Another <br><br> 76 <br><br> important variable in the description of tumor growth is the tumor volume doubling time. Computer programs for the calculation and description of tumor growth are also available, such as the program reported by Rygaard and Spang-Thomsen. Proc. 6th Int. Workshop on Immune-Deficient Animals. Wu and Shengeds. (Basel, 1989), p. 301. It is noted, however, that necrosis and inflammatory responses following treatment may actually result in an 5 increase in tumor size, at least initially. Therefore, these changes need to be carefully monitored, by a combination of a morphometric method and flow cytometric analysis. <br><br> Further, recombinant (transgenic) animal models can be engineered by introducing the coding portion ofthe PRO genes identified herein into the genome of animals of interest, using standard techniques for producing transgenic animals. Animals that can serve as a target for transgenic manipulation include, without limitation, 10 mice, rats, rabbits, guinea pigs, sheep, goats, pigs, and non-human primates, e.g., baboons, chimpanzees and monkeys. Techniques known in the art to introduce a transgene into such animals include pronucleic m icroinjection (U.S. Patent No. 4,873,191); retrovirus-mediated gene transfer into germ lines (e.g., Van der Putten et al, Proc. ^ Natl. Acad. Sci. USA. 82: 6148-615 (1985)); gene targeting in embryonic stem cells (Thompson et al, Cell. 56: 313-321 (1989)): electroporation of embryos (Lo, Mol.Cell. Biol.. 3:1803-1814 (1983)); and sperm-mediated gene 15 transfer. Lavitrano et al. Cell. 57: 717-73 (1989). For a review, see for example, U.S. Patent No. 4,736,866. <br><br> For the purpose of the present invention, transgenic animals include those that carry the transgene only in part of their cells ("mosaic animals"). The transgene can be integrated either as a single transgene, or in concatamers, e.g., head-to-head or head-to-tail tandems. Selective introduction of a transgene into a particular cell type is also possible by following, for example, the technique of Lasko et al., Proc. Natl. Acad. Sci. USA. 89:6232-636 (1992). 20 The expression of the transgene in transgenic animals can be monitored by standard techniques. For example, <br><br> Southern blot analysis or PCR amplification can be used to verify the integration of the transgene. The level of mRNA expression can then be analyzed using techniques such as in situ hybridization. Northern blot analysis, PCR, or immunocytochemistry. The animals are further examined for signs of tumor or cancer development. <br><br> Alternatively, "knock-out" animals can be constructed that have a defective or altered gene encoding a PRO 25 polypeptide identified herein, as a result of homologous recombination between the endogenous gene encoding the ) PRO polypeptide and altered genomic DNA encoding the same polypeptide introduced into an embryonic cell of the animal. For example. cDNA encoding a particular PRO polypeptide can be used to clone genomic DNA encoding that polypeptide in accordance with established techniques. A portion of the genomic DNA encoding a particular PRO polypeptide can be deleted or replaced with another gene, such as a gene encoding a selectable 30 marker that can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector. See, e.g., Thomas and Capecchi, Cell. 51:503 (1987) for a description of homologous recombination vectors. The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected. See, e.g., Li et al., Cell. 69: 915 (1992). The selected cells are then injected into a blastocyst of an 35 animal (e.g., a mouse or rat) to form aggregation chimeras. See, e.g., Bradley, in Teratocarcinomasand Embryonic Stem Cells: A Practical Approach. E. J. Robertson, ed. (IRL: Oxford. 1987), pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term to create a <br><br> 77 <br><br> "knock-out" animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knockout animals can be characterized, for instance, by their ability to defend against certain pathological conditionsand by their development of pathological conditions due to absence of the PRO polypeptide. <br><br> 5 The efficacy of antibodies specifically binding the PRO polypeptides identified herein, and other drug candidates, can be tested also in the treatment of spontaneous animal tumors. A suitable target for such studies is the feline oral squamous cell carcinoma (SCC). Feline oral SCC is a highly invasive, malignant tumor that is the most common oral malignancy of cats, accounting for over 60% of the oral tumors reported in this species. It rarely metastasizes to distant sites, although this low incidence of metastasis may merely be a reflection of the short 10 survival times for cats with this tumor. These tumors are usually not amenable to surgery, primarily because of the anatomy of the feline oral cavity. At present, there is no effective treatment for this tumor. Prior to entry into the study, each cat undergoes complete clinical examination and biopsy, and is scanned by computed tomography (CT). Cats diagnosed with sublingual oral squamous cell tumors are excluded from the study. The tongue can become paralyzed as a result of such tumor, and even if the treatment kills the tumor, the animals may not be able to feed 15 themselves. Each cat is treated repeatedly, over a longer period of time. Photographs of the tumors will be taken daily during the treatment period, and at each subsequent recheck. After treatment, each cat undergoes another CT scan. CT scans and thoracic radiograms are evaluated every 8 weeks thereafter. The data are evaluated for differences in survival, response, and toxicity as compared to control groups. Positive response may require evidence of tumor regression, preferably with improvement of quality of life and/or increased life span. 20 In addition.other spontaneous animal tumors, such as fibrosarcoma, adenocarcinoma, lymphoma, chondroma, <br><br> or leiomyosarcoma of dogs, cats, and baboons can also be tested. Of these, mammary adenocarcinoma in dogs and cats is a preferred model as its appearance and behavior are very similar to those in humans. However, the use of this model is limited by the rare occurrence of this type of tumor in animals. <br><br> Other in vitro and in vivo cardiovascular, endothelial, and angiogenic tests known in the art are also suitable 25 herein. <br><br> ^ ii. Tissue Distribution <br><br> The results of the cardiovascular, endothelial, and angiogenic assays herein can be verified by further studies, such as by determining mRNA expression in various human tissues. <br><br> As noted before, gene amplification and/or gene expression in various tissues may be measured by 30 conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA. 77:5201 -5205 (1980)). dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. <br><br> 35 Gene expression in various tissues, alternatively, may be measured by immunological methods, such as immunohistochemical staining of tissue sections and assay of cell culture or body fluids, to quantitate directly the <br><br> 78 <br><br> expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native-sequence PRO polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to PRO DNA and encoding a specific antibody epitope. <br><br> 5 General techniques for generating antibodies, and special protocols for in situ hybridization are provided hereinbelow. <br><br> iii. Antibody Binding Studies <br><br> The results ofthe cardiovascular.endothelial, and angiogenic study can be further verified by antibody binding studies, in which the ability of anti-PRO antibodies to inhibit the effect ofthe PRO polypeptides on endothelial cells 10 or other cells used in the cardiovascular, endothelial, and angiogenic assays is tested. Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies, the preparation of which will be described hereinbelow. <br><br> Antibody binding studies may be carried out in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of 15 Techniques (CRC Press. Inc.. 1987). pp. 147-158. <br><br> Competitive binding assays rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody. The amount of target protein in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies preferably are insolubilized before or after the competition, so that 20 the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte that remain unbound. <br><br> Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the protein to be detected. In a sandwich assay, the test sample analyte is bound by a first antibody that is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming ^p^5 an insoluble three-part complex. See. e.g., US Pat No. 4.376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme. <br><br> For immunohistochemistry. the tissue sample may be fresh or frozen or may be embedded in paraffin and 30 fixed with a preservative such as formalin, for example. <br><br> iv. Cell-Based Tumor Assavs <br><br> Cell-based assays and animal models for cardiovascular, endothelial, and angiogenic disorders, such as tumors, can be used to verify the findings of a cardiovascular, endothelial, and angiogenic assay herein, and further to understand the relationship between the genes identified herein and the development and pathogenesis of 35 undesirable cardiovascular, endothelial, and angiogenic cell growth. The role of gene products identified herein <br><br> 79 <br><br> in the development and pathology of undesirable cardiovascular, endothelial, and angiogenic cell growth, e.g„ tumor cells, can be tested by using cells or cells lines that have been identified as being stimulated or inhibited by the PRO polypeptide herein. Such cells include, for example, those set forth in the Examples below. <br><br> In a different approach, cells of a cell type known to be involved in a particular cardiovascular, endothelial, 5 and angiogenic disorder are transfected with the cDNAs herein, and the ability of these cDNAs to induce excessive growth or inhibit growth is analyzed. If the cardiovascular, endothelial, and angiogenic disorder is cancer, suitable tumor cells include, for example, stable tumor cells lines such as the B104-1 -1 cell line (stable NIH-3T3 cell line transfected with the neu protooncogene) and nw-transfected NIH-3T3 cells, which can be transfected with the desired gene and monitored for tumorigenic growth. Such transfected cell lines can then be used to test the ability 10 of poly- or monoclonal antibodies or antibody compositions to inhibit tumorigenic cell growth by exerting cytostatic or cytotoxic activity on the growth of the transformed cells, or by mediating antibody-dependent cellular cytotoxicity (ADCC). Cells transfected with the coding sequences of the genes identified herein can further be used to identify drug candidates for the treatment of cardiovascular, endothelial, and angiogenic disorders such as cancer. <br><br> In addition, primary cultures derived from tumors in transgenic animals (as described above) can be used in 15 the cell-based assays herein, although stable cell lines are preferred. Techniques to derive continuous cell lines from transgenic animals are well known in the art. See, e.g., Small et al., Mol. Cell. Biol.. 5: 642-648 (1985). <br><br> v. Gene Therapy <br><br> The PRO polypeptide herein and polypeptidyl agonists and antagonists may be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as gene therapy. 20 There are two major approaches to getting the nucleic acid (optionally contained in a vector) into the patient's cells: in vivo and ex vivo. For in vh o delivery the nucleic acid is injected directly into the patient, usually at the sites where the PRO polypeptide is required, i.e., the site of synthesis of the PRO polypeptide, if known, and the site (e.g., wound) where biological activity of PRO polypeptide is needed. For ex vivo treatment, the patient's cells are removed, the nucleic acid is introduced into these isolated cells, and the modified cells are administered to the 25 patient either directly or, for example, encapsulated within porous membranes that are implanted into the patient (see, e.g., U.S. Pat. Nos. 4.892,538 and 5,283,187). There are a variety of techniques available for introducing nucleic acids into viable cells. The techniques vary depending upon whether the nucleic acid is transferred into cultured cells in vitro, or transferred in vivo in the cells of the intended host. Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, 30 transduction, cell fusion. DEAE-dextran, the calcium phosphate precipitation method, etc. Transduction involves the association of a replication-defective, recombinant viral (preferably retroviral) particle with a cellular receptor, followed by introduction of the nucleic acids contained by the particle into the cell. A commonly used vector for ex vivo delivery of the gene is a retrovirus. <br><br> The currently preferred in vivo nucleic acid transfer techniques include transfection with viral or non-viral 35 vectors (such as adenovirus, lentivirus. Herpes simplex I virus, or adeno-associated virus (AAV)) and lipid-based systems (useful lipids for iipid-mediated transfer of the gene are, for example, DOTMA, DOPE, and DC-Choi; see, <br><br> 80 <br><br> e.g., Tonkinson et al., Cancer Investigation. 14(1): 54-65 (1996)). The most preferred vectors for use in gene therapy are viruses, most preferably adenoviruses, AAV, lentiyiruses, or retroviruses. A viral vector such as a retroviral vector includes at least one transcriptional promoter/enhancer or locus-defining element(s), or other elements that control gene expression by other means such as alternate splicing, nuclear RNA export, or 5 post-translational modification of messenger. In addition, a viral vector such as a retroviral vector includes a nucleic acid molecule that, when transcribed in the presence of a gene encoding PRO polypeptide, is operably linked thereto and acts as a translation initiation sequence. Such vector constructs also include a packaging signal, <br><br> long terminal repeats (LTRs) or portions thereof, and positive and negative strand primer binding sites appropriate to the virus used (if these are not already present in the viral vector). In addition, such vector typically includes a 10 signal sequence for secretion of the PRO polypeptide from a host cell in which it is placed. Preferably the signal sequence for this purpose is a mammalian signal sequence, most preferably the native signal sequence for the PRO polypeptide. Optionally, the vector construct may also include a signal that directs polyadenylation, as well as one ^ or more restriction sites and a translation termination sequence. By way of example, such vectors will typically include a 5' LTR, a tRNA binding site, a packaging signal, an origin of second-strand DNA synthesis, and a 3' LTR 15 or a portion thereof. Other vectors can be used that are non-viral, such as cationic lipids, polylysine, and dendrimers. <br><br> In some situations, it is desirable to provide the nucleic acid source with an agent that targets the target cells, <br><br> such as an antibody specific for a cell-surface membrane protein or the target cell, a ligand for a receptor on the target cell. etc. Where liposomes are employed, proteins that bind to a cell-surface membrane protein associated 20 with endocytosis may be used for targeting and/or to facilitate uptake, e.g., capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins that undergo internalization in cycling, and proteins that target intracellular localization and enhance intracellular half-life. The technique of receptor-mediated endocytosis is described, for example, by Wu et al.. J. Biol. Chem.. 262:4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. <br><br> Sci. USA. 87:3410-3414(1990). For a review of the currently known gene marking and gene therapy protocols, 25 see, Anderson et al.. Science. 256: 808-813 (1992). See also WO 93/25673 and the references cited therein. ^ Suitable gene therapy and methods for making retroviral particles and structural proteins can be found in, e.g., <br><br> U.S. Pat. No. 5,681,746. <br><br> vi. Use of Gene as Diagnostic <br><br> This invention is also related to the use of the gene encoding the PRO 1313 polypeptide as a diagnostic. Detection <br><br> 30 of a mutated form of the PRO polypeptide will allow a diagnosis of a cardiovascular, endothelial, and angiogenic disease or a susceptibility to a cardiovascular, endothelial, and angiogenic disease, such as a tumor, since mutations in the PRO polypeptide may cause tumors. <br><br> Individuals carrying mutations in the genes encoding a human PRO polypeptide may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from 35 blood, urine, saliva, tissue biopsy, and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki et al., Nature. 324: 163-166 (1986)) priorloiu^ysisr Rl^p'rc", office"! <br><br> 1 tjr | <br><br> 81 ' 1 <br><br> iCESVEJ2- <br><br> or cDN A may also be used for the same purpose. As an example, PCR primers complementary to the nucleic acid encoding the PRO polypeptide can be used to identify and analyze PRO polypeptide mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA encoding the PRO 5 polypeptide, or alternatively, radiolabeled antisense DNA sequences encoding the PRO polypeptide. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures. <br><br> Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions 10 and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamidine gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures. See, e.g., Myers et at., Science. 230: 1242 (1985). <br><br> Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase 15 and SI protection or the chemical cleavage method, for example, Cotton et al, Proc. Natl. Acad. Sci. USA. 85: 4397-4401 (1985). <br><br> Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing, or the use of restriction enzymes, e.g., restriction fragment length polymorphisms (RFLP), and Southern blotting of genomic DNA. <br><br> 20 vii. Use to Detect PRO Polypeptide Levels <br><br> In addition to more conventional gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis. <br><br> Expression of nucleic acid encoding the PRO polypeptide may be linked to vascular disease or neovascularization associated with tumor formation. If the PRO polypeptide has a signal sequence and the mRNA 25 is highly expressed in endothelial cells and to a lesser extent in smooth muscle cells, this indicates that the PRO " polypeptide is present in serum. Accordingly, an anti-PRO polypeptide antibody could be used to diagnose vascular disease or neovascularization associated with tumor formation, since an altered level of this PRO polypeptide may be indicative of such disorders. <br><br> A competition assay may be employed wherein antibodies specific to the PRO polypeptide are attached to a 30 solid support and the labeled PRO polypeptide and a sample derived from the host are passed over the solid support and the amount of label detected attached to the solid support can be correlated to a quantity of PRO polypeptide in the sample. <br><br> viii. Chromosome Mapping <br><br> The sequences of the present invention are also valuable for chromosome identification. The sequence is 35 specifically targeted to and can hybridize with a particular location on an individual human chromosome. <br><br> 82 <br><br> Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease. <br><br> 5 Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis for the 3'- untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA. thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment. <br><br> 10 PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner. ^ Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, ^ prescreening with labeled flow-sorted chromosomes, and preselection by hybridization to construct chromosome-15 specific cDNA libraries. <br><br> Fluorescence in situ hybridization (FISH) of a cDN A clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 500 or 600 bases; however, clones larger than 2,000 bp have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. FISH requires use of the clones from which the gene 20 encoding the PRO polypeptide was derived, and the longer the better. For example, 2,000 bp is good. 4,000 bp is better, and more than 4,000 is probably not necessary to get good results a reasonable percentage of the time. For a review of this technique, see, Verma et al., Human Chromosomes: a Manual of Basic Techniques (Pergamon Press, New York, 1988). <br><br> Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence 25 on the chromosome can be correlated with genetic map data. Such data are found, for example, in V. McKusick, ^ Mendelian Inheritance in Man (available online through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region is then identified through linkage analysis (coinheritance of physically adjacent genes). <br><br> Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and 30 unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease. <br><br> With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. <br><br> (This assumes 1 megabase mapping resolution and one gene per 20 kb). <br><br> 35 ix. Screening Assays for Drug Candidates <br><br> This invention encompasses methods of screening compounds to identify those that mimic_ih^,PR^--B: <br><br> r^ELLECTUAl PROVtm Or K'L <br><br> 83 t 5 M 2°05 <br><br> polypeptide (agonists) or prevent the effect of the PRO 1313 polypeptide (antagonists). <br><br> Screening assays for antagonist drug candidates are designed to identify compounds that bind or complex with the PRO polypeptide encoded by the genes identified herein, or otherwise interfere with the interaction of the encoded polypeptides with other cellular proteins. Such screening assays will include assays amenable to high-throughput screening of chemical 5 libraries, making them particularly suitable for identifying small molecule drug candidates. <br><br> The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays, and cell-based assays, which are well characterized in the art. <br><br> All assays for antagonists are common in that they call for contacting the drug candidate with a PRO polypeptide encoded by a nucleic acid identified herein under conditions and for a time sufficient to allow these 10 two components to interact. <br><br> In binding assays, the interaction is binding and the complex formed can be isolated or detected in the reaction mixture. In a particular embodiment, the PRO polypeptide encoded by the gene identified herein or the drug candidate is immobilized on a solid phase, e.g., on a microtiter plate, by covalent or non-covalent attachments. Non-covalent attachment generally is accomplished by coating the solid surface with a solution of the PRO 15 polypeptide and drying. Alternatively, an immobilized antibody, e.g., a monoclonal antibody, specific for the PRO polypeptide to be immobilized can be used to anchor it to a solid surface. The assay is performed by adding the non-immobilized component, which may be labeled by a detectable label, to the immobilized component, e.g., the coated surface containing the anchored component. When the reaction is complete, the non-reacted components are removed, e.g., by washing, and complexes anchored on the solid surface are detected. When the originally non-20 immobilized component carries a detectable label, the detection of label immobilized on the surface indicates that complexing occurred. Where the originally non-immobilized component does not carry a label, complexing can be detected, for example, by using a labeled antibody specifically binding the immobilized complex. <br><br> If the candidate compound interacts with but does not bind to a particular PRO polypeptide encoded by a gene identified herein, its interaction with that polypeptide can be assayed by methods well known for detecting protein-25 protein interactions. Such assays include traditional approaches, such as, e.g., cross-linking, co-^ immunoprecipitation, and co-purification through gradients or chromatographic columns. In addition, protein-protein interactions can be monitored by using a yeast-based genetic system described by Fields and co-workers (Fields and Song, Nature (London). 340:245-246 (1989); Chien et al., Proc. Natl. Acad. Sci. USA. 88:9578-9582 (1991)) as disclosed by Chevray and Nathans, Proc. Natl. Acad. Sci. USA. 89: 5789-5793 (1991). Many 30 transcriptional activators, such as yeast GAL4, consist of two physically discrete modular domains, one acting as the DNA-binding domain, the other one functioning as the transcription-activation domain. The yeast expression system described in the foregoing publications (generally referred to as the "two-hybrid system") takes advantage of this property, and employs two hybrid proteins, one in which the target protein is fused to the DNA-binding domain of GAL4, and another, in which candidate activating proteins are fused to the activation domain. The 35 expression of a GAL 1 -lacZ reporter gene under control of a G AL4-activated promoter depends on reconstitution of GAL4 activity via protein-protein interaction. Colonies containing interacting polypeptides are detected with a chromogenic substrate for P-galactosidase. A complete kit (MATCHMAKER™) for identifying protein-protein o4 J lNTELLECTUAl ^nOPERT/ OFFICE <br><br> Of <br><br> interactionsbetweentwospecific proteins using the two-hybrid technique is commercially available from Clontech. This system can also be extended to map protein domains involved in specific protein interactions as well as to pinpoint amino acid residues that are crucial for these interactions. <br><br> Compounds that interfere with the interaction of a gene encoding a PRO polypeptide identified herein and 5 other intra- or extracellular components can be tested as follows: usually a reaction mixture is prepared containing the product of the gene and the intra- or extracellular component under conditions and for a time allowing for the interaction and binding of the two products. To test the ability of a candidate compound to inhibit binding, the reaction is run in the absence and in the presence of the test compound. In addition, a placebo may be added to a third reaction mixture, to serve as positive control. The binding (complex formation) between the test compound 10 and the intra- or extracellular component present in the mixture is monitored as described hereinabove. The formation of a complex in the control reaction(s) but not in the reaction mixture containing the test compound indicates that the test compound interferes with the interaction of the test compound and its reaction partner. <br><br> If the PRO polypeptide has the ability to stimulate the proliferation of endothelial cells in the presence of the co-mitogen ConA, then one example of a screening method takes advantage of this ability. Specifically, in the 15 proliferation assay, human umbilical vein endothelial cells are obtained and cultured in 96-well flat-bottomed culture plates (Costar, Cambridge, MA) and supplemented with a reaction mixture appropriate for facilitating proliferation of the cells, the mixture containing Con-A (Calbiochem, La Jolla, CA). Con-A and the compound to be screened are added and after incubation at 37°C, cultures are pulsed with 3'H-thymidine and harvested onto glass fiber filters (phD; Cambridge Technology, Watertown, MA). Mean 3'H- thymidine incorporation(cpm)oftriplicate 20 cultures is determined using a liquid scintillation counter (Beckman Instruments, Irvine, CA). Significant J"(H) thymidine incorporation indicates stimulation of endothelial cell proliferation. <br><br> To assay for antagonists, the assay described above is performed; however, in this assay the PRO polypeptide is added along with the compound to be screened and the ability of the compound to inhibit 3"(H)-thymidine incorporation in the presence of the PRO polypeptide indicates that the compound is an antagonist to the PRO 25 polypeptide. Alternatively, antagonists may be detected by combining the PRO polypeptide and a potential antagonist with membrane-bound PRO polypeptide receptors or recombinant receptors under appropriate conditions for a competitive inhibition assay. The PRO polypeptide can be labeled, such as by radioactivity, such that the number of PRO polypeptide molecules bound to the receptor can be used to determine the effectiveness ofthe potential antagonist. The gene encoding the receptor can be identified by numerous methods known to those 30 ofskill in the art, for example, ligand panning and FACS sorting. Coligan etal.. Current Protocols in Immun.. 1(2V. Chapter 5 (1991). Preferably* expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the PRO polypeptide and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the PRO polypeptide. Transfected cells that are grown on glass slides are exposed to the labeled PRO polypeptide. The PRO polypeptide can be labeled by a 35 variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase. Following fixation and incubation, the slides are subjected to autoradiographic analysis. Positive pools are identified and sub-pools are prepared and re-transfected using an interactive sub-pooling and re-screening process, eventually yielding <br><br> 85 <br><br> a single clone that encodes the putative receptor. <br><br> As an alternative approach for receptor identification, labeled PRO polypeptide can be photoaffinity-linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE and exposed to X-ray film. The labeled complex containing the receptor can be excised, resolved into 5 peptide fragments, and subjected to protein micro-sequencing. The amino acid sequence obtained from micro-sequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDN A library to identify the gene encoding the putative receptor. <br><br> In another assay for antagonists, mammalian cells or a membrane preparation expressing the receptor would be incubated with labeled PRO polypeptide in the presence of the candidate compound. The ability of the 10 compound to enhance or block this interaction could then be measured. <br><br> The compositions useful in the treatment of cardiovascular, endothelial, and angiogenic disorders include, without limitation, antibodies, small organic and inorganic molecules, peptides, phosphopeptides, antisense and ribozyme molecules, triple-helix molecules, etc., that inhibit the expression and/or activity of the target gene product. <br><br> 15 More specific examples of potential antagonists include an oligonucleotide that binds to the fusions of immunoglobulin with a PRO polypeptide, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, anti-idiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments. Alternatively, a potential antagonist may be a closely related protein, for example, a mutated form ofthe PRO 20 polypeptide that recognizes the receptor but imparts no effect, thereby competitively inhibiting the action of the PRO polypeptide. <br><br> Another potential PRO polypeptide antagonist or agonist is an antisense RNA or DNA construct prepared using antisense technology, where, e.g., an antisense RNA or DNA molecule acts to block directly the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation. Antisense technology can be used 25 to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5' coding portion of the polynucleotide sequence, which encodes the mature PRO polypeptides herein, is used to design an antisense R.NA oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region ofthe gene involved in transcription (triple helix - see, Lee et al., Nucl. Acids Res.. 6:3073 (1979); Cooney etal., 30 Science. 241: 456 (1988); Dervan et al.. Science. 251:1360 (1991)), thereby preventing transcription and the production of the PRO polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the PRO polypeptide (antisense - Okano, Neurochem.. 56:560 (1991): Oligodeoxvnucleotides as Antisense Inhibitors of Gene Expression (CRC Press: Boca Raton, FL, 1988). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be 35 expressed in vivo to inhibit production of the PRO polypeptide. When antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g., between about -10 and +10 positions of the target gene nucleotide sequence, are preferred. <br><br> 86 <br><br> WO 00/32221 PCT/US99/28313 <br><br> Antisense RMA or DNA molecules are generally at least about 5 bases in length, about 10 bases in length, about 15 bases in length, about 20 bases in length, about 25 bases in length, about 30 bases in length, about 35 bases in length, about 40 bases in length, about 45 bases in length, about 50 bases in length, about 55 bases in length, about 60 bases in length, about 65 bases in length, about 70 bases in length, about 75 bases in length, about 80 bases 5 in length, about 85 bases in length, about 90 bases in length, about 95 bases in length, about 100 bases in length, or more. <br><br> Potential antagonists include small molecules that bind to the active site, the receptor binding site, or growth factor or other relevant binding site of the PRO polypeptide, thereby blocking the normal biological activity of the PRO polypeptide. Examples of small molecules include, but are not limited to, small peptides or peptide-like 10 molecules, preferably soluble peptides, and synthetic non-peptidyl organic or inorganic compounds. <br><br> Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by sequence-specific hybridization to the complementary target RNA, followed by endonucleoiytic cleavage. Specific ribozyme cleavage sites within a potential RNA target can be identified by known techniques. For further details see, e.g., Rossi, Current Biology. 4: 469-471 (1994), and PCT publication No. WO 97/33551 (published 15 September 18,1997). <br><br> Nucleic acid molecules in triple-helix formation used to inhibit transcription should be single-stranded and composed of deoxynucleotides. The base composition of these oligonucleotides is designed such that it promotes triple-helix formation via Hoogsteen base-pairing rules, which generally require sizeabJe stretches of purines or pyrimidines on one strand of a duplex. For further details see, e.g., PCT publication No. WO 97/33551, supra. 20 These small molecules can be identified by any one or more of the screening assays discussed hereinabove and/or by any other screening techniques well known for those skilled in the art. <br><br> x. Types of Cardiovascular. Endothelial, and Angiogenic Disorders to be Treated <br><br> The PRO polypeptides, or agonists or antagonists thereto, that have activity in the cardiovascular, angiogenic, and endothelial assays described herein, and/or whose gene product has been found to be localized to the cardiovascular system, are likely to have therapeutic uses in a variety of cardiovascular, endothelial, and angiogenic disorders, including systemic disorders that affect vessels, such as diabetes mellitus. Their therapeutic utility could include diseases of the arteries, capillaries, veins, and/or lymphatics. Examples of treatments hereunder include treating muscle wasting disease, treating osteoporosis, aiding in implant fixation to stimulate the growth of cells around the implant and therefore facilitate its attachment to its intended site, increasing IGF stability in tissues or 30 in serum, if applicable, and increasing binding to the IGF receptor (since IGF has been shown in vitro to enhance human marrow erythroid and granulocytic progenitor cell growth). <br><br> The PRO polypeptides or agonists or antagonists thereto may also be employed to stimulate erythropoiesis or granulopoiesis, to stimulate wound healing or tissue regeneration and associated therapies concerned with re-growth of tissue, such as connective tissue, skin, bone, cartilage, muscle, lung, or kidney, to promote angiogenesis, 35 to stimulate or inhibit migration of endothelial cells, and to proliferate the growth of vascular smooth muscle and endothelial cell production. The increase in angiogenesis mediated by the PRO polypeptide or antagonist would <br><br> 87 <br><br> WO 00/32221 PCT/US99/28313 - <br><br> be beneficial to ischemic tissues and to collateral coronary development in the heart subsequent to coronary stenosis. Antagonists are used to inhibit the action of such polypeptides, for example, to limit the production of excess connective tissue during wound healing or pulmonary fibrosis if the PRO polypeptide promotes such production. This would include treatment of acute myocardial infarction and heart failure. <br><br> 5 Described herein is the treatment of cardiac hypertrophy, regardless of the underlying cause, by administering a therapeutically effective dose of the PRO polypeptide, or agonist or antagonist thereto. If the objective is the treatment of human patients, the PRO polypeptide preferably is recombinant human PRO polypeptide (rhPRO polypeptide). The treatment for cardiac hypertrophy can be performed at any of its various stages, which may result from a variety of diverse pathologic conditions, including myocardial infarction, 10 hypertension, hypertrophic cardiomyopathy, and valvular regurgitation. The treatment extends to all stages of the progression of cardiac hypertrophy, with or without structural damage of the heart muscle, regardless of the underlying cardiac disorder. <br><br> ^ The decision of whether to use the molecule itself or an agonist thereof for any particular indication, as opposed to an antagonist to the molecule, would depend mainly on whether the molecule herein promotes 15 cardiovascularization, genesis of endothelial cells, or angiogenesis or inhibits these conditions. For example, if the molecule promotes angiogenesis, an antagonist thereof would be useful for treatment of disorders where it is desired to limit or prevent angiogenesis. Examples of such disorders include vascular tumors such as haemangioma, tumor angiogenesis, neovascularization in the retina, choroid, or cornea, associated with diabetic retinopathy or premature infant retinopathy or macular degeneration and proliferative vitreoretinopathy, rheumatoid arthritis, Crohn's 20 disease, atherosclerosis, ovarian hyperstimulation, psoriasis, endometriosis associated with neovascularization, restenosis subsequent to balloon angioplasty, scar tissue overproduction, for example, that seen in a keloid that forms after surgery, fibrosis after myocardial infarction, or fibrotic lesions associated with pulmonary fibrosis. <br><br> If, however, the molecule inhibits angiogenesis, it would be expected to be used directly for treatment of the above conditions. <br><br> 25 On the other hand, if the molecule stimulates angiogenesis it would be used itself (or an agonist thereoO for <br><br> ^ indications where angiogenesis is desired such as peripheral vascular disease, hypertension, inflammatory vasculitides, Reynaud's disease and Reynaud's phenomenon, aneurysms, arterial restenosis, thrombophlebitis, lymphangitis, lymphedema, wound healing and tissue repair, ischemia reperfusion injury, angina, myocardial infarctions such as acute myocardial infarctions, chronic heart conditions, heart failure such as congestive heart 30 failure, and osteoporosis. <br><br> If, however, the molecule inhibits angiogenesis, an antagonist thereof would be used for treatment of those conditions where angiogenesis is desired. = <br><br> Specific types of diseases are described below, where the PRO polypeptide herein or antagonists thereof may serve as useful for vascular-related drug targeting or as therapeutic targets for the treatment or prevention of the 35 disorders. Atherosclerosis is a disease characterized by accumulation of plaques of intimal thickening in arteries, due to accumulation of lipids, proliferation of smooth muscle cells, and formation of fibrous tissue within the arterial wall. The disease can affect large, medium, and small arteries in any organ. Changes in endothelial and <br><br> 88 <br><br> vascular smooth muscle cell function are known to play an important role in modulating the accumulation and regression of these plaques. <br><br> Hypertension is characterized by raised vascular pressure in the systemic arterial, pulmonary arterial, or portal venous systems. Elevated pressure may result from or result in impaired endothelial function and/or vascular 5 disease. <br><br> Inflammatory vasculitides include giant cell arteritis, Takayasu's arteritis, polyarteritis nodosa (including the microangiopathic form), Kawasaki's disease, microscopic polyangiitis, Wegener's granulomatosis, and a variety of infectious-related vascular disorders (including Henoch-Schonlein prupura). Altered endothelial cell function has been shown to be important in these diseases. <br><br> 10 Reynaud's disease and Reynaud's phenomenon are characterized by intermittent abnormal impairment of the circulation through the extremities on exposure to cold. Altered endothelial cell function has been shown to be important in this disease. <br><br> Aneurysms are saccular or fusiform dilatations of the arterial or venous tree that are associated with altered endothelial cell and/or vascular smooth muscle cells. <br><br> 15 Arterial restenosis (restenosis of the arterial wall) may occur following angioplasty as a result of alteration in the function and proliferation of endothelial and vascular smooth muscle cells. <br><br> Thrombophlebitis and lymphangitis are inflammatory disorders of veins and lymphatics, respectively, that may result from, and/or in, altered endothelial cell function. Similarly, lymphedema is a condition involving impaired lymphatic vessels resulting from endothelial cell function. <br><br> 20 The family of benign and malignant vascular tumors are characterized by abnormal proliferation and growth of cellular elements of the vascular system. For example, lymphangiomas are benign tumors of the lymphatic system that are congenital, often cystic, malformations of the lymphatics that usually occur in newborns. Cystic tumors tend to grow into the adjacent tissue. Cystic tumors usually occur in the cervical and axillary region. They can also occur in the soft tissue of the extremities. The main symptoms are dilated, sometimes reticular, structured 25 lymphatics and lymphocysts surrounded by connective tissue. Lymphangiomas are assumed to be caused by ^ improperly connected embryonic lymphatics or their deficiency. The result is impaired local lymph drainage. Griener et al.. Lvmphologv. 4: 140-144(1971). <br><br> Another use for the PRO polypeptides herein or antagonists thereto is in the prevention of tumor angiogenesis, which involves vascularization of a tumor to enable it to growth and/or metastasize. This process is dependent on 30 the growth of new blood vessels. Examples of neoplasms and related conditions that involve tumor angiogenesis include breast carcinomas, lung carcinomas, gastric carcinomas, esophageal carcinomas, colorectal carcinomas, liver carcinomas, ovarian carcinomas, thecomas, arrhenoblastomas, cervical carcinomas, endometrial carcinoma, endometrial hyperplasia, endometriosis, fibrosarcomas, choriocarcinoma, head and neck cancer, nasopharyngeal carcinoma, laryngeal carcinomas, hepatoblastoma, Kaposi's sarcoma, melanoma, skin carcinomas, hemangioma, 35 cavernous hemangioma, hemangioblastoma, pancreas carcinomas, retinoblastoma, astrocytoma, glioblastoma, Schwannoma, oligodendroglioma, medulloblastoma, neuroblastomas, rhabdomyosarcoma, osteogenic sarcoma, leiomyosarcomas, urinary tract carcinomas, thyroid carcinomas, Wilm's tumor, renal cell carcinoma, prostate <br><br> 89 <br><br> carcinoma, abnormal vascuiarproliferationassociatedwith phakomatoses, edema (such as that associated with brain tumors), and Meigs' syndrome. <br><br> Age-related macular degeneration (AMD) is a leading cause of severe visual loss in the elderly population. The exudative form of AMD is characterized by choroidal neovascularization and retinal pigment epithelial cell 5 detachment. Because choroidal neovascularization is associated with a dramatic worsening in prognosis, the PRO polypeptides or antagonist thereto is expected to be useful in reducing the severity of AMD. <br><br> Healing of trauma such as wound healing and tissue repair is also a targeted use for the PRO polypeptides herein or their antagonists. Formation and regression of new blood vessels is essential for tissue healing and repair. This category includes bone, cartilage, tendon, ligament, and/or nerve tissue growth or regeneration, as well as 10 wound healing and tissue repair and replacement, and in the treatment of burns, incisions, and ulcers. A PRO polypeptide or antagonist thereof that induces cartilage and/or bone growth in circumstances where bone is not normally formed has application in the healing of bone fractures and cartilage damage or defects in humans and other animals. Such a preparation employing a PRO polypeptide or antagonist thereof may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints. De novo bone 15 formation induced by an osteogenic agent contributes to the repair of congenital, trauma-induced, or oncologic, resection-induced craniofacial defects, and also is useful in cosmetic plastic surgery. <br><br> PRO polypeptides or antagonists thereto may also be useful to promote better or faster closure of non-healing wounds, including without limitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like. <br><br> 20 It is expected that a PRO polypeptide or antagonist thereto may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, liver, intestine, kidney, skin, or endothelium), muscle (smooth, skeletal, or cardiac), and vascular (including vascular endothelium) tissue, or for promoting the growth of cells comprising such tissues. Part of the desired effects may be by inhibition or modulation of fibrotic scarring to allow normal tissue to regenerate. <br><br> 25 A PRO polypeptide herein or antagonist thereto may also be useful for gut protection or regeneration and treatment of lung or liver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage. Also, the PRO polypeptide or antagonist thereto may be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or cells, or for inhibiting the growth of tissues described above. <br><br> 30 A PRO polypeptide or antagonist thereto may also be used in the treatment of periodontal diseases and in other tooth-repair processes. Such agents may provide an environment to attract bone-forming cells, stimulate growth of bone-forming cells, or induce differentiation of progenitors of bone-forming cells. A PRO polypeptide herein or an antagonist thereto may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and or cartilage repair or by blocking inflammation or processes of tissue destruction 35 (collagenase activity, osteoclast activity, etc.) mediated by inflammatory processes, since blood vessels play an important role in the regulation of bone turnover and growth. <br><br> Another category of tissue regeneration activity that may be attributable to the PRO polypeptide herein or <br><br> 90 <br><br> antagonist thereto is tendon/ligament formation. A protein that induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normally formed has application in the healing of tendon or ligament tears, deformities, and other tendon or ligament defects in humans and other animals. Such a preparation may have prophylactic use in preventing damage to tendon or ligament tissue, as well as use in the improved 5 fixation of tendon or ligament to bone or other tissues, and in repairing defects to tendon or ligament tissue. De novo tendon/ligament-like tissue formation induced by a composition of the PRO polypeptide herein or antagonist thereto contributes to the repair of congenital, trauma-induced, or other tendon or ligament defects of other origin, <br><br> and is also useful in cosmetic plastic surgery for attachment or repair of tendons or ligaments. The compositions herein may provide an environment to attract tendon- or ligament-forming cells, stimulate growth of tendon- or 10 ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/ligament cells or progenitors ex vivo for return in vivo to effect tissue repair. The compositions herein may also be useful in the treatment of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects. The | compositions may also include an appropriate matrix and/or sequestering agent as a carrier as is well known in the art. <br><br> 15 The PRO polypeptide or its antagonist may also be useful for proliferation of neural cells and for regeneration of nerve and brain tissue, i.e., for the treatment of central and peripheral nervous system disease and neuropathies, <br><br> as well as mechanical and traumatic disorders, that involve degeneration, death, or trauma to neural cells or nerve tissue. More specifically, a PRO polypeptide or its antagonist may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and 20 central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington'sdisease,amyotrophiclateral sclerosis, and Shy-Drager syndrome. Further conditions that may be treated as described herein include mechanical arid traumatic disorders, such as spinal cord disorders, head trauma, and cerebrovascular diseases such as stroke. Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a PRO polypeptide herein or antagonist thereto. <br><br> 25 Ischemia-reperfusion injury is another indication. Endothelial cell dysfunction may be important in both the <br><br> ^ initiation of. and in regulation of the sequelae of events that occur following ischemia-reperfusion injury. <br><br> Rheumatoid arthritis is a further indication. Blood vessel growth and targeting of inflammatory cells through the vasculature is an important component in the pathogenesis of rheumatoid and sero-negative forms of arthritis. <br><br> PRO polypeptide or its antagonist may also be administered prophylactically to patients with cardiac 30 hypertrophy, to prevent the progression of the condition, and avoid sudden death, including death of asymptomatic patients. Such preventative therapy is particularly warranted in the case of patients diagnosed with massive left ventricular cardiac hypertrophy (a maximal wall thickness of 35 mm or more in adults, or a comparable value in children), or in instances when the hemodynamic burden on the heart is particularly strong. <br><br> PRO polypeptide or its antagonist may also be useful in the management of atrial fibrillation, which develops 35 in a substantial portion of patients diagnosed with hypertrophic cardiomyopathy. <br><br> Further indications include angina, myocardial infarctions such as acute myocardial infarctions, and heart failure such as congestive heart failure. Additional non-neoplastic conditions include psoriasis, diabetic and other <br><br> 91 intellectual property office of n.z. <br><br> 15 JUN 200S pcrpiv/pn <br><br> WO 00/32221 PCT/US99/28313 - <br><br> proliferative retinopathies including retinopathy of prematurity, retrolental fibroplasia, neovascular glaucoma, thyroid hyperplasias (including Grave's disease), corneal and other tissue transplantation, chronic inflammation, lung inflammation, nephrotic syndrome, preeclampsia, ascites, pericardial effusion (such as that associated with pericarditis), and pleural effusion. <br><br> 5 In view of the above, the PRO polypeptides or agonists or antagonists thereof described herein, which are shown to alter or impact endothelial cell function, proliferation, and/or form, are likely to play an important role in the etiology and pathogenesis of many or all of the disorders noted above, and as such can serve as therapeutic targets to augment or inhibit these processes or for vascular-related drug targeting in these disorders. <br><br> xi. Administration Protocols. Schedules. Doses, and Formulations 10 The molecules herein and agonists and antagonists thereto are pharmaceuticaliy useful as a prophylactic and therapeutic agent for various disorders and diseases as set forth above. <br><br> | Therapeutic compositions of the PRO polypeptides or agonists or antagonists are prepared for storage by mixing the desired molecule having the appropriate degree of purity with optional pharmaceuticaliy acceptable carriers, excipients, or stabilizers (Remington's Pharmaceutical Sciences. 16th edition, Osol, A. ed. (1980)), in the 15 form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such asoctadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; 20 and m-cresol); low molecular weight (less than about 10 residues) polypeptides: proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, giutamine, asparagine, histidine. arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g.. Zn-protein complexes); )25 and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). <br><br> Additional examples of such carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, 30 magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, and polyethylene glycol. Carriers for topical or gel-based forms of antagonist include polysaccharides such as sodium carboxymethylcellulose or methylcellulose, polyvinylpyrrolidone, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wood wax alcohols. For all administrations, conventional depot forms are suitably used. Such forms include, for example, m icrocapsules, nano-capsules, liposomes, plasters, inhalation forms, nose sprays, 35 sublingual tablets, and sustained-release preparations. The PRO polypeptides or agonists or antagonists will typically be formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml. <br><br> 92 <br><br> WO 00/32221 PCTAJS99/28313 <br><br> Another formulation comprises incorporating a PRO polypeptide or antagonist thereof into formed articles. Such articles can be used in modulating endothelial cell growth and angiogenesis. In addition, tumor invasion and metastasis may be modulated with these articles. <br><br> The PRO polypeptide or antagonist to be used for in vivo administration must be sterile. This is readily 5 accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution. The PRO polypeptide ordinarily will be stored in lyophilized form or in solution if administered systemically. If in lyophilized form, the PRO polypeptide or antagonist thereto is typically formulated in combination with other ingredients for reconstitution with an appropriate diluent at the time for use. An example of a liquid formulation of the PRO polypeptide or antagonist is a sterile, clear, colorless unpreserved solution filled 10 in a single-dose vial for subcutaneous injection. Preserved pharmaceutical compositions suitable for repeated use may contain, for example, depending mainly on the indication and type of polypeptide: <br><br> a) a PRO polypeptide or agonist or antagonist thereto; <br><br> | b) a buffer capable of maintaining the pH in a range of maximum stability of the polypeptide or other molecule in solution, preferably about 4-8; <br><br> 15 c) a detergent/surfactant primarily to stabilize the polypeptide or molecule against agitation-induced aggregation; <br><br> d) an isotonifier; <br><br> e) a preservative selected from the group of phenol, benzyl alcohol and a benzethonium haiide, e.g., chloride; <br><br> and <br><br> 20 f) water. <br><br> If the detergent employed is non-ionic, it may, for example, be polysorbates (e.g., POLYSORBATE™ (TWEEN™) 20, 80, etc.) or poloxamers (e.g., POLOXAMER™ 188). The use of non-ionic surfactants permits the formulation to be exposed to shear surface stresses without causing denaturation ofthe polypeptide. Further, such surfactant-containing formulations may be employed in aerosol devices such as those used in a pulmonary 25 dosing, and needleless jet injector guns (see, e.g., EP 257,956). <br><br> ^ An isotonifier may be present to ensure isotonicity of a liquid composition of the PRO polypeptide or antagonist thereto, and includes polyhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, ervthritol, arabitol, xylitol, sorbitol, and mannitol. These sugar alcohols can be used alone or in combination. Alternatively, sodium chloride or other appropriate inorganic salts may be used to render the 30 solutions isotonic. <br><br> The buffer may. for example, be an acetate, citrate, succinate, or phosphate buffer depending on the pH desired. The pH of one type of liquid formulation of this invention is buffered in the range of about 4 to 8, preferably about physiological pH. <br><br> The preservatives phenol, benzyl alcohol and benzethonium halides, e.g., chloride, are known antimicrobial 35 agents that may be employed. <br><br> Therapeutic PRO polypeptide compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. The <br><br> 93 <br><br> WO 00/32221 PCT/US99/28313 <br><br> formulations are preferably administered as repeated intravenous (i.v.), subcutaneous (s.c.), or intramuscular (i.m.) injections, or as aerosol formulations suitable for intranasal or intrapulmonary delivery (for intrapulmonary delivery see, e.g., EP 257,956). <br><br> PRO polypeptides can also be administered in the form of sustained-released preparations. Suitable examples 5 of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the protein, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate) as described by Langer et al. J. Biomed. Mater. Res.. ]5:167-277 (1981) and Langer, Chem. Tech.. 12:98-105 (1982)or poly(vinylalcohol)), polylactides (U.S. Patent No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate 10 (Sidman et al, Biopolvmers. 22: 547-556 (1983)), non-degradable ethylene-vinyl acetate (Langer et al, supra), degradable lactic acid-glycolic acid copolymers such as the Lupron Depot™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid (EP 133,988). <br><br> While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated proteins remain in 15 the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37°C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for protein stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate 20 additives, and developing specific polymer matrix compositions. <br><br> Sustained-release PRO polypeptide compositions also include liposomally entrapped PRO polypeptides. Liposomes containing the PRO polypeptide are prepared by methods known perse: DE 3,218,121; Epstein et al., Proc. Natl. Acad. Sci. USA. 82: 3688-3692 (1985); Hwang et al, Proc. Natl. Acad. Sci. USA. 77: 4030-4034 (1980); EP 52,322; EP 36.676; EP 88,046; EP 143,949; EP 142,641; Japanese patent application 83-118008; U.S. 25 Patent Nos. 4,485,045 and 4,544,545; and EP 102,324. Ordinarily the liposomes are of the small (about 200-800 angstroms) unilamellar type in which the lipid content is greater than about 30 mol. % cholesterol, the selected proportion being adjusted for the optimal therapy. <br><br> The therapeutically effective dose of a PRO polypeptide or antagonist thereto will, of course, vary depending on such factors as the pathological condition to be treated (including prevention), the method of administration, the 30 type of compound being used for treatment, any co-therapy involved, the patient's age, weight, general medical condition, medical history, etc., and its determination is well within the skill of a practicing physician. Accordingly, it will be necessary for the therapist to titerthe dosage and modify the route of administration as required to obtain the maximal therapeutic effect. If the PRO polypeptide has a narrow host range, for the treatment of human patients formulations comprising the human PRO polypeptide, more preferably the native-sequence human PRO 35 polypeptide, are preferred. The clinician will administer the PRO polypeptide until a dosage is reached that achieves the desired effect for treatment of the condition in question. For example, if the objective is the treatment of CHF, the amount would be one that inhibits the progressive cardiac hypertrophy associated with this condition. <br><br> 94 <br><br> The progress of this therapy is easily monitored by echo cardiography. Similarly, in patients with hypertrophic cardiomyopathy, the PRO polypeptide can be administered on an empirical basis. <br><br> With the above guidelines, the effective dose generally is within the range of from about 0.001 to about 1.0 mg/kg, more preferably about 0.01-1.0 mg/kg, most preferably about 0.01-0.1 mg/kg. <br><br> 5 For non-oral use in treating human adult hypertension, it is advantageous to administer the PRO polypeptide in the form of an injection at about 0.01 to 50 mg, preferably about 0.05 to 20 mg. most preferably I to 20 mg, per kg body weight, I to 3 times daily by intravenous injection. For oral administration, a molecule based on the PRO polypeptide is preferably administered at about 5 mg to 1 g, preferably about 10 to 100 mg, per kg body weight, 1 to 3 times daily. It should be appreciated that endotoxin contamination should be kept minimally at a safe level, 10 for example, less than 0.5 ng/mg protein. Moreover, for human administration, the formulations preferably meet sterility, pyrogenicity, general safety, and purity as required by FDA Office and Biologies standards. <br><br> The dosage regimen of a pharmaceutical composition containing a PRO polypeptide to be used in tissue regeneration will be determined by the attending physician considering various factors that modify the action of the polypeptides, e.g., amount of tissue weight desired to be formed, the site of damage, the condition of the 15 damaged tissue, the size of a wound, type of damaged tissue (e.g., bone), the patient's age, sex, and diet, the severity of any infection, time of administration, and other clinical factors. The dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition. For example, the addition of other known growth factors, such as 1GF-1, to the final composition may also affect the dosage. Progress can be monitored by periodic assessment of tissue/bone growth and/or repair, for example, X-rays, 20 histomorphometric determinations, and tetracycline labeling. <br><br> The route of PRO polypeptide or antagonist or agonist administration is in accord with known methods, e.g., by injection or infusion by intravenous, intramuscular, intracerebral, intraperitoneal, intracerobrospinal, subcutaneous, intraocular, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes, or by sustained-release systems as noted below. The PRO polypeptide or antagonists thereof also are suitably 25 administered by intratumoral, peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects. The intraperitoneal route is expected to be particularly useful, for example, in the treatment of ovarian tumors. <br><br> If a peptide or small molecule is employed as an antagonist or agonist, it is preferably administered orally or non-orally in the form of a liquid or solid to mammals. <br><br> 30 Examples of pharmacologically acceptable salts of molecules that form salts and are useful hereunder include alkali metal salts (e.g., sodium salt, potassium salt), alkaline earth metal salts (e.g., calcium salt, magnesium salt), ammonium salts, organic base salts (e.g., pyridine salt, triethylamine salt), inorganic acid salts (e.g., hydrochloride, sulfate, nitrate), and salts of organic acid (e.g., acetate, oxalate, p-toluenesulfonate). <br><br> For compositions herein that are useful for bone, cartilage, tendon, or ligament regeneration, the therapeutic 35 method includes administering the composition topically, systemically, or locally as an implant or device. When administered, the therapeutic composition for use is in a pyrogen-free, physiologically acceptable form. Further, the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, <br><br> 95 <br><br> cartilage, or tissue damage. Topical adm inistration may be suitable for wound healing and tissue repair. Preferably, for bone and/or cartilage formation, the composition would include a matrix capable of delivering the protein-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and preferably capable of being resorbed into the body. Such matrices may be formed of materials 5 presently in use for other implanted medical applications. <br><br> The choice of matrix material is based on biocompatibility, biodegradability, mechanical properties, cosmetic appearance, and interface properties. The particular application of the compositions will define the appropriate formulation. Potential matrices for the compositions may be biodegradable and chemically defined calcium sulfate, tricalcium phosphate, hydroxyapatite, polylactic acid, polyglycolic acid, and polyanhydrides. Other potential 10 materials are biodegradable and biologically well-defined, such as bone or dermal collagen. Further matrices are comprised of pure proteins or extracellular matrix components. Other potential matrices are nonbiodegradable and chemically defined, such as sintered hydroxyapatite, bioglass, aluminates, or other ceramics. Matrices may be comprised of combinations of any of the above-mentioned types of material, such as polylactic acid and I hydroxyapatite or collagen and tricalcium phosphate. The bioceramics may be altered in composition, such as in 15 calcium-aluminate-phosphate and processing to alter pore size, particle size, particle shape, and biodegradability. <br><br> One specific embodiment is a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns. In some applications, it will be useful to utilize a sequestering agent, such as carboxymethyl cellulose or autologous blood clot, to prevent the polypeptide compositions from disassociating from the matrix. <br><br> 20 One suitable family of sequestering agents is cellulosic materials such as alkylcelluloses (including hydroxyalkylcelluloses).includingmethylcellulose,ethylcellulose,hydoxyethylcellulose,hydroxypropylcellulose, hydroxypropylmethylcellulose, and carboxymethylcellulose, one preferred being cationic salts of carboxymethylcellulose (CMC). Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer, and poly(vinyl alcohol). The amount of 25 sequestering agent useful herein is 0.5-20 wt%, preferably 1-10 wt%, based on total formulation weight, which represents the amount necessary to prevent desorption ofthe polypeptide (or its antagonist) from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the polypeptide (or its antagonist) the opportunity to assist the osteogenic activity of the progenitor cells. <br><br> 30 xii. Combination Therapies <br><br> The effectiveness of the PRO polypeptide or an agonist or antagonist thereof in preventing or treating the disorder in question may be improved by administering the active agent serially or in combination with another agent that is effective for those purposes, either in the same composition or as separate compositions. <br><br> For example, for treatment of cardiac hypertrophy, PRO polypeptide therapy can be combined with the 35 administration of inhibitors of known cardiac myocyte hypertrophy factors, e.g., inhibitors of a-adrenergic agonists such as phenylephrine: endothelin-1 inhibitors such as BOSENTAN™ and MOXONODIN™; inhibitors to CT-1 <br><br> 96 <br><br> WO 00/32221 PCT/US99/28313 . <br><br> (US Pat. No. 5,679,545); inhibitors to LIF; ACE inhibitors; des-aspartate-angiotensin I inhibitors (U.S. Pat. No. 5,773,415), and angiotensin 11 inhibitors. <br><br> For treatment of cardiac hypertrophy associated with hypertension, a PRO polypeptide can be administered in combination with P-adrenergic receptor blocking agents, e.g., propranolol, timolol, tertaiolol, carteolol, nadolol, 5 betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, or carvedilol; ACE inhibitors, e.g., quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, or lisinopril; diuretics, e.g., chlorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, or indapamide; and/or calcium channel blockers, e.g., diltiazem, nifedipine, verapamil, or nicardipine. Pharmaceutical compositions comprising the therapeutic agents identified herein by their generic names are commercially available, and are to 10 be administered following the manufacturers' instructions for dosage, administration, adverse effects, contraindications, etc. See. e.g.. Physicians' Desk Reference (Medical Economics Data Production Co.: Montvale, N.J., 1997), 51th Edition. <br><br> Preferred candidates for combination therapy in the treatment of hypertrophic cardiomyopathy are P-adrenergic-blocking drugs (e.g.. propranolol, timolol, tertaiolol, carteolol, nadolol, betaxolol, penbutolol, 15 acetobutolol, atenolol, metoprolol. or carvedilol), verapamil, difedipine, or diltiazem. Treatment of hypertrophy associated with high blood pressure may require the use of antihypertensive drug therapy, using calcium channel blockers, e.g., diltiazem. nifedipine, verapamil, or nicardipine; P-adrenergic blocking agents; diuretics, e.g., chlorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, or indapamide; and/or ACE-inhibitors, e.g., quinapril, captopril, enalapril, ramipril, benazepril, 20 fosinopril, or lisinopril. <br><br> For other indications, PRO polypeptides or their antagonists may be combined with other agents beneficial to the treatment of the bone and/or cartilage defect, wound, or tissue in question. These agents include various growth factors such as EGF, PDGF, TGF-a or TGF-P, IGF, FGF, and CTGF. <br><br> In addition, PRO polypeptides or their antagonists used to treat cancer may be combined with cytotoxic, 25 chemotherapeutic, or growth-inhibitory agents as identified above. Also, for cancertreatment, the PRO polypeptide or antagonist thereof is suitably administered serially or in combination with radiological treatments, whether involving irradiation or administration of radioactive substances. <br><br> The effective amounts of the therapeutic agents administered in combination with the PRO polypeptide or antagonist thereof will be at the physician's or veterinarian's discretion. Dosage administration and adjustment is 30 done to achieve maximal management ofthe conditions to be treated. For example, for treating hypertension, these amounts ideally take into account use of diuretics or digitalis, and conditions such as hyper- or hypotension, renal impairment, etc. The dose will additionally depend on such factors as the type of the therapeutic agent to be used and the specific patient being treated. Typically, the amount employed will be the same dose as that used, if the given therapeutic agent is administered without the PRO polypeptide. <br><br> 35 xiii. Articles of Manufacture <br><br> An article of manufacture such as a kit containing a PRO polypeptide or antagonists thereof useful for the <br><br> 97 <br><br> WO 00/32221 PCT/US99/28313 <br><br> diagnosis or treatment of the disorders described above comprises at least a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition that is effective for diagnosing or treating the condition and may have a sterile access port (for example, the container may be an intravenous 5 solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agent in the composition is the PRO polypeptide or an agonist or antagonist thereto. The label on, or associated with, the container indicates that the composition is used for diagnosing or treating the condition of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, and dextrose solution. It may further include other materials desirable 10 from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. The article of manufacture may also comprise a second or third container with another active agent as described above. <br><br> ^ E. Antibodies <br><br> Some of the most promising drug candidates according to the present invention are antibodies and antibody 15 fragments that may inhibit the production or the gene product of the genes identified herein and/or reduce the activity of the gene products. <br><br> i. Polyclonal Antibodies <br><br> Methods of preparing polyclonal antibodies are known to the skilled artisan. Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. 20 Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. The immunizing agent may include the PRO polypeptide or a fusion protein thereof, it may be useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include, but are not limited to, keyhole limpet hemocyanin, ^ serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. Examples of adjuvants that may be employed 25 include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A or synthetic trehalose dicorynomvcolate). The immunization protocol may be selected by one skilled in the art without undue experimentation. <br><br> ii. Monoclonal Antibodies <br><br> The anti-PRO antibodies may, alternatively, be monoclonal antibodies. Monoclonal antibodies may be 30 prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature. 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes may be immunized in vitro. <br><br> The immunizing agent will typically include the PRO polypeptide or a fusion protein thereof. Generally, <br><br> 98 <br><br> either peripheral blood lymphocytes ("PBLs") are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell. Goding, Monoclonal Antibodies: Principles and Practice (New York: Academic Press, 1986), pp. 59-103. immortalized 5 cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine, and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT). the culture medium for the hybridomas typically will include hypoxanthine, 10 aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells. <br><br> Preferred immortalized cell lines are those that fuse efficiently, support stable high-level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center. San Diego, California and the American Type Culture Collection, Manassas, 15 Virginia. Human myelomaand mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies. Kozbor, J. Immunol.. 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications (Marcel Dekker, Inc.: New York, 1987) pp. 51-63. <br><br> The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the PRO polypeptide. Preferably, the binding specificity of monoclonal 20 antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem.. 107:220 (1980). <br><br> After the desired hybridoma cells are identified, the clones may be subcloned by limiting dilution procedures 25 and grown by standard methods. Goding, supra. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells may be grown in vivo as ascites in a mammal. <br><br> The monoclonal antibodies secreted by the subclones may be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purificationprocedures such as, for example, protein A-Sepharose, 30 hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. <br><br> The monoclonal antibodies may also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the 35 invention serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal <br><br> 99 <br><br> antibodies in the recombinant host cells. The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; Morrison et al., supra) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide 5 can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody. <br><br> The antibodies may be monovalent antibodies. Methods for preparing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy-chain 10 crosslinking. Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent crosslinking. <br><br> In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments thereof, particularly Fab fragments, can be accomplished using routine techniques known in the art. <br><br> iii. Human and Humanized Antibodies 15 The anti-PRO antibodies may further comprise humanized antibodies or human antibodies. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab. Fab1, F(ab')2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-20 human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two. variable domains, in which all or substantially all of the CDR regions correspond 25 to those of a non-human immunoglobulin, and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody preferably also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Jones et al., Nature. 321:522-525 (1986); Riechmann et al, Nature. 332: 323-329 (1988); Presta, Curr. Op. Struct. Biol.. 2:593-596 (1992). <br><br> Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody 30 has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al, Nature. 321:522-525 (1986); Riechmann et al, Nature. 332: 323-327 (1988); Verhoeven et al, Science. 239: 1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. 35 Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from <br><br> 100 <br><br> a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies. <br><br> Human antibodies can also be produced using various techniques known in the art, including phage display libraries. Hoogenboom and Winter. J. Mol. Biol.. 227: 381 (1991); Marks et al., J. Mol. Biol.. 222: 581 (1991). <br><br> 5 The techniques of Cole et al. and Boemer et al. are also available for the preparation of human monoclonal antibodies. Cole et al.. Monoclonal Antibodies and Cancer Therapy. Alan R. Liss, p. 77 (1985) and Boerner etal., J. Immunol.. 147(1): 86-95 (1991). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed that closely resembles 10 that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016, and in the following scientific publications: Marks et a!., Bio/Technology. H): 779-783 (1992); Lonberg eta!., Nature, 368:856-859(1994): Morrison,Nature, 368:812-813 (1994); Fishwilderal., Nature Biotechnology. 14: 845-851 (1996); Neuberger, Nature Biotechnology, 14: 826 (1996); Lonberg and Huszar, Intern. Rev. 15 Immunol.. 13: 65-93 (1995V <br><br> iv. Bispecific Antibodies <br><br> Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for the PRO polypeptide, the other one is for any other antigen, and preferably for a cell-surface protein or receptor or receptor 20 subunit. <br><br> Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities. Milstein and Cuello, Nature. 305: 537-539 (1983). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a 25 potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification ofthe correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker etal.. EMBO J.. 10:3655-3659(1991). <br><br> Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant-domain sequences. The fusion preferably is with an immunoglobulin heavy-30 chain constant domain, comprising at least part ofthe hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies, see, for example, Suresh et al., Methods in Enzvmology. 121: 210 35 (1986). <br><br> 101 <br><br> v. Heteroconiueate Antibodies Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune-system cells to unwanted cells (U.S. Patent No. 4.676,980), and for treatment of HIV infection. WO 91/00360; WO 92/200373; EP 03089. It is contemplated that the antibodies may 5 be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins may be constructed using a disulfide-exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980. <br><br> vi. Effector Function Engineering <br><br> 10 It may be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, <br><br> e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) may be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See, Caron et at., J. Exp. Med.. 176: 1191-1195 (1992) and 15 Shopes, J. Immunol.. 148:2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al., Cancer Research. 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See, Stevenson et al., Anti-Cancer Drug Design. 3: 219-230 (1989). <br><br> vii. Immunoconjugates <br><br> 20 The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate). <br><br> Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active 25 fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleuritesfordii proteins, dianthin proteins, Phytolaca americana proteins (PAP1, PAP1I, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin. restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 2,2Bi, l3lI, mIn, WY, and l86Re. 30 Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agentssuchasN-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane(IT), bifunctional derivatives of imidoesters (such as dimethyl adipim idate HC1), active esters (such as disuccinimidy 1 suberate), aldehydes (such as glutareldehvde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzovl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-35 active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be <br><br> 102 <br><br> prepared as described in Vitetta et al. Science. 238: 1098(1987). Carbon- 14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaaceiic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See. W094/11026. <br><br> In another embodiment, the antibody may be conjugated to a "receptor" (such as streptavidin) for utilization 5 in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) that is conjugated to a cytotoxic agent (e.g., a radionucleotide). <br><br> viii. lmmunoliposomes <br><br> The antibodies disclosed herein may also be formulated as immunoliposomes. Liposomes containing the 10 antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA. 82: 3688 (1985); Hwang et al, Proc. Natl. Acad. Sci. USA. IT. 4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556. <br><br> Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-15 PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al, J. Biol. Chem.. 257: 286-288 (1982) via a disulfide-interchange reaction. A chemotherapeutic agent (such as Doxorubicin) is optionally contained within the liposome. See, Gabizon et al. J. National Cancer Inst.. 81(19): 1484(1989). <br><br> 20 ix. Pharmaceutical Compositions of Antibodies <br><br> Antibodies specifically binding a PRO polypeptide identified herein, as well as other molecules identified by the screening assays disclosed hereinbefore, can be administered for the treatment of various disorders as noted above and below in the form of pharmaceutical compositions. <br><br> If the PRO polypeptide is intracellular and whole antibodies are used as inhibitors, internalizing antibodies 25 are preferred. However, lipofections or liposomes can also be used to deliver the antibody, or an antibody fragment, into cells. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the binding domain of the target protein is preferred. For example, based upon the variable-region sequences of an antibody, peptide molecules can be designed that retain the ability to bind the target protein sequence. Such peptides can be synthesized chemically and/or produced by recombinant DNA technology. See, e.g., Marasco et 30 al, Proc. Natl. Acad. Sci. USA. 90: 7889-7893 (1993). <br><br> The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Alternatively, or in addition, the composition may comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent. Such molecules are 35 suitably present in combination in amounts that are effective for the purpose intended. <br><br> 103 <br><br> The active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example. hydTOxymethylcellulose or gelatin-microcapsules and poly-(methy Imethacy late) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions. Such techniques 5 are disclosed in Remington's Pharmaceutical Sciences, supra. <br><br> The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. <br><br> Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form 10 of shaped articles, e.g.. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT ™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. 15 While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body fora long time, they may denature or aggregate as a result of exposure to moisture at 37°C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be 20 intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions. <br><br> x. Methods of Treatment using the Antibody It is contemplated that the antibodies to a PRO polypeptide may be used to treat various cardiovascular, 25 endothelial, and angiogenic conditions as noted above. <br><br> The antibodies are administered to a mammal, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal. subcutaneous, intra-articular, intrasynovial. intrathecal, oral, topical, or inhalation routes. Intravenous administration of the antibody is preferred. <br><br> 30 Other therapeutic regimens may be combined with the administration of the antibodies of the instant invention as noted above. For example, if the antibodies are to treat cancer, the patient to be treated with such antibodies may also receive radiation therapy. Alternatively, or in addition, a chemotherapeutic agent may be administered to the patient. Preparation and dosing schedules for such chemotherapeutic agents may be used according to manufacturers' instructions or as determined empirically by the skilled practitioner. Preparation and dosing 35 schedules for such chemotherapy are also described in Chemotherapy Service. Ed., M.C. Perry (Williams &amp; Wilkins: Baltimore, MD. 1992). The chemotherapeutic agent may precede, or follow administration of the <br><br> 104 <br><br> antibody, or may be given simultaneously therewith. The antibody may be combined with an ariti-estrogen compound such as tamoxifen or EVISTA™ or an anti-progesterone such as onapristone (see, EP 6)6812) in dosages known for such molecules. <br><br> If the antibodies are used for treating cancer, it may be desirable also to administer antibodies against other 5 tumor-associated antigens, such as antibodies that bind to one or more of the ErbB2. EGFR, ErbB3, ErbB4, or VEGF receptor(s). These also include the agents set forth above. Also, the antibody is suitably administered serially or in combination with radiological treatments, whether involving irradiation or administration of radioactive substances. Alternatively, or in addition, two or more antibodies binding the same or two or more different antigens disclosed herein may be co-administered to the patient. Sometimes, it may be beneficial also to 10 administer one or more cytokines to the patient. In a preferred embodiment, the antibodies herein are coadministered with a growth-inhibitory agent. For example, the growth-inhibitory agent may be administered first, followed by an antibody of the present invention. However, simultaneous administration or administration ofthe antibody of the present invention first is also contemplated. Suitable dosages for the growth-inhibitory agent are those presently used and may be lowered due to the combined action (synergy) ofthe growth-inhibitory agent and 15 the antibody herein. <br><br> In one embodiment, vascularization of tumors is attacked in combination therapy. The anti-PRO polypeptide and another antibody (e.g., anti-VEGF) are administered to tumor-bearing patients at therapeutically effective doses as determined, for example, by observing necrosis of the tumor or its metastatic foci, if any. This therapy is continued until such time as no further beneficial effect is observed or clinical examination shows no trace of the 20 tumor or any metastatic foci. Then TNF is administered, alone or in combination with an auxiliary agent such as alpha-, beta-, orgamma-interferon. anti-HER2 antibody, heregulin, anti-heregulin antibody, D-factor, interleukin-1 (IL-1), interleukin-2 (1L-2), granulocyte-macrophage colony stimulating factor (GM-CSF), or agents that promote microvascular coagulation in tumors, such as anti-protein C antibody, anti-protein S antibody, or C4b binding protein (see. WO 91/01753, published 21 February 1991), or heat or radiation. <br><br> 25 Since the auxiliary agents will vary in their effectiveness, it is desirable to compare their impact on the tumor by matrix screening in conventional fashion. The administration of anti-PRO polypeptide antibody and TNF is repeated until the desired clinical effect is achieved. Alternatively, the anti-PRO polypeptide antibody is administered together with TNF and, optionally, auxiliary agent(s). In instances where solid tumors are found in the limbs or in other locations susceptible to isolation from the general circulation, the therapeutic agents described 30 herein are administered to the isolated tumor or organ. In other embodiments, a FGF or PDGF antaigonist, such as an anti-FGF or an anti-PDGF neutralizing antibody, is administered to the patient in conjunction with the anti-PRO polypeptide antibody. Treatment with anti-PRO polypeptide antibodies preferably may be suspended during periods of wound healing or desirable neovascularization. <br><br> Forthe prevention or treatment of cardiovascular, endothelial,and angiogenic disorder, the appropriate dosage 35 of an antibody herein will depend on the type of disorder to be treated, as defined above, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody <br><br> 105 <br><br> is suitably administered to the patient at one time or over a series of treatments. <br><br> For example, depending on the type and severity of the disorder, about I ngfkg to 50 mg/kg (e.g., 0.1-20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily or weekly dosage might range from about 5 1 A&lt;g/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated or sustained until a desired suppression of disorder symptoms occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays, including, for example, radiographic tumor imaging. <br><br> xi. Articles of Manufacture with Antibodies <br><br> 10 An article of manufacture containing a container with the antibody and a label is also provided. Such articles are described above, wherein the active agent is an anti-PRO antibody. <br><br> xii. Diagnosis and Prognosis of Tumors using Antibodies If the indication for which the antibodies are used is cancer, while cell-surface proteins, such as growth receptors over expressed in certain tumors, are excellent targets for drug candidates or tumor (e.g., cancer) 15 treatment, the same proteins along with PRO polypeptides find additional use in the diagnosis and prognosis of tumors. For example, antibodies directed against the PRO polypeptides may be used as tumor diagnostics or prognostics. <br><br> For example, antibodies, including antibody fragments, can be used qualitatively or quantitatively to detect the expression of genes including the gene encoding the PRO polypeptide. The antibody preferably is equipped 20 with a detectable, e.g., fluorescent label, and binding can be monitored by light microscopy, flow cytometry, fluorimetry. or other techniques known in the art. Such binding assays are performed essentially as described above. <br><br> In situ detection of antibody binding to the marker gene products can be performed, for example, by immunofluorescence or immunoeiectron microscopy. For this purpose, a histological specimen is removed from the patient, and a labeled antibody is applied to it, preferably by overlaying the antibody on a biological sample. This procedure also allows for determining the distribution of the marker gene product in the tissue examined. It will be apparent to those skilled in the art that a wide variety of histological methods are readily available for in situ detection. <br><br> The following Examples are offered for illustrative purposes only, and are not intended to limit the scope of 30 the present invention in any way. <br><br> The disclosures of all patent and literature references cited in the present specification are hereby incorporated by reference in their entirety. <br><br> 106 <br><br> EXAMPLES <br><br> Commercially available reagents referred to in the Examples were used according to manufacturer's instructions unless otherwise indicated. The source of those cells identified in the following Examples, and throughout the specification, by ATCC accession numbers is the American Type CultureCollection, Manassas, VA. <br><br> 5 Unless otherwise noted, the present invention uses standard procedures of recombinant DNA technology, such as those described hereinabove and in the following textbooks: Sambrook et al.. supra; Ausubel et al., Current Protocols in Molecular Biology (Green Publishing Associates and Wiley Interscience, N.Y., 1989); lnnis et al., PCR Protocols: A Guide to Methods and Applications (Academic Press, Inc.: N.Y., 1990); Harlow et al.. Antibodies: A Laboratory Manual (Cold Spring Harbor Press: Cold Spring Harbor, 1988); Gait, Oligonucleotide 10 Synthesis (IRL Press: Oxford, 1984): Freshney, Animal Cell Culture. 1987; Coiigan et al., Current Protocols in Immunology. 1991. <br><br> EXAMPLE 1 <br><br> Extracellular Domain Homology Screening to Identify Novel Polypeptides and cDNA Encoding Therefor The extracellular domain (ECD) sequences (including the secretion signal sequence, if any) from about 950 15 known secreted proteins from the Swiss-Prot public database were used to search EST databases. The EST databases included public-databases (e.g., Dayhoff, GenBank), and proprietary databases (e.g. LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA). The search was performed using the computer program BLAST or BLAST-2 (Altschul et al., Methods in Enzvmology. 266:460-480 (1996)) as a comparison of the ECD protein sequences to a 6 frame translation of the EST sequences. Those comparisons with a BLAST score of 70 (or in some cases 90) 20 or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green. University of Washington, Seattle, WA). <br><br> Using this extracellular domain homology screen, consensus DNA sequences were assembled relative to the other identified EST sequences using phrap. In addition, the consensus DNA sequences obtained were often (but not always) extended using repeated cycles of BLAST or BLAST-2 and phrap to extend the consensus sequence 25 as far as possible using the sources of EST sequences discussed above. <br><br> Based upon the consensus sequences obtained as described above, oligonucleotides were then synthesized and used to identify by PCR a cDN'A library that contained the sequence of interest and for use as probes to isolate a clone of the full-length coding sequence for a PRO polypeptide. Forward and reverse PCR primers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The 30 probe sequences are typically 40-55 bp in length. In some cases, additional oligonucleotides are synthesized when the consensus sequence is greater than about 1-1.5 kbp. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel et al.. Current Protocols in Molecular Biology, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. <br><br> 35 The cDNA libraries used to isolate the cDNA clones were constructed by standard methods using commercially available reagents such as those from lnvitrogen, San Diego, CA. The cDN A was primed with oligo <br><br> 107 <br><br> dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl sites. <br><br> 5 EXAMPLE 2 <br><br> Isolation of cDNA clones by Amylase Screening <br><br> 1. Preparation of oligo dT primed cDNA library mRNA was isolated from a human tissue of interest using reagents and protocols from Invitrogen, San Diego, CA (Fast Track 2). This RNA was used to generate an oligo dT primed cDNA library in the vector pRK5D using 10 reagents and protocols from Life Technologies, Gaithersburg, MD (Super Script Plasmid System). In this procedure, the double stranded cDNA was sized to greater than 1000 bp and the Sall/Notl linkered cDNA was cloned into Xhol/NotI cleaved vector. pRK5D is a cloning vector that has an sp6 transcription initiation site followed by an Sfil restriction enzyme site preceding the Xhol/NotI cDNA cloning sites. <br><br> 2. Preparation of random primed cDNA library <br><br> 15 A secondary cDNA library was generated in order to preferentially represent the 5" ends of the primary cDNA <br><br> clones. Sp6 RNA was generated from the primary library (described above), and this RNA was used to generate a random primed cDNA library in the vector pSST-AMY.O using reagents and protocols from Life Technologies (Super Script Plasmid System, referenced above). In this procedure the double stranded cDNA was sized to 500-1000 bp, linkered with blunt to Notl adaptors, cleaved with Sfil, and cloned into Sfil/NotI cleaved vector. pSST-20 AMY.O is a cloning vector that has a yeast alcohol dehydrogenase promoter preceding the cDNA cloning sites and the mouse amylase sequence (the mature sequence without the secretion signal) followed by the yeast alcohol dehydrogenase terminator, after the cloning sites. Thus, cDNAs cloned into this vector that are fused in frame with amylase sequence will lead to the secretion of amylase from appropriately transfected yeast colonies. <br><br> 3. Transformation and Detection <br><br> 25 DNA from the library described in paragraph 2 above was chilled on ice to w hich was added electrocompetent <br><br> DH10B bacteria (Life Technologies. 20 ml). The bacteria and vector mixture was then electroporated as recommended by the manufacturer. Subsequently, SOC media (Life Technologies, 1 ml) was added and the mixture was incubated at 37°C for 30 minutes. The transformants were then plated onto 20 standard 150 mm LB plates containing ampicillin and incubated for 16 hours (37°C). Positive colonies were scraped off the plates and 30 the DNA was isolated from the bacterial pellet using standard protocols, e.g., CsCI-gradient. The purified DNA was then carried on to the yeast protocols below. <br><br> The yeast methods were divided into three categories: (1) Transformation of yeast with the plasmid/cDNA combined vector; (2) Detection and isolation of yeast clones secreting amylase: and (3) PCR amplification of the insert directly from the yeast colony and purification of the DNA for sequencing and further analysis. <br><br> 108 <br><br> The yeast strain used was HD56-5A (ATCC-90785). This strain has the following genotype: MAT alpha, ura3-52, leu2-3, Ieu2-112, his3-1I.his3-I5, MAL", SUC\ GAL'. Preferably, yeast mutants can be employed that have deficient post-translational pathways. Such mutants may have translocation deficient alleles in seel I, secll, sec62, with truncated secll being most preferred. Alternatively, antagonists (including antisense nucleotides 5 and/or ligands) which interfere with the normal operation of these genes, other proteins implicated in this post translation pathway (e.g., SEC61p, SEC72p, SEC62p, SEC63p, TDJlp or SSAlp-4p) or the complex formation of these proteins may also be preferably employed in combination with the amylase-expressing yeast. <br><br> Transformation was performed based on the protocol outlined by Gietz et al., Nucl. Acid. Res.. 20:1425 (1992). Transformed cells were then inoculated from agar into YEPD complex media broth (100 ml) and grown 10 overnight at 30°C. The YEPD broth was prepared as described in Kaiser et al., Methods in Yeast Genetics. Cold Spring Harbor Press, Cold Spring Harbor, NY, p. 207 (1994). The overnight culture was then diluted to about 2 x 104 cells/ml (approx. OD600=0.1) into fresh YEPD broth (500 ml) and regrown to 1 x 107 cells/ml (approx. 00^=0.4-0.5). <br><br> The cells were then harvested and prepared for transformation by transfer into GS3 rotor bottles in a Sorval 15 GS3 rotor ai 5.000 rpm for 5 minutes, the supernatant discarded, and then resuspended into sterile water, and centrifuged again in 50 ml falcon tubes at 3,500 rpm in a Beckman GS-6KR centrifuge. The supernatant was discarded and the cells were subsequently washed with LiAc/TE (10 ml, 10 mM Tris-HCl, 1 mM EDTA pH 7.5, 100 mM LuOOCCH,), and resuspended into LiAc/TE (2.5 ml). <br><br> Transformation took place by mixing the prepared cells (100 juI) with freshly denaturedsingle stranded salmon 20 testes DNA (Lofstrand Labs, Gaithersburg, MD) and transforming DNA (1 ug- vol. &lt; 10 tA) in microfuge tubes. The mixture was mixed briefly by vortexing, then 40% PEG/TE (600 pd, 40% polyethylene glycol-4000,10 mM Tris-HCl. 1 mM EDTA, 100 mM Li:OOCCH3, pH 7.5) was added. This mixture was gently mixed and incubated at 30°C while agitating for 30 minutes. The cells were then heat shocked at 42°C for 15 minutes, and the reaction vessel centrifuged in a microfuge at 12,000 rpm for 5-10 seconds, decanted and resuspended into TE (500 /ul, 10 25 mM Tris-HCl. 1 mM EDTA pH 7.5) followed by recentrifugation. The cells were then diluted into TE (1 ml) and aliquots (200 ^1) were spread onto the selective media previously prepared in 150 mm growth plates (VWR). <br><br> Alternatively, instead of multiple small reactions, the transformation was performed using a single, large scale reaction, wherein reagent amounts were scaled up accordingly. <br><br> The selective media used was a synthetic complete dextrose agar lacking uracil (SCD-Ura) prepared as 30 described in Kaiser et al.. Methods in Yeast Genetics. Cold Spring Harbor Press. Cold Spring Harbor, NY. p. 208-210 (1994). Transformants were grown at 30°C for 2-3 days. <br><br> The detection of colonies secreting amylase was performed by including red starch in the selective growth media. Starch was coupled to the red dye (Reactive Red-120, Sigma) as per the procedure described by Biely et al.. Anal. Biochem.. 172:176-179 (1988). The coupled starch was incorporated into the SCD-Ura agar plates at 35 a final concentration of 0.15% (w/v), and was buffered with potassium phosphate to a pH of7.0 (50-100 mM final concentration). <br><br> The positive colonies were picked and streaked across fresh selective media (onto 150 mm plates) in order <br><br> 109 <br><br> to obtain well isolated and identifiable single colonies. Well isolated single colonies positive for amylase secretion were detected by direct incorporation of red starch into buffered SCD-Ura agar. Positive colonies were determined by their ability to break down starch resulting in a clear halo around the positive colony visualized directly. <br><br> 4. Isolation of DNA bv PCR Amplification 5 When a positive colony was isolated, a portion of it was picked by a toothpick and diluted into sterile water <br><br> (30 p&lt;l) in a 96 well plate. At this time, the positive colonies were either frozen and stored for subsequent analysis or immediately amplified. An aliquot of cells (5 ptl) was used as a template for the PCR reaction in a 25 pd volume containing: 0.5 jul Klentaq (Clontech, Palo Alto, CA); 4.0 tA 10 mM dNTP's (Perkin Elmer-Cetus); 2.5 p\ Kentaq buffer (Clontech); 0.25 p\ forward oligo 1; 0.25 n\ reverse oligo 2; 12.5 ju\ distilled water. The sequence of the 10 forward oligonucleotide 1 was: <br><br> 5'-TGTAAAACC.ACGGCCAGTTAAATAGACCTGCAATTATTAATCT-3' (SEQ IDNO:l) The sequence of reverse oligonucleotide 2 was: <br><br> 5'-CAGGAAACAGCTATGACCACCTGCACACCTGCAAATCCATT-3' (SEQ ID NO:2) <br><br> PCR was then performed as follows: <br><br> 15 <br><br> a. <br><br> Denature <br><br> 92 °C, <br><br> 5 minutes <br><br> b. <br><br> 3 cycles of: <br><br> Denature <br><br> Anneal <br><br> Extend <br><br> 92°C, 59°C, 72 °C, <br><br> 30 seconds 30 seconds 60 seconds <br><br> 20 <br><br> c. <br><br> 3 cycles of: <br><br> Denature <br><br> Anneal <br><br> Extend <br><br> 92 °C, 57°C, 72 °C, <br><br> 30 seconds 30 seconds 60 seconds <br><br> d. <br><br> 25 cycles of: <br><br> Denature <br><br> Anneal <br><br> Extend <br><br> 92 °C, 55 °C, 72 °C, <br><br> 30 seconds 30 seconds 60 seconds <br><br> 25 <br><br> e. <br><br> Hold <br><br> 4°C <br><br> The underlined regions of the oligonucleotides annealed to the ADH promoter region and the amylase region, respectively, and amplified a 307 bp region from vector pSST-AMY.O when no insert was present. Typically, the first 18 nucleotides of the 5' end of these oligonucleotides contained annealing sites for the sequencing primers. Thus, the total product of the PCR reaction from an empty vector was 343 bp. However, signal sequence-fused 30 cDNA resulted in considerably longer nucleotide sequences. <br><br> Following the PCR. an aliquot of the reaction (5 ptl) was examined by agarose gel electrophoresis in a 1% agarose gel using a Tris-Borate-EDTA (TBE) buffering system as described by Sambrook et al., supra. Clones resulting in a single strong PCR product larger than 400 bp were further analyzed by DNA sequencing after purification with a 96 Qiaquick PCR clean-up column (Qiagen Inc., Chatsworth, CA). <br><br> 110 <br><br> EXAMPLE 3 <br><br> Isolation of cDNA Clones Using Signal Algorithm Analysis Various polypeptide-encoding nucleic acid sequences were identified by applying a proprietary signal sequence finding algorithm developed by Genentech, Inc., (South San Francisco, CA) upon ESTs as well as 5 clustered and assembled EST fragments from public (e.g., GenBank) and/or private (L1FESEQ®, lncyte Pharmaceuticals, Inc., Palo Alto. CA) databases. The signal sequence algorithm computes a secretion signal score based on the character of the DNA nucleotides surrounding the first and optionally the second methionine codon(s) (ATG) at the 5'-end of the sequence or sequence fragment under consideration. The nucleotides following the first ATG must code for at least 35 unambiguous amino acids without any stop codons. If the first ATG has the required 10 amino acids, the second is not examined. If neither meets the requirement, the candidate sequence is not scored. In order to determine whether the EST sequence contains an authentic signal sequence, the DNA and corresponding amino acid sequences surrounding the ATG codon are scored using a set of seven sensors (evaluation parameters) known to be associated with secretion signals. Use of this algorithm resulted in the identification of numerous polypeptide-encoding nucleic acid sequences. <br><br> 15 EXAMPLE 4 <br><br> Isolation of cDNA Clones Encoding Human PRO 172 A consensus DNA sequence encoding a delta-like homologue was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA28765. Based on the DNA28765 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library 20 that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0172. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> 2876S-f (0L1644V <br><br> 5"-GGATCTCGAGAACAGCTACTCC-3' (SEQ ID NO:5) <br><br> 25 28765-r {QLI64S1: <br><br> 5'-TCGTCCACGTTGTCGTCACATG-3' (SEQ ID NO:6) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA28765 sequence which had the following nucleotide sequence: <br><br> 28765.p (OL1643) hybridization probe: 30 5'-AAATCTGTGAATTGAGTGCCATGGACCTGTTGCGGACGGCCCTTGCTT-3' (SEQ ID NO:7) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PROl 72 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal kidney tissue. <br><br> Ill <br><br> DNA sequencing ofthe isolated clones isolated as described above gave the full-length DNA sequence for DNA35916-1161 [Figure I, SEQ ID NO:3]; and the derived protein sequence for PR0172. <br><br> The entire coding sequence of DNA35916-1161 is included in Figure 1 (SEQ IDNO:3). Clone DNA35916-1161 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 38-40, 5 and an apparent stop codon at nucleotide positions 2207-2209. The predicted polypeptide precursor is 723 amino acids long. Analysis of the full-length PRO 172 sequence shown in Figure 2 (SEQ ID NO:4) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 172 polypeptide shown in Figure 2 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 21; a transmembrane 10 domain from about amino acid 546 to about amino acid 566; an N-glycosylation site from about amino acid 477 to about amino acid 481: a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 660 to about amino acid 664; tyrosine kinase phosphorylation sites from about amino acid 176 to about amino acid 185, and from about amino acid 252 to about amino acid 261; N-myristoylation sites from about amino acid 2 to about amino acid 8, from about amino acid 37 to about amino acid 43, from about amino acid 40 to about amino 15 acid 46, from about amino acid 98 to about amino acid 104, from about amino acid 99 to about amino acid 105, from about amino acid 262 to about amino acid 268, from about amino acid 281 to about amino acid 287, from about amino acid 282 to about amino acid 288, from about amino acid 301 to about amino acid 307, from about amino acid 310 to about amino acid 316, from about amino acid 328 to about amino acid 334, from about amino acid 340 to about amino acid 344. from about amino acid 378 to about amino acid 384, from about amino acid 387 20 to about amino acid 393. from about amino acid 512 to about amino acid 518, from about amino acid 676 to about amino acid 682, from about amino acid 683 to about amino acid 689, and from about amino acid 695 to about amino acid 701; aspartic acid and asparagine hydroxylation sites from about amino acids 343 to about amino acid 355, from about amino acid 420 to about amino acid 432. and from about amino acid 458 to about amino acid 470; a prokaryotic membrane lipoprotein lipid attachment site from about amino acid 552 to about amino acid 563; and 25 EGF-like domain cysteine pattern signatures from about amino acid 243 to about amnino acid 255, from about amino acid 274 to about amino acid 286, from about amino acid 314 to about amino acid 326, from about amino acid 352 to about amino acid 364, from about amino acid 391 to about amino acid 403, from about amino acid 429 to about amino acid 441. from about amino acid 467 to about amino acid 479. and from about amino acid 505 to about amino acid 517. Clone DNA35916-1161 has been deposited with the ATCC on October 28, 1997 and is 30 assigned ATCC deposit no. 209419. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 2 (SEQ 1DN0:4), PR0172 shows 89% amino acid sequence identity to delta-1 mouse protein. <br><br> EXAMPLE 5 Isolation of cDNA Clones Encoding Human PRO 178 35 An expressed sequence tag (EST) DNA database ( LIFESEQ®, lncyte Pharmaceuticals, Palo Alto, CA) <br><br> was searched and an EST was identified which showed homology to the human TIE ligand family. <br><br> 112 <br><br> RNA for construction of cDNA libraries was then isolated from human fetal lung tissue. The cDNA libraries used to isolate the cDNA clones encoding human PRO 178 were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA. The cDNA was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately 5 by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl. <br><br> Oligonucleotides probes based upon the above described EST sequence were then synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the 10 full-length coding sequence for PRO 178. Forward and reverse PCR primers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel et al., Current Protocols in Molecular Biology, supra, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe 15 oligonucleotide and one of the primer pairs. <br><br> The oligonucleotide probes employed were as follows: <br><br> NL8.5-1: <br><br> 5'-ACGTAGTTCCAGTATGGTGTGAGCAGCAACTGGA-3* (SEQ ID NO: 10) <br><br> NL8.3-1: <br><br> 20 5'-AGTCCAGCCTCCACCCTCCAGTTGCT-3' (SEQ ID NO: 11) <br><br> NL8.3-2: <br><br> 5'-CCCCAGTCCTCCAGGAGAACCAGCA-3' (SEQ ID NO: 12) <br><br> A full length clone [DNA23339-1130]was identified that contained a single open reading frame with an apparent translational initiation site at nucleotide positions 118-120 and a stop signal at nucleotide positions 1528-25 1530 (Figure 3, SEQ ID NO:8). The predicted polypeptide precursor is 470 amino acids long, has a calculated | molecular weight of approximately 51,694 daltons and an estimated pi of approximately 8.86. Analysis of the full-length PROl 78 sequence shown in Figure 4 (SEQ ID NO:9) evidences the presence of a variety of important polypeptide domains as shown in Figure 4, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 178 polypeptide shown in Figure 4 evidences 30 the presence of the following: a signal peptide from about amino acid 1 to about amino acid 20; N-glycosylation sites from about amino acid 58 to about amino acid 62, and from about amino acid 145 to about amino acid 149; acAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 97 to about amino acid 101; a tyrosine kinase phosphorylation site from about amino acid 441 to about amino acid 448; N-myristoylation sites from about amino acid 16 to about amino acid 22; from about amino acid 23 to about amino acid 29, from 35 about amino acid 87 to about amino acid 93, from about amino acid 108 to about amino acid 114, from about amino acid 121 to about amino acid 127.from about amino acid 125 to about amino acid 131,from about amino acid 129 to about amino acid 135, from about amino acid 187 to about amino acid 193, from about amino acid 293 to about <br><br> 113 <br><br> amino acid 299, from about amino acid 353 to about amino acid 359. from about amino acid 378 to about amino acid 384. from about amino acid 445 to about amino acid 451, and from about amino acid 453 to about amino acid 459; a cell attachment site from about amino acid 340 to about amino acid 343; and a fibrinogen beta and gamma chains C-terminal domain signature from about amino acid 418 to about amino acid 431. Clone DM A23339-1130 5 has been deposited with ATCC on September 18, 1997 and is assigned ATCC deposit no. 209282. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 4 (SEQ ID NO:9), PROl 78 (herein designated NL8) shows a 23% amino acid sequence identity to both ligand 1 and ligand 2 of the TIE2 receptor. Ligand I and ligand 2 of the TIE-2 receptor are 64% identical and 40-43% identical, respectively, to PROl 78. The abbreviation "TIE" is an acronym which stands for "tyrosine kinase containing Ig 10 and EGF homology domains" and was coined to designate a new family of receptor tyrosine kinases. <br><br> EXAMPLE 6 Isolation of cDNA Clones Encoding Human PRO 179 A cDNA sequence isolated in the amylase screen described in Example 2 above was found, by BLAST and FastA sequence alignment, to have sequence homology to a nucleotide sequence encoding various angiopoietin 15 proteins. This cDN A sequence is herein designated DNA 10028 and/or DNA25250. Based on the sequence homology, probes were generated from the sequence of the DNA 10028 molecule and used to screen a human fetal liver library (LIB6) prepared as described in paragraph 1 of Example 2 above. The cloning vector was pRK5B (pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)), and the cDNA size cut was less than 2800 bp. <br><br> 20 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PRO 179 gene using a probe oligonucleotide and one of the PCR primers. <br><br> A full length clone [DNA 16451-1388] was identified that contained a single open reading frame with an apparent transiational initiation site at nucleotide positions 37-39, and a stop signal at nucleotide positions 1417-25 1419 (Figure 5; SEQ ID NO: 13). The predicted polypeptide precursor is 460 amino acids long, and has a calculated molecular weight of approximately 53,637 daltons and an estimated pi of approximately 6.61. Analysis of the full-length PRO 179 sequence shown in Figure 6 (SEQ ID NO: 14) evidences the presence of a variety of important polypeptide domains as shown in Figure 6, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 179 polypeptide shown in Figure 6 evidences 30 the presence of the following: a signal peptide from about amino acid I to about amino acid 16, N-glycosylation sites from about amino acid 23 to about amino acid 27, from about amino acid 115 to about amino acid 119, from about amino acid 296 to about amino acid 300, and from about amino acid 357 to about amino acid 361; cAMP-and cGMP-dependent protein kinase phosphorylation sites from about amino acid 100 to about amino acid 104, and from about amino acid 204 to about amino acid 208; a tyrosine kinase phosphorylation site from about amino 35 acid 342 to about amino acid 35 l;N-myristoylation sites from about amino acid 279 to about amino acid 285, from about amino acid 352 to about amino acid 358, and from about amino acid 367 to about amino acid 373; and <br><br> 114 <br><br> zipper patterns from about amino acid 120 to about amino acid 142, and from about amino acid 127 to about amino acid 149. Clone DNA16451-1388 was deposited with the ATCC on April 14,1998, and is assigned ATCC deposit no. 209776. <br><br> Analysis of the amino acid sequence of the full-length PR0179 polypeptide shown in Figure 6 (SEQ ID 5 NO: 14) suggests that it possesses significant similarity to the angiopoietin family of proteins, thereby indicating that PRO 179 may be a novel angiopoietin family member. More specifically, an analysis of the Dayhoff database (version 35.45 SwissProt 35) evidenced significant homology between the PRO 179 amino acid sequence and the following Dayhoff sequences: AF004326_1, PJ194605, HSU83508_1, PJR94603, P_R94317, AF025818_1, HSY16132_1, P_R65760,137391 and HUMRSC192_1. <br><br> 10 EXAMPLE 7 <br><br> Isolation of cDNA Clones Encoding Human PROl 82 The nucleic acid sequence of the relaxin molecule, a member of the insulin family of proteins, was used to search for homologous sequences in a human colon cDNA library of expressed sequence tags (ESTs) from Incyte, Inc. (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA). Two ESTs were obtained, Incyte EST nos. 15 INC2328985 and INC778319, each having approximately 40% homology to the region of the relaxin nucleic acid sequence, and represent sequences within a gene of an insulin-like polypeptide (BUP). <br><br> A cDNA library was constructed from human uterus mRNA obtained from Clontech Laboratories, Inc. Palo Alto, CA, catalog no. 6537-1. The full-length nucleic acid sequence of PR0182 was obtained by screening a plasmid cDNA library described above, by colony hybridization using oligonucleotides designed based on the EST 20 sequences fromlncyte, Inc. (Ihcyte EST INC2328985 and Incyte EST INC778319). The cDNA libraries used to isolate the cDNA clones encoding human PROl 82 were constructed by standard methods using commercially available reagents such as those from Ihvitrogen, San Diego, CA. The cDNA was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Nod, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B 25 is a precursor of pRK5D that does not contain the Sfil site; see, Holmes etal, Science. 253:1278-1280 (1991)) in the unique Xhol and Notl. <br><br> Oligonucleotides probes based upon the above described ESTsequences were then synthesized: l)to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PRO 182. Forward and reverse PCR primers generally range from 20 to 30 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. Hie probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel et al., Current Protocols in Molecular Biology, supra, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. <br><br> 35 The primer oligonucleotides sequences used are as follows: <br><br> 5'-CACATrCAGTCCTCAGCAAAATGAA-3' (SEQ ID NO: 17) <br><br> 115 <br><br> S'-GAGAATAAAAACAGAGTGAAAATGGAGCCCTTCATTTTGC-S' (SEQ ID NO: 18) 5'-CTCAGCTTGCTGAGCTTGAGGG A-3' (SEQ ID NO: 19) <br><br> A full length clone [DNA27865-I091]was identified that contained a single open reading frame with an apparent translational initiation site at nucleotide positions 39-41 and a stop signal at nucleotide positions 444-446 5 (Figure 7, SEQ ID NO: 15). The predicted polypeptide precursor is 135 amino acids long, and has a calculated molecular weight of approximately 15,319 daltons and an estimated pi of approximately 7.39. Analysis of the full-length PRO 182 sequence shown in Figure 8 (SEQ ID NO: 16) evidences the presence of a variety of important polypeptide domains as shown in Figure 8, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 182 polypeptide shown in Figure 8 evidences 10 the presence of the following: a signal peptide from about amino acid I to about amino acid 18; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 107 to about amino acid 111; M-myristoylation sites from about amino acid 3 to about amino acid 9, from about amino acid 52 to about amino acid 58, from about amino acid 96 to about amino acid 102, and from about amino acid 125 to about amino acid 131; and an insulin family signature from about amino acid 121 to about amino acid 136. Clone DNA27865-I091 has 15 been deposited with ATCC on September 23, 1997 and is assigned ATCC deposit no. 209296. <br><br> Based on a BLAST and FastA sequence alignment analysis (using the ALIGN computer program) of the full-length sequence shown in Figure 8 (SEQ ID NO: 16), the PROl 82 polypeptide sequence was homologous to but clearly different from any known polypeptide molecule, and therefore the PRO 182 polypeptide constitutes a novel member of the insulin family of proteins. <br><br> 20 EXAMPLE 8 <br><br> Isolation of cDNA Clones Encoding Human PROl 87 An expressed sequence tag (EST) DNA database ( LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) was searched and an EST (no. IN843193) was identified which showed homology to fibroblast growth factor (FGF-8) also known as androgen-induced growth factor. <br><br> ^^25 RNA for construction of cDNA libraries was then isolated from human fetal lung tissue; The cDNA libraries used to isolate the cDNA clones encoding human PROl 87 were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA. The cDNA was primed with oligo dT containing a Notl site, linked w ith blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; 30 pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes el alScience. 253:1278-1280 (1991)) in the unique Xhol and Notl. <br><br> Oligonucleotides probes based upon the above described EST sequence were then synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PRO 187. Forward and reverse PCR primers generally range from 20 to 30 35 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries <br><br> 116 <br><br> was screened by PCR amplification, as per Ausubel et alCurrent Protocols in Molecular Biology, supra, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. <br><br> Several libraries from various tissue sources were screened by PCR amplification with the following 5 oligonucleotide probes: <br><br> IN843193.f (OLI315): <br><br> 5'-CAGTACGTGAGGGACCAGGGCGCCATGA-3' (SEQ ID NO:22) <br><br> IN843193.r (OL1317): <br><br> 5'-CCGGTGACCTGCACGTGCTTGCCA-3' (SEQ ID NO:23) <br><br> 10 A positive library was then used to isolate clones encoding the FGF-8 homologue gene using one of the above oligonucleotides and the following oligonucleotide probe: <br><br> IN843193.p (OLI316): <br><br> 5'-GCGGATCTGCCGCCTGCTCANCTGGTCGGTCATGGCGCCCT-3' (SEQ ID NO:24) <br><br> A full length clone (DNA27864-1155} was identified that contained a single open reading frame with an 15 apparent translational initiation site at nucleotide positions 26-28 and a stop signal at nucleotide positions 641-643 (Figure 9, SEQ ID N0:20). The predicted polypeptide precursor is 205 amino acids long, has a calculated molecular weight of approximately 23,669 daltons and an estimated pi of approximately 10.75. Analysis of the full-length PROl 87 sequence shown in Figure 10 (SEQ lDNO:21) evidences the presence of a variety of important polypeptide domains as shown in Figure 10, wherein the locations given for those important polypeptide domains 20 are approximate as described above. Analysis of the full-length PROl87 polypeptide shown in Figure 10 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 22; N-glycosylation sites from about amino acid 9 to about amino acid 13. and from about amino acid 126 to about amino acid 130: a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 60 to about ^ amino acid 64; tyrosine kinase phosphorylation sites from about amino acid 39 to about amino acid 48, and from 25 about amino acid 89 to about amino acid 97; N-myristoylation sites from about amino acid 69 to about amino acid 75, and from about amino acid 188 to about amino acid 194; an amidation site from about amino acid 58 to about amino acid 62; and a HBGF/FGF family signature from about amino acid 103 to about amino acid 128. Clone DNA27864-1155 has been deposited with ATCC on October 16, 1997 and is assigned ATCC deposit no. 209375. Based on a BLAST and FastA sequence alignment analysis (using the ALIGN computer program) of the full-30 length sequence shown in Figure 10 (SEQ ID NO:21), PRO 187 shows 74% amino acid sequence identity to human fibroblast growth factor-8 (androgen-induced growth factor). <br><br> EXAMPLE 9 Isolation of cDNAClones Encoding Human PROl 88 An expressed sequence tag (EST) DNA database ( LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) <br><br> 117 <br><br> was searched and an EST was identified which showed homology to the human TIE ligand family. <br><br> RNA for construction of cDNA libraries was then isolated from human fetal lung tissue. The cDNA libraries used to isolate the cDNA clones encoding human PRO 188 were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA. The cDNA was primed with oligo 5 dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does riot contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl. <br><br> Oligonucleotides probes based upon the above described EST sequence were then synthesized: 1) to identify 10 by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PROl88. Forward and reverse PCR primers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel el al., Current Protocols in Molecular Biology, supra, with the 15 PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. <br><br> The oligonucleotide probes employed were as follows: <br><br> NL5.5-1: <br><br> 5'-CAGGTTATCCCAGAGATTTAATGCCACCA-3' (SEQ ID NO:27) <br><br> 20 NL5.3-1: <br><br> 5'-TTGGTGGGAGAAGTTGCCAGATCAGGTGGTGGCA-3' (SEQ ID NO:28) <br><br> NL5.3-2: <br><br> 5'-TTCACACCATAACTGCATTGGTCCA-3' (SEQ ID NO:29) <br><br> A full length clone [DNA28497-1130] was identified that contained a single open reading frame with an 25 apparent translational initiation site at nucleotide positions 449-451 and a stop signal at nucleotide positions 1922-1924 (Figure 11, SEQ ID NO:25). The predicted polypeptide precursor is 491 amino acids long, and has a calculated molecular weight of approximately 56,720 daltons and an estimated pi of approximately 8.56. Analysis of the full-length PRO 188 sequence shown in Figure 12 (SEQ ID NO:26) evidences the presence of a variety of important polypeptide domains as shown in Figure 12, wherein the locations given for those important polypeptide 30 domains are approximate as described above. Analysis of the full-length PROl 88 polypeptide shown in Figure 12 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 23; N-glycosylation sites from about amino acid 160 to about amino acid 164, and from about amino acid 188 to about amino acid 192; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 120 to about amino acid 124: tyrosine kinase phosphorylation sites from about amino acid 173 to about amino acid 180, 35 and from about amino acid 387 to about amino acid 396; N-myristoylation sites from about amino acid 70 to about amino acid 76. from about amino acid 110 to about amino acid 116, from about amino acid 232 to about amino acid <br><br> 118 <br><br> 238, from about amino acid 343 to about amino acid 349, from about amino acid 400 to about amino acid 406, from about amino acid 467 to about amino acid 473, and from about amino acid 475 to about amino acid 487; and a fibrinogen beta and gamma chains C-terminal domain signature from about amino acid 440 to about amino acid 453. Clone DNA28497-I130 has been deposited with ATCC on September 18,1997 and is assigned ATCC deposit 5 no. 209279. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 12 (SEQ ID NO:26), PRO 188 (herein designated NL5) shows 24% amino acid sequence identity to both ligand 1 and ligand 2 ofthe TIE2 receptor, Ligand 1 and ligand 2 ofthe TIE-2 receptor are 64% identical and 40-43% identical, respectively, to PROl 88. The abbreviation "TIE" is an acronym which stands for "tyrosine kinase containing Ig 10 and EGF homology domains" and was coined to designate a new family of receptor tyrosine kinases. <br><br> EXAMPLE 10 Isolation of cDNA Clones Encoding Human PROl 95 A cDNA sequence isolated in the amylase screen described in Example 2 above is herein designated DNA 13199ABI2. The DNA13199 AB12 sequence was then compared to a variety of expressed sequence tag 15 (EST) databases which included public EST databases (e.g., GenBank) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 (Altshul et al., Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green. University of Washington, Seattle, Washington). The consensus sequence 20 identified is herein designated DNA22778. <br><br> Based on the DNA 13199ABI2 sequence and DNA22778 sequences, oligonucleotide probes were generated and used to screen a human placenta library (LIB89) prepared as described in paragraph 1 of Example 2 above. The cloning vector was pRK5B (pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)), and the cDNA size cut was less than 2800 bp. <br><br> *25 PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (22778.0: <br><br> 5'-ACAAGCTGAGCTGCTGTGACAG-3' (SEQ ID NO:32) <br><br> reverse PCR primer (22778.r): <br><br> 5-TGATTCTGGCAACCAAGATGGC-3' (SEQ ID NO:33) <br><br> 30 Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA22778 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe (22778.pV <br><br> 5-ATGGCCTTGGCCGGAGGTTCGGGGACCGCTTCGGCTGAAG-3' (SEQ ID NO:34) <br><br> 119 <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0195 gene using a probe oligonucleotide and one of the PCR primers. <br><br> A full length clone (DNA26847-1395] was identified that contained a single open reading frame with an 5 apparent translational initiation site at nucleotide positions 70-72, and a stop signal at nucleotide positions 1039-1041 (Figure 13; SEQ ID N0:30). The predicted polypeptide precursor is 323 amino acids long, and has a calculated molecular weight of approximately 36,223 daltons and an estimated pi of approximately 5.06. Analysis ofthe full-length PROl 95 sequence shown in Figure 14 (SEQ ID NO:31) evidences the presence of a variety of important polypeptide domains as shown in Figure 14, wherein the locations given for those important polypeptide 10 domains are approximate as described above. Analysis of the full-length PROl 95 polypeptide shown in Figure 14 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 31, a transmembrane domain from about amino acid 242 to about amino acid 262; an N-glycosylation site from about amino acid 90 to about amino acid 94; and N-myristoylation sites from about amino acid 28 to about amino acid 34, from about amino acid 29 to about amino acid 35, from about amino acid 31 to about amino acid 37, and from 15 about amino acid 86 to about amino acid 92. Clone DNA26847-1395 was deposited with the ATCC on April 14, 1998 , and is assigned ATCC deposit no. 209772. <br><br> Analysis of the amino acid sequence of the full-length PRO 195 polypeptide (Figure 14; SEQ ID NO:31) suggests that it possesses no significant similarity to any known protein. However, an analysis of the Dayhoff database (version 35.45 SwissProt 35) evidenced some degree of homology between the PR0195 amino acid 20 sequence and the following Dayhoff sequences: P_P91380, AF035118 1, HUMTROPCSl, NUODSALTY and E70002. <br><br> EXAMPLE 11 Isolation of cDNA Clones Encoding Human PRQ212 A consensus DNA sequence was assembled relative to other EST sequences (including an EST proprietary 25 to Genentech) using phrap as described in Example I above. Based on the assembled consensus sequence, oligonucleotides were synthesized: I) to identify by PCR a cDNA libraiy that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0212. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR nrimer: <br><br> 30 5'-CACGCTGGTTTCTGCTTGGAG-3' (SEQIDNO:37) <br><br> reverse PCR primer: <br><br> 5'-AGCTGGTGCACAGGGTGTCATG-3' (SEQ IDNO:38) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus sequence which had the following nucleotide sequence: <br><br> 120 <br><br> hybridization probe: <br><br> 5-CCCAGGCACCTTCTCAGCCAGCCAGCAGCTCCAGCTCAGAGCAGTGCCAGCCC-3{SEQIDNO:39) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones 5 encoding the PR0212 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal lung tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA30942-1134 [Figure 15, SEQ ID NO:35]; and the derived protein sequence for PR0212. <br><br> The entire coding sequence of DNA30942-1134 is included in Figure 15 (SEQ ID NO:35). Clone 10 DNA30942-1134 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 101-103, and an apparent stop codon at nucleotide positions 1001-1003. The predicted polypeptide precursor is 300 amino acids long. Analysis of the full-length PR02I2 sequence shown in Figure 16 (SEQ ID NO:36) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0212 15 polypeptide shown in Figure 16 evidences the presence ofthe following: a signal peptide from about amino acid 1 to about amino acid 23; an N-glycosylation site from about amino acid 173 to about amino acid 177; cAMP- and cGMP-dependent protein kinase phosphorylation sites from about amino acid 63 to about amino acid 67, and from about amino acid 259 to about amino acid 263; a tyrosine kinase phosphorylation site from about amino acid 28 to about amino acid 37; N-myristoylation sites from about amino acid 156 to about amino acid 162, from about 20 amino acid 178 to about amino acid 184, from about amino acid 207 to about amino acid 213, from about amino acid 266 to about amino acid 272, and from about amino acid 287 to about amino acid 293. Clone DNA30942-1134 has been deposited with the ATCC on September 16, 1997 and is assigned ATCC deposit no. 209254. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 16 (SEQ ID NO:36), PR0212 shows some amino acid sequence identity to TNFR2 (28.7%). <br><br> ^25 EXAMPLE 12 <br><br> Isolation of cDNA Clones Encoding Human PRQ214 <br><br> A consensus DNA sequence encoding an EGF-like homologue was assembledreiativeto other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA28744. Based on the assembled DNA28744 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a 30 cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0214. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (01.1556): <br><br> 5'-ATTCTGCGTGAACACTGAGGGC-3* (SEQ ID NO:42) <br><br> 121 <br><br> reverse PCR primer (OLI557): 5'-ATCTGCTTGTAGCCCTCGGCAC-3' <br><br> (SEQ ID NO:43) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA28744 consensus sequence which had the following nucleotide sequence: <br><br> 5 hybridization probe (OLI555): <br><br> 5'-CCTGGCTATCAGCAGGTGGGCTCCAAGTGTCTCGATGTGGATGAGTGTGA-3' (SEQ ID NO:44) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR02I4 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction 10 of the cDNA libraries was isolated from human fetal lung tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA32286-1191 [Figure 17, SEQ ID N0:40]; and the derived protein sequence for PR0214. <br><br> The entire coding sequence of DNA32286-1191 is included in Figure 17 (SEQ ID N0:40). Clone DNA32286-1191 contains a single open reading frame with an apparent translational initiation site at nucleotide 15 positions 103-105, and an apparent stop codon at nucleotide positions 1363-1365. The predicted polypeptide precursor is 420 amino acids long. Analysis of the full-length PR02I4 sequence shown in Figure 18 (SEQ ID NO:41) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR02I4 polypeptide shown in Figure 18 evidences the presence of the following: a signal peptide from about amino acid 20 1 to about amino acid 29; a transmembrane domain from about amino acid 342 to about amino acid 392; N-glycosylation sites from about amino acid 79 to about amino acid 83, and from about amino acid 205 to about amino acid 209; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 290 to about amino acid 294: an aspartic acid and asparagine hydroxylation site from about amino acid 321 to about amino acid 333; and an EGF-Iike domain cysteine pattern signature from about amino acid 181 to about amino acid 25 193. Clone DNA32286-1191 has been deposited with the ATCC on October 16, 1997 and is assigned ATCC deposit no. 209385. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 18 (SEQ ID NO:41), PR02I4 shows amino acid sequence identity to HT protein and/or Fibulin (49% and 38%, respectively). <br><br> 30 EXAMPLE 13 <br><br> Isolation of cDNA Clones Encoding Human PRQ2I7 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA28760. Based on the assembled DNA28760 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that <br><br> contained the sequence of interest and 2) for use as probes to isolate a clone ofthe full-length coding sequence for PR0217. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 5 5-AAAGACGCATCTGCGAGTGTCC-3' (SEQ ID NO:47) <br><br> reverse PCR primer: <br><br> 5'-TGCT(jATTTCACACTGCTCTCCC-3' (SEQ ID NO:48) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA28760 consensus sequence which had the following nucleotide sequence: <br><br> 10 hybridization probe: <br><br> 5'-CCCACGATGTATGAATGGTGGACTTTGTGTGACTCCTGGTTTCTGCATC-3' (SEQ ID NO:49) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0217 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction 15 of the cDNA libraries was isolated from human fetal lung tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA33094-I131 [Figure 19, SEQ ID NO:45]; and the derived protein sequence for PR0217. <br><br> The entire coding sequence of DNA33094-1131 is included in Figure 19 (SEQ ID NO:45). Clone DNA33094-1131 contains a single open reading frame with an apparent translational initiation site at nucleotide 20 positions 146-148, and an apparent stop codon at nucleotide positions 1283-1285. The predicted polypeptide precursor is 379 amino acids long with a molecular weight of approximately 41.528 daltons and an estimated pi of about 7.97. Analysis of the full-length PR0217 sequence shown in Figure 20 (SEQ ID NO:46) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide | domains are approximate as described above. Analysis of the full-length PR0217 polypeptide shown in Figure 20 25 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 28; N-glycosylation sites from about amino acid 88 to about amino acid 92, and from about amino acid 245 to about amino acid 249; a tyrosine kinase phosphorylation site from about amino acid 370 to about amino acid 378; N-myristoylation sites from about amino acid 184 to about amino acid 190, from about amino acid 185 to about amino acid 191, from about amino acid 189 to about amino acid 195, and from about amino acid 315 to about amino acid 30 321 ; an ATP/GTP-binding site motif A (P-loop) from about amino acid 285 to about amino acid 293; and EGF-iike domain cysteine pattern signatures from about amino acid 198 to about amino acid 210, from about amino acid 230 to about amino acid 242. from about amino acid 262 to about amino acid 274. from about amino acid 294 to about amino acid 306, and from about amino acid 326 to about amino acid 338. Clone DNA33094-1131 has been deposited with the ATCC on September 16, 1997 and is assigned ATCC deposit no. 209256. <br><br> 35 Based on a BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 20 <br><br> (SEQ ID NO:46), PR0217 appears to be a novel EGF-like homologue. <br><br> EXAMPLE 14 Isolation of cDNA Clones Encoding Human PRQ224 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 5 Example 1 above. This consensus sequence is designated herein as DNA30845. Based on the assembled DNA30845 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0224. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> 10 forward PCR primer: <br><br> 5'-AAGTTCCAGTGCCGCACCAGTGGC-3' (SEQ ID NO:52) <br><br> reverse PCR primer: <br><br> 5'-TTGGTTCCACAGCCGAGCTCGTCG-3' (SEQ ID NO:53) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30845 consensus 15 sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5-GAGGAGGAGTGCAGGATTGAGCCATGTACCCAGAAAGGGCAATGCCCACC-3'(SEQ IDNO:54) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones 20 encoding the PR0224 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal liver tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA3322I-1133 [Figure 21, SEQ ID N0:50]; and the derived protein sequence for PR0224. <br><br> The entire coding sequence of DNA33221-1133 is included in Figure 21 (SEQ ID N0:50). Clone 25 DNA33221-1133 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 33-35. and an apparent stop codon at nucleotide positions 879-881. The predicted polypeptide precursor is 282 amino acids long with a molecular weight of approximately 28,991 daltons and an estimated pi of about 4.62. Analysis of the full-length PR0224 sequence shown in Figure 22 (SEQ ID NO:51) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are 30 approximate as described above. Analysis of the full-length PR0224 polypeptide shown in Figure 22 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 30; a transmembrane domain from about amino acid 231 to about amino acid 248; N-glycosylation sites from about amino acid 126 to about amino acid 130, from about amino acid 195 to about amino acid 199, and from about amino acid 213 to about amino acid 217: N-myristoylation sites from about amino acid 3 to about amino acid 9. from about amino acid 10 <br><br> 124 <br><br> to about amino acid 16, from about amino acid 26 to about amino acid 32, from about amino acid 30 to about amino acid 36, from about amino acid 112 to about amino acid 118, from about amino acid 166 to about amino acid 172, from about amino acid 212 to about amino acid 218, from about amino acid 224 to about amino acid 230, from about amino acid 230 to about amino acid 236, and from about amino acid 263 to about amino acid 269; a 5 prokaryotic membrane lipoprotein lipid attachment site from about amino acid 44 to about amino acid 55; and a leucine zipper pattern from about amino acid 17 to about amino acid 39. Clone DNA3322I-1133 has been deposited with the ATCC on September 16, 1997 and is assigned ATCC deposit no. 209263. <br><br> Analysis of the amino acid sequence of the full-length PR0224 sequence suggests that it has homology to very low-density lipoprotein receptors, apolipoprotein E receptor and chicken oocyte receptor P95. Based on a 10 BLAST and FastA sequence alignment analysis of the full-length sequence shown in Figure 22 (SEQ ID NO:51), PR0224 has amino acid identity to portions of these proteins in the range from 28% to 45%, and overall identity with these proteins of about 33%. <br><br> EXAMPLE 15 Isolation of cDNA Clones Encoding Human PRQ231 15 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in <br><br> Example 1 above. This consensus sequence is designated herein as DNA30933. wherein the encoded polypeptide showed some similarity to a putative acid phosphatase protein. Based on the assembled DNA30933 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0231. 20 A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer 1: <br><br> 5-CCAACTACCAAAGCTGCTGGAGCC-3' (SEQ ID NO:57) <br><br> forward PCR primer 2: <br><br> 5-GCAGCTCTATTACCACGGGAAGGA-3' (SEQ ID NO:58) <br><br> 25 reverse PCR primer: <br><br> 5'-TCCTTCCCGTGGTAATAGAGCTGC-3' (SEQ ID NO:59) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30933 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 30 5'-GGCAGAGAACCAGAGGCCGGAGGAGACTGCCTCTTTACAGCCAGG-3' (SEQ ID N0:60) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PR0231 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal liver tissue. <br><br> 125 <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA34434-1139 [Figure 23, SEQ ID NO:55]; and the derived protein sequence for PR0231. <br><br> The entire coding sequence of DNA34434-1139 is included in Figure 23 (SEQ ID NO:55). Clone DNA34434-1139 contains a single open reading frame with an apparent translational initiation site at nucleotide 5 positions 173-175, and an apparent stop codon at nucleotide positions 1457-1459. The predicted polypeptide precursor is 428 amino acids long with a molecular weight of approximately 48,886 daltons and an estimated pi of about 6.39. Analysis of the full-length PR0231 sequence shown in Figure 24 (SEQ ID NO:56) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0231 polypeptide shown in Figure 24 10 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 23; a cAMP-and cGMP-dependent protein kinase phosphorylation site from about amino acid 218 to about amino acid 222; a tyrosine kinase phosphorylation site from about amino acid 280 to about amino acid 288; N-myristoylation sites from about amino acid 15 to about amino acid 21, from about amino acid 117 to about amino acid 123, from about amino acid 118 to about amino acid 124, from about amino acid 179 to about amino acid 185, from about amino 15 acid 240 to about amino acid 246. and from about amino acid 387 to about amino acid 393; an amidation site from about amino acid 216 to about amino acid 220; a leucine zipper pattern from about amino acid 10 to about amino acid 32; and a histidine acid phosphatases phosphohistidine signature from about amino acid 50 to about amino acid 65. Clone DNA34434-1139 has been deposited with the ATCC on September 16, 1997 and is assigned ATCC deposit no. 209252. <br><br> 20 Analysis of the amino acid sequence of the full-length PR0231 sequence suggests that it possesses 30% and <br><br> 31% amino acid identity with the human and rat prostatic acid phosphatase precursor proteins, respectively. <br><br> EXAMPLE 16 Isolation of cDNA Clones Encoding Human PRQ235 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 25 Example 1 above. This consensus sequence is designated herein as DNA30927. Based on the assembled DNA30927 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0235. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> 30 forward PCR primer: <br><br> 5'-TGGAATACCGCCTCCTGCAG-3' (SEQIDNO:63) <br><br> reverse PCR primer: <br><br> 5'-CTTCTGCCCTTTGGAGAAGATGGC-3' (SEQ ID NO:64) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30927 consensus 35 sequence which had the following nucleotide sequence: <br><br> 126 <br><br> hybridization brobe: <br><br> 5'-GGACTCACTGGCCCAGGCCTTCAATATCACCAGCCAGGACGAT-3' (SEQ ID NO:65) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones 5 encoding the PR0235 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal liver tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA35558-1167 [Figure 25, SEQ ID NO:61]; and the derived protein sequence for PR0235. <br><br> The entire coding sequence of DNA35558-1167 is included in Figure 25 (SEQ ID NO:61). Clone 10 DNA35558-1167 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 667-669, and an apparent stop codon at nucleotide positions 2323-2325. The predicted polypeptide ^ precursor is 552 amino acids long with a molecular weight of approximately 61,674 daltons and an estimated pi ^ of about 6.95. Analysis of the full-length PR0235 sequence shown in Figure 26 (SEQ ID NO:62) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide 15 domains are approximate as described above. Analysis of the full-length PR0235 polypeptide shown in Figure 26 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 32; a transmembrane domain from about amino acid 71 to about amino acid 86; N-glycosylation sites from about amino acid 130 to about amino acid 134. from about amino acid 145 to about amino acid 149, from about amino acid 217 to about amino acid 221. and from about amino acid 380 to about amino acid 385; N-myristoylation sites from 20 about amino acid 220 to about amino acid 226, from about amino acid 319 to about amino acid 325, from about amino acid 353 to about amino acid 359, from about amino acid 460 to about amino acid 466, and from about amino acid 503 to about amino acid 509. Clone DNA35558-1167 has been deposited with the ATCC on October 16,1997 and is assigned ATCC deposit no. 209374. <br><br> Analysis of the amino acid sequence of the full-length PR0235 sequence shown in Figure 26 (SEQ ID NO:62), suggests that portions of it possess significant homology to the human, mouse and Xenopus plexin protein, thereby indicating that PR0235 may be a novel plexin protein. <br><br> EXAMPLE 17 Isolation of cDNA Clones Encoding Human PRQ245 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 30 Example 1 above. This consensus sequence is designated herein as DNA30954. wherein the polypeptide showed some structural homology to transmembrane protein receptor tyrosine kinase proteins. Based on the assembled DNA30954 consensus sequence, oligonucleotides were synthesized: I) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0245. <br><br> 35 A pair of PCR primers (forward and reverse) were synthesized: <br><br> 127 <br><br> forward PCR primer: <br><br> 5'-ATCGTTGTGAAGTTAGTGCCCC-3' (SEQ ID NO:68) <br><br> reverse PCR primer: <br><br> S'-ACCTGCG AT ATCC A ACAG A ATTG-3' (SEQ ID NO:69) <br><br> 5 Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30954 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5-GGAAGAGGATACAGTCACTCTGGAAGTATTAGTGGCTCCAGCAGTTCC-3' (SEQ ID NO:70) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened 10 by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0245 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDN A libraries was isolated from human fetal liver tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA35638-1141 [Figure 27, SEQ ID NO:66]; and the derived protein sequence for PR0245. 15 The entire coding sequence of DNA35638-1141 is included in Figure 27 (SEQ ID NO:66). Clone <br><br> DNA35638-1141 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 89-91, and an apparent stop codon at nucleotide positions 1025-1027. The predicted polypeptide precursor is 312 amino acids long with a molecular weight of approximately 34.554 daltons and an estimated pi of about 9.39. Analysis of the full-length PR0245 sequence shown in Figure 28 (SEQ ID NO:67) evidences the 20 presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0245 polypeptide shown in Figure 28 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 20; a transmembrane domain from about amino acid 237 to about amino acid 258: N-glycosylation sites from about amino acid 98 to about amino acid 102, from about amino acid 187 to about amino acid 191, from about amino 25 acid 236 to about amino acid 240. and from about amino acid 277 to about amino acid 281; N-myristoylation sites from about amino acid 182 to about amino acid 188, from about amino acid 239 to about amino acid 245, from about amino acid 255 to about amino acid 261, from about amino acid 257 to about amino acid 263, and from about amino acid 305 to about amino acid 311; and an amidation site from about amino acid 226 to about amino acid 230. Clone DNA35638-1141 has been deposited with the ATCC on September 16,1997 and is assigned ATCC deposit 30 no. 209265. <br><br> Analysis of the amino acid sequence of the full-length PR0245 sequence shown in Figure 28 (SEQ ID NO:67), suggests that a portion of it possesses 60% amino acid identity with the human c-myb protein and, therefore, may be a new member of the transmembrane protein receptor tyrosine kinase family. <br><br> 128 <br><br> EXAMPLE 18 Isolation of cDNA Clones Encoding Human PRQ26I A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA30843. Based on the assembled 5 DNA30843 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0261. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 10 5'-AA AGGTGCGTACCC AGCTGTGCC-3' (SEQIDNO.-73) <br><br> reverse PCR primer: <br><br> 5'-TCCAGTCGGCAGAAGCGGTTCTGG-3' (SEQ ID NO:74) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30843 consensus sequence which had the following nucleotide sequence: <br><br> 15 hybridization probe: <br><br> 5'-CCTGGTGCTGGATGGCTGTGGCTGCTGCCGGGTATGTGCACGGCGGCTGGG-3' (SEQ ID NO:75) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR026I gene using the probe oligonucleotide and one of the PCR primers. RNA for construction 20 of the cDNA libraries was isolated from human fetal lung tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA33473-1176 [Figure 29, SEQ ID NO:7l]; and the derived protein sequence for PR0261. <br><br> The entire coding sequence of DNA33473-1176 is included in Figure 29 (SEQ ID NO:71). Clone DNA33473-1176 contains a single open reading frame with an apparent translational initiation site at nucleotide 25 positions 10-12, and an apparent stop codon at nucleotide positions 760-762. The predicted polypeptide precursor is 250 amino acids long with a molecular weight of approximately 26,825 daltons and an estimated pi of about 8.36. Analysis of the full-length PR0261 sequence shown in Figure 30 (SEQ ID NO:72) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR026I polypeptide shown in Figure 30 evidences 30 the presence of the following: a signal peptide from about amino acid I to about amino acid 23; N-myristoylation sites from about amino acid 3 to about amino acid 9, from about amino acid 49 to about amino acid 55, from about amino acid 81 to about amino acid 87, from about amino acid 85 to about amino acid 91, from about amino acid 126 to about amino acid 132, from about amino acid 164 to about amino acid 170, from about amino acid 166 to about amino acid 172, from about amino acid 167 to about amino acid 173, from about amino acid 183 to about 35 amino acid 189, and from about amino acid 209 to about amino acid 215; an insulin-like growth factor binding <br><br> 129 <br><br> protein signature from about amino acid 49 to about amino acid 65: a von Willebrand C1 domain from about amino acid 107 to about amino acid 124: a thrombospondin 1 homology block from about amino acid 201 to about amino acid 216; and an IGF binding protein site from about amino acid 49 to about amino acid 58. Clone DNA33473-1176 has been deposited with the ATCC on October 17, 1997 and is assigned ATCC deposit no. 209391. <br><br> 5 Analysis of the amino acid sequence of the full-length PR026I sequence shown in Figure 30 (SEQ ID <br><br> NO:72), suggests that portions of it possess significant homology to CTGF, thereby indicating that PR0261 is a novel growth factor. <br><br> 9 <br><br> EXAMPLE 19 Isolation of cDNA Clones Encoding Human PRQ269 10 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in <br><br> Example 1 above. This consensus sequence is designated herein as DNA35705. Based on the assembled DNA35705 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0269. <br><br> 15 PCR primers (three forward and two reverse) were synthesized: <br><br> forward PCR primer 1: <br><br> 5-TGGAAGGAGATGCGATGCCACCTG-3' (SEQ IDNO:78) <br><br> forward PCR primer 2: <br><br> 5-TGACCAGTGGGGAAGGACAG-3' (SEQ ID NO:79) <br><br> 20 forward PCR primer 3: <br><br> 5-ACAGAGCAGAGGGTGCCTTG-3' (SEQ ID N0:80) <br><br> reverse PCR primer 1 <br><br> 5-TC AGGG AC A AGTGGTGTCTCTCCC-3' (SEQ 1D NO:81) <br><br> reverse PCR primer 2: <br><br> 25 5'-TCAGGGAAGGAGTGTGCAGTTCTG-3* (SEQIDNO:82) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA35705 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5'-ACAGCTCCCGATCTCAGTTACTTGCATCGCGGACGAAATCGGCGCTCGCT-3' (SEQ ID NO:83) <br><br> 30 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primers identified above. A positive library was then used to isolate clones encoding the PR0269 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal kidney tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for <br><br> 130 <br><br> DNA38260-1180 [Figure 31, SEQ ID NO:76); and the derived protein sequence for PR0269. <br><br> The entire coding sequence of DNA38260-1180 is included in Figure 31 (SEQ ID NO:76). Clone DNA38260-1 180 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 314-316, and an apparent stop codon at nucleotide positions 1784-1786. The predicted polypeptide 5 precursor is 490 amino acids long with a molecular weight of approximately 51.636 daltons and an estimated pi of about 6.29. Analysis of the full-length PR0269 sequence shown in Figure 32 (SEQ ID NO:77) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0269 polypeptide shown in Figure 32 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 16; a 10 transmembrane domain from about amino acid 397 to about amino acid 418; N-glycosylation sites from about amino acid 189 to about amino acid 193, and from about amino acid 381 to about amino acid 385; a glycosaminogiycan attachment site from about amino acid 289 to about amino acid 293; cAMP- and cGMP-dependent protein kinase phosphorylation sites from about amino acid 98 to about amino acid 102, and from about amino acid 434 to about amino acid 438; N-myristoylation sites from about amino acid 30 to about amino acid 36, 15 from about amino acid 35 to about amino acid 41, from about amino acid 58 to about amino acid 64. from about amino acid 59 to about amino acid 65, from about amino acid 121 to about amino acid 127, from about amino acid 151 to about amino acid 157, from about amino acid 185 to about amino acid 191, from about amino acid 209 to about amino acid 215, from about amino acid 267 to about amino acid 273, from about amino acid 350 to about amino acid 356, from about amino acid 374 to about amino acid 380, from about amino acid 453 to about amino 20 acid 459, from about amino acid 463 to about amino acid 469, and from about amino acid 477 to about amino acid 483; and an aspartic acid and asparagine hydroxylation site from about amino acid 262 to about amino acid 274. Clone DNA38260-1180 has been deposited with the ATCC on October 17, 1997 and is assigned ATCC deposit no. 209397. <br><br> Analysis of the amino acid sequence of the full-length PR0269 sequence shown in Figure 32 (SEQ ID 25 NO:77), suggests that portions of it possess significant homology to the human thrombomodulin proteins, thereby indicating that PR0269 may possess one or more thrombomodulin-like domains. <br><br> EXAMPLE 20 Isolation of cDNA Clones Encoding Human PRQ287 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 30 Example 1 above. This consensus sequence is designated herein as DNA28728 or an extended consensus sequence encoding PR0287 was assembled and which showed similarity to type 1 procollagen C-proteinase enhancer protein and type 1 procollagen C-proteinase enhancer protein precursor. Based on the assembled DNA28728 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0287. 35 A pair of PCR primers (forward and reverse) were synthesized: <br><br> 131 <br><br> forward PCR primer: <br><br> 5'-CCGATTCATAGACCTCGAGAGT-3, (SEQ ID NO:86) <br><br> reverse PCR primer: <br><br> 5'-GTCAAGGAGTCCTCCACAATAC-3* (SEQ ID NO:87) <br><br> 5 Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA28728 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5'-GTGTACAATGGCCATGCCAATGGCCAGCGCATTGGCCGCTTCTGT-3' (SEQ1DN0:88) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened 10 by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0287 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal kidney tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA39969-J185 [Figure 33, SEQ ID NO:84]; and the derived protein sequence for PR0287. 15 The entire coding sequence of DNA39969-1185 is included in Figure 33 (SEQ ID NO:84). Clone <br><br> DNA39969-1185 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 307-309, and an apparent stop codon at nucleotide positions 1552-1554. The predicted polypeptide precursor is 415 amino acids long with a molecular weight of approximately 45,716 daltons and an estimated pi of about 8.89. Analysis of the full-length PR0287 sequence shown in Figure 34 (SEQ ID NO:85) evidences the 20 presence of a variety of important polypeptide domains, wherein the locations given forthose important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0287 polypeptide shown in Figure 34 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 23; an N-glycosylation site from about amino acid 355 to about amino acid 359; a tyrosine kinase phosphorylation site from about amino acid 199 to about amino acid 208; N-myristoylation sites from about amino acid 34 to about amino 25 acid 40, from about amino acid 35 to about amino acid 41, from about amino acid 100 to about amino acid 106, from about amino acid 113 to about amino acid 119, from about amino acid 218 to about amino acid 224, from about amino acid 289 to about amino acid 295, from about amino acid 305 to about amino acid 311, from about amino acid 309 to about amino acid 315, from about amino acid 320 to about amino acid 326, and from about amino acid 330 to about amino acid 336; and a cell attachment sequence from about amino acid 149 to about amino 30 acid 152. Clone DNA39969-1185 has been deposited with the ATCC on October 17,1997 and is assigned ATCC deposit no. 209400. <br><br> Analysis of the amino acid sequence of the full-length PR0287 sequence shown in Figure 34 (SEQ ID NO:85), suggests that it may possess one or more procollagen C-proteinase enhancer protein precursor or procollagen C-proteinase enhancer protein-like domains. Based on a BLAST and FastA sequence alignment 35 analysis of the full-length sequence. PR0287 shows amino acid sequence identity to procollagen C-proteinase <br><br> 132 <br><br> enhancer protein precursor and procollagen C-proteinase enhancer protein (47% and 54%, respectively). <br><br> EXAMPLE 21 Isolation of cDNA Clones Encoding Human PRQ301 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 5 Example 1 above. This consensus sequence is designated herein as DNA35936. Based on the assembled DNA35936 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone ofthe full-length coding sequence for PR0301. <br><br> PCR primers (three forward and two reverse) were synthesized: <br><br> 10 forward PCR primer 1: <br><br> 5-TCGCGGAGCTGTGTTCTGTTTCCC-3* (SEQ IDNO:91) <br><br> forward PCR primer 2: <br><br> 5'-ACACCTGGTTCAAAGATGGG-3' (SEQ ID NO:92) <br><br> forward PCR primer 3: <br><br> 15 5'-TTGCCTT ACTC AGGTGCT AC-3' (SEQIDNO:93) <br><br> reverse PCR primer I: <br><br> 5'-TAGGAAGAGTTGCTGAAGGCACGG-3' (SEQ ID NO:94) <br><br> reverse PCR primer 2: <br><br> 5'-ACTCAGCAGTGGTAGGAAAG-3' (SEQ IDNO:95) <br><br> 20 Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA35936 consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> ( 5-TGATCGCGATGGGGACAAAGGCGCAAGCTCGAGAGGAAACTGTTGTGCCT-3' (SEQ IDNO:96) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened 25 by PCR amplification with the PCR primers identified above. A positive library was then used to isolate clones encoding the PR0301 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal kidney tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA40628-1216 (Figure 35, SEQ IDNO:89]; and the derived protein sequence for PR0301. 30 The entire coding sequence of DNA40628-1216 is included in Figure 35 (SEQ ID NO:89). Clone <br><br> DNA40628-1216 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 52-54, and an apparent stop codon at nucleotide positions 949-951. The predicted polypeptide precursor is299 amino acids long with a molecular weight of approximately 32,583 daltons and an estimated pi of about 8.29. <br><br> 133 <br><br> Analysis of the full-length PR0301 sequence shown in Figure 36 (SEQ ID N0:90) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0301 polypeptide shown in Figure 36 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 27; a transmembrane 5 domain from about amino acid 235 to about amino acid 256; an N-glycosylation site from about amino acid 185 to about amino acid 189; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 270 to about amino acid 274; N-myristoylation sites from about amino acid 105 to about amino acid 111, from about amino acid 116 to about amino acid 122, from about amino acid 158 to about amino acid 164, from about amino acid 219 to about amino acid 225, from about amino acid 237 to about amino acid 243, and from about 10 amino acid 256 to about amino acid 262. Clone DNA40628-1216 has been deposited with the ATCC on November 7, 1997 and is assigned ATCC deposit no. 209432. <br><br> Based on a BLAST and FastA sequence alignment analysis of the full-length PR0301 sequence shown in Figure 36 (SEQ ID N0:90), PR0301 shows amino acid sequence identity to A33 antigen precursor (30%) and coxsackie and adenovirus receptor protein (29%). <br><br> 15 EXAMPLE 22 <br><br> Isolation of cDNA Clones Encoding Human PRQ323 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA30875. Based on the assembled DNA30875 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that 20 contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0235. <br><br> PCR primers (two forward and one reverse) were synthesized: <br><br> forward PCR primer 1: <br><br> 5'-AGTTCTGGTCAGCCTATGTGCC-3' (SEQ ID NO:99) <br><br> 25 forward PCR primer 2: <br><br> 5'-CGTGATGGTGTCTTTGTCCATGGG-3' (SEQ ID NO: 100) <br><br> reverse PCR primer 1: <br><br> 5'-CTCCACCAATCCCGATGAACTTGG-3' (SEQ ID NO. 101) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA30875 consensus 30 sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5'-GAGCAGATTGACCTCATACGCCGCATGTGTGCCTCCTATTCTGAGCTGGA-3" (SEQ ID NO: 102) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones <br><br> 134 <br><br> encoding the PR0323 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal liver tissue (LIB6). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA35595-1228 [Figure 37, SEQ ID NO:97]; and the derived protein sequence for PR0323. Additional probes 5 were made from the DNA35595 sequence as follows for use in characterization of the DNA35595 sequence and for screening other cDNA libraries: <br><br> forward PCR primer: <br><br> 5'-TGCTGCTGCTCCAGCCTGTAACC-3' (SEQ ID NO: 103) <br><br> reverse PCR primer: <br><br> 10 5'-CTGGCCGTAGCTGAAATTGCGC-3' (SEQ ID NO: 104) <br><br> hybridization probe: <br><br> 5'-ACTCAGTAGTCCCAGCACCCAGGGCCTGCAAGAGCAGGCACGG-3' (SEQ ID NO: 105) <br><br> The entire coding sequence of DNA35595-1228 is included in Figure 37 (SEQ ID NO:97). Clone DNA35595-1228 contains a single open reading frame with an apparent translational initiation site at nucleotide 15 positions 110-112, and an apparent stop codon at nucleotide positions 1409-1411. The predicted polypeptide precursor is 433 amino acids long with a molecular weight of approximately 47,787 daltons and an estimated pi of about 6.11. Analysis of the full-length PR0323 sequence shown in Figure 38 (SEQ ID NO:98) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0323 polypeptide shown in Figure 38 20 evidences the presence of the following: N-glycosylation sites from about amino acid 58 to about amino acid 62, from about amino acid 123 to about amino acid 127, from about amino acid 182 to about amino acid 186, and from about amino acid 273 to about amino acid 277; N-myristoylation sites from about amino acid 72 to about amino acid 78, from about amino acid 133 to about amino acid 139, from about amino acid 234 to about amino acid 240, ^ from about amino acid 264 to about amino acid 270, from about amino acid 334 to about amino acid 340, and from 25 about amino acid 389 to about amino acid 395; and a renal dipeptidase active site from about amino acid 134 to about amino acid 157. Clone DNA35595-1228 has been deposited with the ATCC on December 10, 1997 and is assigned ATCC deposit no. 209528. <br><br> Analysis of the amino acid sequence of the full-length PR0323 sequence shown in Figure 38 (SEQ ID NO:98), suggests that portions of it possess significant homology to various dipeptidase proteins, thereby indicating 30 that PR0323 may be a novel dipeptidase protein. <br><br> EXAMPLE 23 Isolation of cDNA Clones Encoding Human PRQ331 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. Based on the assembled DNA consensus sequence, oligonucleotides were synthesized: 1) to <br><br> 135 <br><br> identify by PCR a cDN A library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0331. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 5 5'-GCCTTTGACAACCTTCAGTCACTAGTGG-3' (SEQ ID NO: 108) <br><br> reverse PCR primer: <br><br> 5'-CCCCATGTGTCCATG ACTGTTCCC-3' (SEQ ID NO: 109) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the DNA consensus sequence which had the following nucleotide sequence: <br><br> 10 hybridization probe: <br><br> 5'-TACTGCCTCATGACCTCTTCACTCCCTTGCATCATCTTAGAGCGG-3' (SEQ ID NO:l 10) <br><br> ^ In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0331 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction 15 of the cDNA libraries was isolated from human fetal brain tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA40981-1234 [Figure 39, SEQ ID N0:106]; and the derived protein sequence for PR033I. <br><br> The entire coding sequence of DNA40981-1234 is included in Figure 39 (SEQ ID NO: 106). Clone DNA40981-1234 contains a single open reading frame with an apparent translational initiation site at nucleotide 20 positions 812-814, and an apparent stop codon at nucleotide positions 2732-2734. The predicted polypeptide precursor is 640 amino acids long with a molecular weight of approximately 71.950 daltons and an estimated pi of about 7.12. Analysis of the full-length PR0331 sequence shown in Figure 40 (SEQ ID NO: 107) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide ^ domains are approximate as described above. Analysis of the full-length PR0331 polypeptide shown in Figure 40 25 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 44; a transmembrane domain from about amino acid 528 to about amino acid 543; N-glycosylation sites from about amino acid 278 to about amino acid 282, from about amino acid 364 to about amino acid 368, from about amino acid 390 to about amino acid 394, from about amino acid 412 to about amino acid 416, from about amino acid 415 to about amino acid 419, from about amino acid 434 to about amino acid 438, from about amino acid 442 to about 30 amino acid 446, from about amino acid 488 to about amino acid 492, and from about amino acid 606 to about amino acid 610; a cAMP- and cGMP-dependent protein kinase phosphoiylation site from about amino acid 183 to about amino acid 187; N-myristoylation sites from about amino acid 40 to about amino acid 46, from about amino acid 73 to about amino acid 79. from about amino acid 118 to about amino acid 124, from about amino acid 191 to about amino acid 197. from about amino acid 228 to about amino acid 234, from about amino acid 237 to 35 about amino acid 243, from about amino acid 391 to about amino acid 397, from about amino acid 422 to about <br><br> 136 <br><br> amino acid 428, from about amino acid 433 to about amino acid 439, and from about amino acid 531 to about amino acid 537. Clone DNA40981 -1234 has been deposited with the ATCC on November 7, 1997 and is assigned ATCC deposit no. 209439. <br><br> Analysis of the amino acid sequence of the full-length PR0331 sequence shown in Figure 40 (SEQ ID 5 NO: 107), suggests that portions of it possess significant homology to the LIG-1 protein, thereby indicating that PR0331 may be a novel LJG-1 -related protein. <br><br> EXAMPLE 24 Isolation of cDNA Clones Encoding Human PRQ356 An expressed sequence tag (EST) DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) was 10 searched and an EST (#2939340) was identified that had homology to the TIE ligand family. To clone PR0356, a human fetal lung library prepared from mRNA purchased from Clontech, Inc., (Palo Alto, CA), catalog # 6528-1 _ was used, following the manufacturer's instructions. <br><br> ^ The cDNA libraries used to isolate the cDNA clones encoding human PR0356 were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA. The cDNA was 15 primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et a!., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl. <br><br> Oligonucleotide probes based upon the above described EST sequence were then synthesized: I) to identify 20 by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0356. Forward and reverse PCR primers generally range from 20-30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel ei al., Current Protocols in Molecular Biology, supra, with the ^^5 PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. <br><br> The oligonucleotide sequences used were as follows: <br><br> 5'-TTCAGCACCAAGGACAAGGACAATGACAACT-3' (SEQ ID NO: 113) 5'-TGTGCACACTTGTCCAAGCAGTTGTCATTGTC-3' (SEQ ID NO: 114) <br><br> 30 5'-GTAGT AC ACTCCATTG AGGTTGG-3' (SEQ ID NO:l 15) <br><br> A cDNA clone was identified and sequenced in entirety. The entire nucleotide sequence of DNA47470-1130-P1 is shown in Figure 41 (SEQ ID NO: 111). Clone DNA47470-1130-P1 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 215-217, and a stop codon at nucleotide positions 1253-1255 (Figure 41; SEQ ID NO:l 11). The predicted polypeptide precursor is 346 amino 35 acids long. The full-length PR0356 protein is shown in Figure 42 (SEQ ID NO: 112). <br><br> Analysis ofthe full-length PR0356 sequence shown in Figure 42 (SEQ ID NO: 112) evidences the presence <br><br> 137 <br><br> of important polypeptide domains as shown in Figure 42, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0356 sequence (Figure 42; SEQ ID NO: 112) evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 26; N-glycosylation sites from about amino acid 58 to about amino acid 62, from about amino acid 253 5 to about amino acid 257, and from about amino acid 267 to about amino acid 271; a glycosaminogiycan attachment site from about amino acid 167 to about amino acid 171; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 176 to about amino acid 180; N-myristoylation sites from about amino acid 168 to about amino acid 174, from about amino acid 196 to about amno acid 202, from about amino acid241 to about amino acid 247, from about amino acid 252 to about amino acid 258, from about amino acid 256 to about 10 amino acid 262, and from about amino acid 327 to about amino acid 333; and a cell attachment sequence from about amino acid 199 to about amino acid 202. <br><br> Clone DNA47470-1130-P1 has been deposited with ATCC on October 28, t997 and is assigned ATCC deposit no. 209422. It is understood that the deposited clone has the actual correct sequence rather than the representations provided herein. <br><br> 15 An analysis of the Dayhoff database (version 35.45 SwissProt 35), using the ALIGN-2 sequence alignment analysis of the full-length sequence shown in Figure 42 (SEQ ID NO: 112), shows amino acid sequence identity between the PR0356 amino acid sequence and both TIE-2LI (32%) and TIE-2L2 (34%). The abbreviation "TIE" is an acronym which stands for "tyrosine kinase containing Ig and EGF homology domains" and was coined to designate a new family of receptor tyrosine kinases. <br><br> 20 EXAMPLE 25 <br><br> Isolation of cDNA Clones Encoding Human PRQ364 An expressed sequence tag (EST) DNA database (LJFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) was searched and an EST(lncyte EST no. 3003460) was identified that encoded a polypeptide which showed homology to members of the tumor necrosis factor receptor (TNFR) family of polypeptides. <br><br> &gt;25 A consensus DNA sequence was then assembled relative to Incyte EST no. 3003460 and other EST <br><br> sequences using repeated cycles of BLAST (Altshul et al., Methods in Enzvmology. 266:460-480 (1996)) and "phrap" (Phil Green, University of Washington, Seattle, Washington). This consensus sequence is herein designated "&lt;consen01 &gt;", also designated herein as DNA44825. Based upon the DNA44825 and "&lt;consen01 &gt;'"consensus sequences, oligonucleotide probes were then synthesized: 1) to identify by PCR a cDNA 30 library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0364. Forward and reverse PCR primers generally range from 20-30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In order to screen several libraries for a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel et al., Current Protocols in Molecular Biology, supra, with the PCR primer pair. A 35 positive library was then used to isolate clones encoding the gene of interest using the probe oligonucleotide and one of the primer pairs. , <br><br> 138 <br><br> The oligonucleotide sequences used were as follows: <br><br> forward PCR primer (44825.flV. <br><br> 5'-CACAGCACG&lt;jGGCGATGGG-3' (SEQ ID NO: 118) <br><br> forward PCR primer (44825.Q1: <br><br> 5 5,-GCTCTGCGTTCTGCTCTG-3, (SEQ ID NO: 119) <br><br> forward PCR primer f44825.GITR.fi: <br><br> S'-GGCACAGCACGGGGCGATGGGCGCGTTT-3' (SEQ ID NO: 120) <br><br> reverse PCR primer (44825.rI1: <br><br> 5*-CTGGTCACTGCCACCTTCCTGCAC-3' (SEQ ID NO:121) <br><br> 10 reverse PCR primer (44825.r21: <br><br> 5-CGCTGACCCAGGCTGAG-3' (SEQ ID NO: 122) <br><br> reverse PCR primer (44825.GITR.r1: <br><br> ^ 5'-GAAGGTCCCCGAGGCACAGTCGATACA-3' (SEQ ID NO: 123) <br><br> Additionally, synthetic oligonucleotide hybridization probes were constructed from the consensus DNA44825 15 sequence which had the following nucleotide sequences: <br><br> hybridization probe (44825.PI1: <br><br> 5'-GAGGAGTGCTGTTCCGAGTGGGACTGCATGTGTGTCCAGC-3' (SEQ ID NO: 124) hybridization probe (44825.GITR.pl: <br><br> 5'-AGCCTGGGTCAGCGCCCCACCGGGGGTCCCGGGTGCGGCC-3* (SEQ ID NO: 125) <br><br> 20 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR <br><br> amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PR0364 gene using the probe oligonucleotides and one of the PCR primers. <br><br> ^ RNA for construction of the cDN A libraries was isolated from human small intestine tissue (LIB231). The cDNA libraries used to isolate the cDNA clones were constructed by standard methods using commercially <br><br> 25 available reagents such as those from Invitrogen, San Diego, CA. The cDN A was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B <br><br> is a precursor of pRK5D that does not contain the Sfil site; see, Holmes etal., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl sites. <br><br> 30 DNA sequencing of the clones isolated as described above gave the full-length DNA sequence for PR0364 <br><br> [herein designated as DNA47365-1206]. The entire nucleotide sequence of DNA47365-1206 is shown in Figure 43 (SEQ ID NO: 116). Clone DNA47365-1206 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 121-123, and a stop codon at nucleotide positions 844-846 (Figure 43; SEQ ID NO: 116). The predicted polypeptide precursor is 241 amino acids long, and has an estimated molecular weight <br><br> 139 <br><br> of about 26,000 daltons, and a pi of about 6.34. The full-length PR0364 protein is shown in Figure 44 (SEQ ID NO: 117). <br><br> Analysis ofthe full-length PR0364 sequence shown in Figure 44 (SEQ ID NO: 117) evidences the presence of important polypeptide domains as shown in Figure 44. wherein the locations given for those important 5 polypeptide domains are approximate as described above. Analysis of the full-length PR0364 sequence (Figure 44; SEQ ID NO:l 17) evidences the presence ofthe following: a signal peptide from about amino acid I to about amino acid 25; a transmembrane domain from about amino acid 163 to about amino acid 183; an N-glycosylation site from about amino acid 146 to about amino acid 150; N-myristoylation sites from about amino acid 5 to about amino acid 11, from about amino acid 8 to about amno acid 14, from about amino acid 25 to about amino acid 31, 10 from about amino acid 30 to about amino acid 36, from about amino acid 33 to about amino acid 39, from about amino acid 118 to about amino acid 124, from about amino acid 122 to about amino acid 128, and from about amino acid 156 to about amino acid 162; a prokaryotic membrane lipoprotein lipid attachment site from about amino acid 166 to about amino acid 177; and a leucine zipper pattern from about amino acid 171 to about amino * acid 193. Clone DNA47365-I206 has been deposited with ATCC on November 7, 1997 and is assigned ATCC 15 deposit no. 209436. <br><br> An analysis of the full-length PR0364 sequence shown in Figure 44 (SEQ ID NO: 117), suggests that portions of it possess significant homology to members of the tumor necrosis factor receptor family, thereby indicating that PR0364 may be a novel member of the tumor necrosis factor receptor family. <br><br> A detailed review of the amino acid sequence of the full-length PR0364 polypeptide and the nucleotide 20 sequence that encodes that amino acid sequence evidences sequence homology with the mouse GITR protein reported by Nocenti et al., Proc. Natl. Acad. Sci. USA. 94:6216-6221 (1997). It is possible, therefore, that PR0364 represents the human counterpart to the mouse GITR protein reported by Nocentini el al. <br><br> EXAMPLE 26 Isolation of cDNA Clones Encoding Human PRQ526 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. An initial consensus DNA sequence was identified and herein is designated DNA39626.init. The initial consensus DNA sequence was extended using repeated cycles of BLAST and phrap to extend the initial consensus sequence as far as possible using the sources of EST sequences discussed above. The extended assembly sequence is herein designated &lt;consen01&gt;. Bas§d on the assembled &lt;consen0l&gt; DNA consensus sequence, 30 oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0526. <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 5'-TGGCTGCCCTGCAGTACCTCTACC-3' (SEQ ID NO: 128) <br><br> 35 reverse PCR primer: <br><br> 5'-CCCTGCAGGTCATTGGCAGCTAGG-3' (SEQ ID NO: 129) <br><br> 140 <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructedfrom the &lt;consenO 1 &gt;DN A consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5'-AGGCACTGCCTGATGACACCTTCCGCGACCTGGGCAACCTCACAC-3' (SEQ JD NO: 130) <br><br> 5 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0526 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal liver tissue (LIB228). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for 10 DNA44184-1319 [Figure 45, SEQ ID NO: 126]; and the derived protein sequence for PR0526. <br><br> The entire coding sequence of DNA44184-1319 is included in Figure 45 (SEQ ID NO: 126). Clone DNA44184-1319 contains a single open reading frame with an apparent translational initiation site at nucleotide V positions 514-516, and an apparent stop codon at nucleotide positions 1933-1935. The predicted polypeptide precursor is 473 amino acids long with a molecular weight of approximately 50,708 daltons and an estimated pi 15 of about 9.28. Analysis of the full-length PR0526 sequence shown in Figure 46 (SEQ ID NO: 127) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0526 polypeptide shown in Figure 46 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 26; a leucine zipper pattern from about amino acid 135 to about amino acid 157; a glycosaminogiycan attachment site from about 20 amino acid 436 to about amino acid 440; N-glycosylation sites from about amino acid 82 to about amino acid 86, from about amino acid 179 to about amino acid 183, from about amino acid 237 to about amino acid 241, from about amino acid 372 to about amino acid 376, and from about amino acid 423 to about amino acid 427; and a von Willebrand Factor type C domain from about amino acid 411 to about amino acid 427. Clone DNA44184-1319 has been deposited with the ATCC on March 26, 1998 and is assigned ATCC deposit no. 209704. ^5 Analysis of the amino acid sequence of the full-length PR0526 sequence shown in Figure 46 (SEQ ID <br><br> NO: 127), suggests that portions of it possess significant homology to the leucine repeat rich proteins including ALS, SLIT, carboxypeptidase and platelet glycoprotein V, thereby indicating that PR0526 is a novel protein which is involved in protein-protein interactions. <br><br> EXAMPLE 27 <br><br> 30 Isolation of cDNA Clones Encoding Human PRQ538 <br><br> An expressed sequence tag (EST) DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) was searched and an Incyte EST (INC3574209) was identified that had 61 % identity to murine GFRa3 ("glial-cell-line-derived neurotrophic factor family receptor alpha"). To clone the corresponding full-length cDNA for PR0538, a panel of cDNA libraries were screened with primers: <br><br> 141 <br><br> newa3.F: <br><br> 5'-GCCTCTCGCAGCCGG AG ACC-3' (SEQ ID NO: 133) newa3.R: <br><br> 5'-CAGGTGGGATCAGCCTGGCAC-3' (SEQ ID NO: 134) <br><br> 5 DNA from the libraries was screened by PCR amplification, as per Ausubel et al., Current Protocols in Molecular Biology (1995). with the PCR primer pair. A strong PCR product was identified in all libraries analyzed (fetal lung, fetal kidney, and placenta). <br><br> To isolate a cDNA clone encoding this protein, a human fetal lung-pRK5 vector library was selected and enriched for positive cDNA clones by extension of single stranded DNA from plasmid libraries grown in dug-10 /bung- host using the newa3.R primer. RNA for construction of the cDNA libraries was isolated from human fetal lung tissue. <br><br> The cDNA libraries used to isolate the cDNA clones encoding human PR0538 were constructed by standard ^ methods using commercially available reagents such as those from lnvitrogen, San Diego, CA. The cDNA was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, 15 sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl. To enrich for positive cDNA clones the primer extension reaction contained 10^1 of lOx PCR Buffer (Perkin Elmer, USA), 1 dNTP (20 mM), 1 n\ library DNA (200 ng), 1 fA primer, 86.5 //I H;0 and I ^1 of Amplitaq (Perkin Elmer, USA) added after a hot start. The reaction was 20 denatured for 1 minute at 95°C, annealed for I minute at 60°C, and then extended for 15 minutes at 72°C. The DNA was extracted with phenol/chloroform, precipitated with ethanol, and then transformed by electroporation into a DH10B host bacteria. The entire transformation mixture was plated onto 10 plates and colonies allowed to form. Colonies were lifted onto nylon membranes and screened with an oligonucleotide probe derived from the Incyte EST: <br><br> newa3. probe: <br><br> 5'-TCTCGCAGCCGGAGACCCCCTTCCCACAGAAAGCCGACTCA-3' (SEQ ID NO: 135) <br><br> Five positive clones were identified. Pure positive clones were obtained after colony purification and secondary screening. Two of the isolated clones were sequenced, designated herein as DNA48613 and DNA48614 (an alternatively spliced form of DNA48613). <br><br> 30 The entire nucleotide sequence of DNA48613-1268 is shown in Figure 47 (SEQ ID NO:131). Clone <br><br> DNA48613-1268 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 38-40, and a stop codon at nucleotide positions 1238-1240 (Figure 47; SEQ ID NO: 131). The predicted polypeptide precursor is 400 amino acids long, with an estimated molecular weight of about 44,511 daltons and a pi of about 8.15. The full-length PR0538 protein is shown in Figure 48 (SEQ ID NO: 132). 35 Analysis ofthe full-length PR0538 sequence shown in Figure 48 (SEQ ID NO: 132) evidences the presence <br><br> 142 <br><br> of important polypeptide domains as shown in Figure 48, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0538 sequence (Figure 48; SEQ ID MO: 132) evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 26; N-glycosylation sites from about amino acid 95 to about amino acid 99. from about amino acid 148 5 to about amino acid 152, and from about amino acid 309 to about amino acid 313; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 231 to about amino acid 235; N-myristoylation sites from about amino acid 279 to about amino acid 285, and from about amino acid 294 to about amino acid 300; and prokaryotic membrane lipoprotein lipid attachment sites from about amino acid 306 to about amino acid 317, and from about amino acid 379 to about amino acid 390. Clone DNA48613-1268 has been deposited with ATCC on 10 April 7, 1998 and is assigned ATCC deposit no. 209752. <br><br> As discussed below, a sequence comparison ofthe protein encoded by DNA48613 to GFRal and GFRa2 indicated that the human protein PR0538 is a new member of the GFRa receptor family, and is a human homolog ^ of murine GFRa3. Accordingly, DN A48613-1268encodes a protein designated as human GFRa3, and DNA48614 encodes its splice variant. <br><br> 15 Amino acid sequence comparisons between GFRa family members are provided below, based on a BLAST-2 <br><br> and FastA sequence alignment analysis of the full-length PR0538 polypeptide (shown in Figure 48; SEQ ID NO: 132): <br><br> Sequence Identity Between Members of the GFRa Family Proteins Compared Percent Identity <br><br> 20 rGFRal versus hGFRal 92% <br><br> rGFRa2 versus hGFRa2 94% <br><br> mGFRa3 versus hGFRa3 77% <br><br> 34% <br><br> 34% <br><br> 48% <br><br> From the sequence comparisons it can be seen that human GFRa3 is less related to its rodent homolog than is either GFRal or GFRa2. In addition. GFRa3 appears to be more distantly related to GFRal and GFRa2 than GFRal and GFRa2 are to each other. <br><br> hGFRa3 versus hGFRal hGFRa3 versus hGFRa2 hGFRal versus hGFRa2 <br><br> EXAMPLE 28 <br><br> 30 Isolation ofcDNA Clones Encoding Human PRQ7I3 <br><br> An expressed sequence tag (EST) DNA database (LIFESEQ®, Incyte Pharmaceuticals. Palo Alto, CA) was searched and an EST (Incyte EST no. 1302516) was identified that encoded a polypeptide which showed homology to VEGF. Probes based on the the Incyte EST no. 1302516 were used to screen a cDNA library derived from the human glioma cell line G61. In particular, Incyte clone INC 1302516 was used to generate the following four <br><br> 143 <br><br> probes: <br><br> 5'-ACTTCTCAGTGTCCATAAGGG-3' (SEQ ID NO: 138) <br><br> S'-GAACTAAAGAGAACCGATACCATTTTCTGGCCAGGTTGTC-S' (SEQ ID NO: 139) 5'-CACCACAGCGTTTAACCAGG (SEQ ID N0:140) <br><br> 5 5'-ACA ACAGGCACAGTTCCCAC-3' (SEQ ID NO: 141) <br><br> Nine positives were identified and characterized. Three clones contained the full coding region and were identical in sequence. Partial clones were also identified from a fetal lung library and were identical with the glioma-derived sequence with the exception of one nucleotide change which did not alter the encoded amino acid. <br><br> DNA sequencing of the clones isolated as described above gave the full-length DNA sequence for PR0713 10 [herein designated as DNA29I01-1122], The entire nucleotide sequence of DNA2910I-1122 is shown in Figure 49 (SEQ ID NO: 136). Clone DN A29101 -1122 contains a single open reading frame with an apparent translational - initiation site at nucleotide positions 285-287, and a stop codon at nucleotide positions 1320-1322 (Figure 49; SEQ ^ ID NO: 136). The predicted polypeptide precursor is 345 am ino acids long, and has an estimated molecular weight of about 39,029 daltons, and a pi of about 6.06. The full-length PR0713 protein is shown in Figure 50 (SEQ ID 15 NO: 137). <br><br> Analysis ofthe full-length PR0713 sequence shown in Figure 50 (SEQ ID NO: 13 7) evidences the presence of important polypeptide domains as shown in Figure 50, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0713 sequence (Figure 50; SEQ ID N0:137) evidences the presence of the following: a signal peptide from about amino acid 1 to about 20 amino acid 14; N-glycosylation sites from about amino acid 25 to about amino acid 29, from about amino acid 55 to about amino acid 59, and from about amino acid 254 to about amino acid 258; N-myristoylation sites from about amino acid 15 to about amino acid 21. from about amino acid 117 to about amno acid 123, from about amino acid 127 to about amino acid 133. from about amino acid 281 to about amino acid 287. from about amino acid 282 to about amino acid 288, and from about amino acid 319 to about amino acid 325: and an amidation site from about amino acid 229 to about amino acid 233. Clone DNA29101-1122 has been deposited with ATCC on March 5, 1998 and is assigned ATCC deposit no. 209653. <br><br> An analysis of the full-length PR0713 sequence shown in Figure 50 (SEQ ID NO: 137), suggests that it is a novel VEGF-related protein (VEGF-E). <br><br> EXAMPLE 29 <br><br> 30 Isolation of cDNA Clones Encoding Human PRQ719 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA44851. which also corresponds exactly to Incyte EST clone no. 179903. Based on the DNA44851 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDN A library that contained the sequence of interest, and 2) for use as probes to isolate a 35 clone of the full-length coding sequence for PR0719. <br><br> 144 <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (44851 .fl): <br><br> 5'-GTGAGCATGAGCGAGCCGTCCAC-3' (SEQ ID NO: 144) <br><br> reverse PCR primer (4485 l.rl): <br><br> 5 5'-GCTATTACAACGGTTCTTGCGGCAGC-3' (SEQ ID NO: 145) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA44851 sequence which had the following nucleotide sequence: <br><br> hybridization probe (44851.pH: <br><br> 5'-TTGACTCTCTGGTGAATCAGGACAAGCCGAGTTTTGCCTTCCAG-3' (SEQ ID NO: 146) <br><br> 10 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones ^ encoding the PR07I9 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human placenta tissue (L1B90). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for 15 DNA49646-1327 [Figure 51. SEQ ID NO:142]; and the derived protein sequence for PR07I9. <br><br> The entire coding sequence of DNA49646-1327 is included in Figure 51 (SEQ ID NO: 142). Clone DNA49646-1327 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 223-225. and an apparent stop codon at nucleotide positions 1285-1287. The predicted polypeptide precursor is 354 amino acids long, and has an estimated molecular weight of about 39,362 daltons and a pi of about 20 8.35. Analysis ofthe full-length PR0719 sequence shown in Figure 52 (SEQ ID NO: 143) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0719 polypeptide shown in Figure 52 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 16; N-glycosylation sites from about amino acid 80 to about amino acid 84, and from about amino acid 136 to about amino acid 140; cAMP- and cGMP-dependent protein kinase phosphorylation sites from about amino acid 206 to about amino acid 210, and from about amino acid 329 to about amino acid 333; N-myristoylation sites from about amino acid 63 to about amino acid 69, from about amino acid 96 to about amino acid 102, from about amino acid 171 to about amino acid 177, from about amino acid 191 to about amino acid 197, from about amino acid 227 to about amino acid 233, from about amino acid 251 to about amino acid 257, from about amino acid 306 to about amino acid 312, and from 30 about amino acid 346 to about amino acid 352; and a lipases, serine active site from about amino acid 163 to about amino acid 173. Clone DNA49646-1327 has been deposited with the ATCC on March 26, 1998 and is assigned ATCC deposit no. 209705. <br><br> Analysis of the amino acid sequence of the full-length PR0719 polypeptide suggests that it possesses significant sequence similarity to the lipoprotein lipase H protein, thereby indicating that PR0719 may be a novel 35 lipoprotein lipase homolog. More specifically, an analysis ofthe Dayhoff database (version 35.45 SwissProt 35) <br><br> 145 <br><br> evidenced significant homology between the PR07I9 amino acid sequence and the following Dayhoff sequences: L1PLHUMAN, LIPHHUMAN, D83548J, A24059J, P_R30740, D88666_l, A43357, A46696, B43357 and A49488. <br><br> EXAMPLE 30 <br><br> 5 Isolation of cDNA Clones Encoding Human PRQ771 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example I above. This consensus sequence is designated herein as &lt;consen0l&gt;. Based on the &lt;consen01&gt; DNA consensus sequence, oligonucleotides were synthesized: I) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0771. 10 A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 5'-CAGCAATATTCAGAAGCGGCAAGGG-3' (SEQ ID NO: 149) <br><br> ^ reverse PCR primer: <br><br> 5-CATCATGGTCATCACCACCATCATCATC-3' (SEQ ID NO: 150) <br><br> 15 Additionally, a synthetic oligonucleotide hybridization probe was constructed from the &lt;consen01&gt; DNA consensus sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 5-GGTTACTACAAGCCAACACAATGTCATGGCAGTGTTGGACAGTGCTGG-3' (SEQ ID NO: 151) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened 20 by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PR0771 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human fetal kidney tissue (LIB28). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA49829-1346 [Figure 53, SEQ ID NO:I47]; and the derived protein sequence for PR0771. 25 The entire coding sequence of DNA49829-1346 is included in Figure 53 (SEQ ID NO:147). Clone <br><br> DNA49829-1346 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 134-136, and an apparent stop codon at nucleotide positions 1442-1444. The predicted polypeptide precursor is 436 amino acids long, and has an estimated molecular weight of about 49,429 daltons and a pi of about 4.80. Analysis of the full-length PR0771 sequence shown in Figure 54 (SEQ ID NO: 148) evidences the presence 30 of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0771 polypeptide shown in Figure 54 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 16; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 115 to about amino acid 119; a tyrosine k inase phosphorylation site from about amino acid 62 to about amino acid 70; N-myristoylation sites from <br><br> 146 <br><br> about amino acid 357 to about amino acid 363, from about amino acid 371 to about amino acid 377, and from about amino acid 376 to about amino acid 382; and a leucine zipper pattern from about amino acid 246 to about amino acid 268. Clone DNA49829-1346 has been deposited with the ATCC on April 7, 1998 and is assigned ATCC deposit no. 209749. <br><br> 5 Analysis ofthe amino acid sequence ofthe full-length PR0771 polypeptide suggests that portions of it possess significant sequence homology to the testican protein, thereby indicating that PR0771 may be a novel testican homologue. <br><br> EXAMPLE 31 Isolation of cDNA Clones Encoding Human PRQ788 10 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as "&lt;consen01&gt;". Based on the data provided herein, ^ Incyte EST 2777282 was identified which showed homology to ARS and E48 anitigen. Based on the assembled P "&lt;consenO 1 &gt;" sequence and other data provided herein, Incyte EST 2777282 was obtained and sequenced in full which gave the full-length DNA sequence for PR0788 herein designated as DN A56405-1357 (Figure 55; SEQ ID 15 NO: 152), and the derived protein sequence for PR0788. <br><br> The entire coding sequence of DNA56405-1357 is included in Figure 55 (SEQ ID NO: 152). Clone DNA56405-1357 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 84-86, and an apparent stop codon at nucleotide positions 459-461. The predicted polypeptide precursor is 125 amino acids long, and has an estimated molecular weight of about 13,115 daltons and a pi of about 5.90. 20 Analysis of the full-length PR0788 sequence shown in Figure 56 (SEQ ID NO: 153) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PR0788 polypeptide shown in Figure 56 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 17; an N-glycosylation • site from about amino acid 46 to about amino acid 50; N-myristoylation sites from about amino acid 3 to about J2S amino acid 9, from about amino acid 33 to about amino acid 39, and from about amino acid 84 to about amino acid 90: and a prokaryotic membrane lipoprotein lipid attachment site from about amino acid 6 to about amino acid 17. Clone DNA56405-1357 has been deposited with the ATCC on May 6, 1998 and is assigned ATCC deposit no. 209849. <br><br> EXAMPLE 32 <br><br> 30 Isolation of cDNA Clones Encoding Human PRQ792 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA38I06, which also corresponds exactly to Incyte EST clone no. 1988930. Based on the DNA38106 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes 35 to isolate a clone of the full-length coding sequence for PR0792. <br><br> 147 <br><br> A pair of PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (38106.fl): <br><br> 5'-GCGAGAACTGTGTCATGATGCTGC-3' (SEQ ID NO: 156) <br><br> reverse PCR primer (38106,rl V 5 5-GTTTCTGAGACTCAGCAGCGGTGG-3' (SEQ ID NO: 157) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA38106 sequence which had the following nucleotide sequence: <br><br> hybridization probe (38106.P 1): <br><br> 5-CACCGTGTGACAGCGAGAAGGACGGCTGGATCTGTGAGAAAAGGCACAAC-3' (SEQ ID NO: 158) <br><br> 10 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones )) encoding the PR0792 gene using the probe oligonucleotide and one ofthe PCR primers. RNA for construction of the cDNA libraries was isolated from human bone marrow tissue (LIB255). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for 15 DNA56352-I358 [Figure 57. SEQ ID NO: 154]; and the derived protein sequence for PR0792. <br><br> The entire coding sequence of DNA56352-1358 is included in Figure 57 (SEQ ID NO:I54). Clone DNA56352-1358 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 67-69,and an apparent stop codon at nucleotide positions 946-948. The predicted polypeptide precursor is 293 amino acids long, and has an estimated molecular weight of about 32,562 daltons and a pi of about 6.53. 20 Analysis of the full-length PR0792 sequence shown in Figure 58 (SEQ ID NO: 155) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0792 polypeptide shown in Figure 58 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 46; a possible type II transmembrane domain from about amino acid 31 to about amino acid 54; N-glycosylation sites from about amino 25 acid 73 to about amino acid 77, and from about amino acid 159 to about amino acid 163; N-myristoylation sites from about amino acid 18 to about amino acid 24, from about amino acid 133 to about amino acid 139, and from about amino acid 242 to about amino acid 248; a C-type lectin domain signature from about amino acid 264 to about amino acid 288; and a leucine zipper pattern from about amino acid 102 to about amino acid 124. Clone DNA56352-1358 has been deposited with the ATCC on May 6, 1998 and is assigned ATCC deposit no. 209846. 30 Analysis of the amino acid sequence of the full-length PR0792 polypeptide suggests that it possesses significant sequence similarity to the CD23 protein, thereby indicating that PR0792 may be a novel CD23 homolog. More specifically, an analysis of the Dayhoff database (version 35.45 SwissProt 35) evidenced significant homology between the PR0792 amino acid sequence and the following Dayhoff sequences: S34198, A07100-I, A05303 1, P_R41689. P P82839, A10871 J, P R 12796, P_R47199, A46274 and PR32188. <br><br> 148 <br><br> EXAMPLE 33 Isolation of cDNA Clones Encoding Human PRQ812 DN A59205-1421 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster 5 sequence from the LIFESEQ8 database, designated Incyte EST cluster no. 170079. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank)and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) lo identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 (Altshul etal, Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score 10 of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as DNA55721. <br><br> ^ In light of the sequence homology between the DNA55721 sequence and Incyte EST no. 388964, Incyte EST <br><br> ^ no. 388964 was purchased and the cDNA insert was obtained and sequenced. The sequence of this cDNA insert 15 is shown in Figure 59 (SEQ ID NO: 159) and is herein designated as DNA59205-1421. <br><br> The entire coding sequence of DNA59205-142I is included in Figure 59 (SEQ ID NO:159). Clone DNA59205-1421 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 55-57 and ending at the stop codon at nucleotide positions 304-306 (Figure 59). The predicted polypeptide precursor is 83 amino acids long (Figure 60; SEQ ID NO: 160). The full-length PR0812 protein shown 20 in Figure 60 has an estimated molecular weight of about 9,201 daltons and a pi of about 9.30. Analysis of the full-length PR0812 sequence shown in Figure 60 (SEQ ID NO: 160) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0812 sequence shown in Figure 60 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 15; and a cAMP- and cGMP-dependent 25 protein kinase phosphorylation site from about amino acid 73 to about amino acid 77. Clone DNA59205-1421 ^ has been deposited with ATCC on June 23, 1998 and is assigned ATCC deposit no. 203009. <br><br> An analysis ofthe Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 60 (SEQ ID NO: 160), evidenced significant homology between the PR0812 amino acid sequence and the following Dayhoff sequences: P W35802, P_W35803, 30 PSC1 _RAT, S68231, GEN 13917, PSC2_RAT, CC10_HUMAN, UTER RABIT, AF008595J, and A56413. <br><br> EXAMPLE 34 Isolation of cDNA Clones Encoding Human PRQ865 A cDNA sequence isolated in the amylase screen described in Example 2 above is herein designated DNA37642 or DN A37642.init. The DNA37642 sequence was then compared to a variety of expressed sequence 35 tag (EST) databases which included public EST databases (eg., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals. Palo Alto, CA) to identify homologies there between. The homology search <br><br> 149 <br><br> was performed using the computer program BLAST or BLAST2 (Altshul etal., Methods in Enzvmology. 266:46-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program uphrap" (Phil Green, University of Washington, Seattle, Washington). The assembly and the consensus sequence obtained 5 is herein designated DNA48615. <br><br> Based on the DNA48615 consensus sequence, probes were generated from the sequence of the DNA48615 molecule and used to screen a human fetal kidney (LIB227) library prepared as described in paragraph 1 of Example 2 above. The cloning vector was pRK5B (pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al., Science. 253:1278-1280 (1991)), and the cDNA size cut was less than 2800 bp. 10 PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer 1 (486l5.fl): <br><br> S'-AAGCTGCCGGAGCTGCAATG-3' (SEQ ID NO: 163) <br><br> forward PCR primer 2 (48615.f2): <br><br> ^ 5'TTGCTTCTTAATCCTGAGCGC-3' (SEQ ID NO: 164) <br><br> 15 forward PCR primer 3 (48615.f3): <br><br> 5VAAAGGAGGACTTTCGACTGC-3' (SEQ ID NO: 165) <br><br> reverse PCR primer 1 (48615x11: <br><br> S'-AGAGATTCATCCACTGCTCCAAGTCG-3' (SEQ ID NO: 166) <br><br> reverse PCR primer 2 (48615.r2): <br><br> 20 5'-TGTCCAGAAACAGGCACATATCAGC-3' (SEQ ID NO: 167) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA48615 sequence which had the following nucleotide sequence: <br><br> hybridization probe (48615.pl): <br><br> 5'-AGACAGCGGCACAGAGGTGCTTCTGCCAGGTTAGTGGTTACTTGGATGAT-3' (SEQ ID NO: 168) <br><br> 25 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PR0865 gene using the probe oligonucleotide and one of the PCR primers. <br><br> A full length clone [DNA53974-I401] was identified that contained a single open reading frame with an apparent translational initiation site at nucleotide positions 173-175, and a stop signal at nucleotide positions 1577- <br><br> 30 1579 (Figure 61; SEQ ID NO:161). The predicted polypeptide precursor is 468 amino acids long, and has a calculated molecular weight of approximately 54,393 daltons and an estimated pi of approximately 5.63, Analysis ofthe full-length PR0865 sequence shown in Figure 62 (SEQ ID NO: 162) evidences the presence of a variety of important polypeptide domains as shown in Figure 62, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR0865 polypeptide shown in Figure <br><br> 150 <br><br> 62 evidences the presence of the following: a signal peptide from about amino acid I toaboutamino acid 23; N-glycosylation sites from about amino acid 280 to about amino acid 284. and from about amino acid 384 to about amino acid 388; an amidation site from about amino acid 94 to about amino acid 98; glycosaminogiycan attachment sites from about amino acid 20 to about amino acid 24, and from about amino acid 223 to about amino acid 227; 5 an aminotransferases class-V pyridoxal-phosphate site from about amino acid 216 to about amino acid 223; and an interleukin-7 protein site from about amino acid 338 to about amino acid 344. Clone DNA53974-1401 was deposited with the ATCC on April 14, 1998 , and is assigned ATCC deposit no. 209774. <br><br> Analysis of the amino acid sequence of the full-length PR0865 (Figure 62; SEQ ID NO: 162) polypeptide suggests that it possesses no significant similarity to any known protein. However, an analysis of the Dayhoff 10 database (version 35.45 SwissProt 35) evidenced some degree of homology between the PR0865 amino acid sequence and the following Dayhoff sequences: YMNO YEAST. ATFCA4 43, S44168, P W14549 and RABTCRG41. <br><br> ^ EXAMPLE 35 <br><br> Isolation of cDNA Clones Encoding Human PROl 075 15 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in <br><br> Example 1 above. This consensus sequence is designated herein as DNA34363. ESTs proprietary to Genentech were employed in the consensus assembly. The Genentech ESTs are herein designated DNA 13059 and DNA 19463. Based on the DNA34363 consensus sequence, oligonucleotides were synthesized: I) to identify by PCR a cDNA library that contained the sequence of Interest, and 2) for use as probes to isolate a clone of the full-20 length coding sequence for PROl 075. <br><br> PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (34363.fl): <br><br> 5'-TG AGAGGCCTCTCTGGAAGTTG-3' (SEQ ID NO: 171) <br><br> forward PCR primer (34363.12): <br><br> 25 5'-GTCAGCGATCAGTGAAAGC-3' (SEQ ID NO: 172) <br><br> forward PCR primer (34363.QV. <br><br> 5-CCAGAATGAAGTAGCTCGGC-3' (SEQ ID NO: 173) <br><br> forward PCR primer (34363.f4): <br><br> 5'-CCGACTCAAAATGCATTGTC-3' (SEQ ID NO: 174) <br><br> 30 forward PCR primer (34363.f5): <br><br> 5'-CATTTGGCAGGAATTGTCC-3' (SEQ ID NO: 175) <br><br> forward PCR primer (34363.f6V. <br><br> 5'-GGTGCTATAGGCCAAGGG-3' (SEQ ID NO: 176) <br><br> reverse PCR primer (34363.rl): <br><br> 35 5'-CTGTATCTCTGGGCTATGTCAGAG-3' (SEQ ID NO: 177) <br><br> 151 <br><br> reverse PCR primer f34363,r2): . 5-CTACATATAATGGCACATGTCAGCC-3' <br><br> (SEQ ID NO: 178) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA34363 sequence which had the following nucleotide sequence: <br><br> 5 hybridization probe (34363.pl): <br><br> 5'-CGTCTTCCTATCCTTACCCGACCTCAGATGCTCCCTTCTGCTCCTG-3' (SEQ ID NO: 179) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PRO 1075 gene using the probe oligonucleotide and one of the PCR primers. RNA for 10 construction of the cDNA libraries was isolated from human skin tumor tissue (LIB324). <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for ^ DNA57689-1385 [Figure 63. SEQ ID NO: 169]; and the derived protein sequence for PROl 075. <br><br> The entire coding sequence of DNA57689-1385 is included in Figure 63 (SEQ ID NO: 169). Clone DNA57689-1385 contains a single open reading frame with an apparent translational initiation site at nucleotide 15 positions 137-139, and an apparent stop codon at nucleotide positions 1355-1357. The predicted polypeptide precursor is 406 amino acids long, and has an estimated molecular weight of about 46,927 daltons and a pi of about 5.21. Analysis ofthe full-length PRO 1075 sequence shown in Figure 64 (SEQ ID NO: 170) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR01075 polypeptide shown in Figure 64 20 evidences the presence of the following: a signal peptide from about amino acid 1 to about am ino acid 29; a tyrosine kinase phosphorylation site from about amino acid 203 to about amino acid 212; a N-myristoylation site from about amino acid 225 to about amino acid 231: and an endoplasmic reticulum targeting sequence from about amino acid 403 to about amino acid 408. Clone DNA57689-1385 has been deposited with the ATCC on May 14,1998 and is assigned ATCC deposit no. 209869. <br><br> 25 Analysis of the amino acid sequence of the full-length PR01075 polypeptide suggests that it possesses significant sequence similarity to protein disulfide isomerase, thereby indicating that PR01075 may be a novel protein disulfide isomerase. More specifically, an analysis of the Dayhoff database (version 35.45 SwissProt 35) evidenced significant homology between the PROl 075 amino acid sequence and the following Dayhoff sequences: CELC30H7 2, CELC06A6_3, CELF42G8J3, S57942, ER72 CAEEL, CELC07A12_3, CEH0600I_4 and 30 P_R51696. <br><br> EXAMPLE 36 Isolation of cDNA Clones Encoding Human PROl 126 <br><br> DNA60615-1483 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster <br><br> 152 <br><br> sequence from the LIFESEQ® database, designated Incyte EST cluster no. 121249. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, GA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 5 (Altshul et al.. Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as DNA56250. <br><br> In light of the sequence homology between the DNA56250 sequence and Incyte EST clone no. 1437250, 10 Incyte EST no. 1437250 was purchased and the cDNA insert was obtained and sequenced. The sequence of this cDNA insert is shown in Figure 65 (SEQ ID NO: 180) and is herein designated as DNA60615-1483. <br><br> The entire coding sequence of DNA60615-1483 is included in Figure 65 (SEQ ID NO: 180). Clone ^ DNA60615-1483 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 110-112 and ending at the stop codon at nucleotide positions 1316-1318 (Figure 65). The predicted 15 polypeptide precursor is 402 amino acids long (Figure 66; SEQ ID NO: 181). The full-length PROl 126 protein shown in Figure 66 has an estimated molecular weight of about 45,921 daltons and a pi of about 8.60. Analysis of the full-length PROl 126 sequence shown in Figure 66 (SEQ ID NO: 181) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PROl 126 sequence shown in Figure 66 evidences the 20 presence of the following: a signal peptide from about amino acid 1 to about amino acid 25; and N-glycosylation sites from about amino acid 66 to about amino acid 70, from about amino acid 138 to about amino acid 142, and from about amino acid 183 to about amino acid 187. Clone DN A60615-1483 has been deposited with ATCC on June 16, 1998 and is assigned ATCC deposit no. 209980. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment 25 analysis of the full-length sequence shown in Figure 66 (SEQ ID NO: 181), evidenced significant homology between the PROl 126 amino acid sequence and the following Dayhoff sequences: 173636, NOMR HUMAN. MMUSMYOC31, HS454G6J, P_R98225, RNU78105J, RNU72487J, AF035301 J, CEELC48E7_4, and CEF11C33. <br><br> EXAMPLE 37 <br><br> 30 Isolation of cDNA Clones Encoding Human PROl 130 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example I above. This consensus sequence is designated herein as DNA34360. Based on the DNA34360 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PROl 130. 35 PCR primers (forward and reverse) were synthesized: <br><br> 153 <br><br> forward PCR primer C34360.fl'): <br><br> 5,-GCCATAGTCAGGACATGGATG-3, (SEQIDNO:184) <br><br> forward PCR primer (34360.f2): <br><br> S'-GGATGGCCAGAGCTGCTG-S' (SEQIDNO:185) <br><br> 5 forward PCR primer (3436Q.f3): <br><br> 5'-AAAGTACAAGTGTGGCCTC ATCAAGC-3' (SEQ ID NO:186) <br><br> reverse PCR primer (34360.rl): <br><br> 5'-TCTGACTCCTAAGTCAGG€AGGAG-3' (SEQ ED NO:187) <br><br> reverse PCR primer (34363.r2): <br><br> 10 5'-ATTCTCTCCACAGACAGCTGGTTC-3' (SEQIDNO:188) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA34360 sequence which had the following nucleotide sequence: <br><br> hybridization probe (34360.01"): <br><br> 5'-GTACAAGTGTGGCCTCATCAAGCCCIX3CCCAGCCAACTACTTTGCG-3' (SEQ ID NO:189) <br><br> 15 In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PRO 1130 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human aortic endothelial cell tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for 20 DNA59814-1486 [Figure 67, SEQ ID NO: 182]; and the derived protein sequence for PROl 130. <br><br> The entire coding sequence of DNA59814-1486 is included in Figure 67 (SEQ ID NO:182). Clone DNA59814-1486 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 312-314, and an apparent stop codon at nucleotide positions 984-986. The predicted polypeptide precursor is 224 amino acids long, and has an estimated molecular weight of about24,963 daltons and a pi of about 25 9.64. Analysisofthefull-lengthPRO1130sequenceshowninFigure68(SEQIDNO:183)evidencesthepresence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PROl 130 polypeptide shown in Figure 68 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 15; an ATP/GTP-binding site motif A (P-loop) from about amino acid 184 to about amino acid 192; and an N-30 glycosylation site fromabout amino acid 107 to about amino acid 111. CloneDNA59814-1486hasbeendeposited with the ATCC on October 20,1998 and is assigned ATCC deposit no. 203359. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 68 (SEQ ID NO: 183), evidenced significant homology between the PROl 130 amino acid sequence and the following Dayhoff sequences: P_W06547,216JHUMAN, D87120_l, 35 MMU72677_1, LAU04889_1, and D69319. <br><br> 154 <br><br> EXAMPLE 38 Isolation of cDNA Clones Encoding Human PROl 154 <br><br> DNA59846-1503 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster 5 sequence from the LIFESEQ® database. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 (Altshiil et al.. Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that 10 did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as DNA56250. <br><br> In light of the discoveries provided herein, the Incyte clone which included EST 2169375 (library 309-ENDCNOTOS-from a microvascular endothelial cell library) was purchased and further examined and sequenced. 15 The sequence of this cDN A insert is shown in Figure 69 (SEQ ID NO: 190) and is herein designated as DNA59846-1503. <br><br> The entire coding sequence of DNA59846-1503 is included in Figure 69 (SEQ ID NO: 190). Clone DNA59846-1503 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 86-88 and ending at the stop codon at nucleotide positions 2909-2911 (Figure 69). The predicted 20 polypeptide precursor is 941 amino acids long (Figure 70; SEQ IDNO:19I). The full-length PROl 154 protein shown in Figure 70 has an estimated molecular weight of about 107,144 daltons and a pi of about 6.26. Analysis of the full-length PROl 154 sequence shown in Figure 70 (SEQ ID NO: 191) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PROl 154 sequence shown in Figure 70 evidences the 25 presence of the following: a signal peptide from about amino acid I to about amino acid 34; N-glycosylation sites { from about amino acid 70 to about amino acid 74, from about amino acid 154 to about amino acid 158, from about amino acid 414 to about amino acid 418, from about amino acid 760 to about amino acid 764, and from about amino acid 901 to about amino acid 905; and a neutral zinc metallopeptidases, zinc-binding region signature from about amino acid 350 to about amino acid 360. Clone DNA59846-1503 has been deposited with ATCC on June 30 16, 1998 and is assigned ATCC deposit no. 209978. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis ofthe full-length sequence shovfrn in Figure 70 (SEQ lDNO:19l). evidenced sequence identity between the PROI154 amino acid sequence and the following Dayhoff sequences: AB0110971, AMPNHUMAN, RNU76997I, 159331, GEN 14047, HSU62768J, PR51281, CET07FIOJ, SSU66371J, and 35 AMPRE HUMAN. <br><br> 155 <br><br> EXAMPLE 39 Isolation of cDNA Clones Encoding Human PRO 1244 DNA64883-1526 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster 5 sequence from the LIFESEQ® database, designated Incyte EST cluster no. 7874. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. One or more of the ESTs was derived from a library constructed from tissue of the corpus cavernosum. The homology search was performed using the computer program BLAST or BLAST2 (Altshul et 10 al., Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The assembly included EST sequences designated "DNA18313", "DNA22812", and Incyte EST no. 3202349. The consensus sequence obtained therefrom is herein designated as DNA56011. <br><br> 15 In light of the sequence homology between the DNA56011 sequence and Incyte EST clone no. 3202349, <br><br> Incyte EST no. 3202349 was purchased and the cDNA insert was obtained and sequenced. The sequence of this cDNA insert is shown in Figure 71 (SEQ ID NO: 192) and is herein designated as DNA64883-1526. <br><br> The entire coding sequence of DNA64883-1526 is included in Figure 71 (SEQ ID NO: 192). Clone DNA64883-1526 contains a single open reading frame with an apparent translational initiation site at nucleotide 20 positions 9-11 and ending at the stop codon at nucleotide positions 1014-1016 (Figure 71). The predicted polypeptide precursor is 335 amino acids long (Figure 72; SEQ ID NO:193). The full-length PR01244 protein shown in Figure 72 has an estimated molecular weight of about 38,037 daltons and a pi of about 9.87. Analysis ofthe full-length PRO 1244 sequence shown in Figure 72 (SEQ ID NO: 193) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are 25 approximate as described above. Analysis of the full-length PR01244 sequence shown in Figure 72 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 29; transmembrane domains from about amino acid 183 to about amino acid 205, from about amino acid 217 to about amino acid 237, from about amino acid 217 to about amino acid 287, and from about amino acid 301 to about amino acid 321; N-glycosylation sites from about amino acid 71 to about amino acid 75, and from about amino acid 215 to about 30 amino acid 219; and a cell attachment sequence from about amino acid 150 to about amino acid 153. Clone DNA64883-1526 has been deposited with ATCC on September 9,1998 and is assigned ATCC deposit no. 203253. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), usinga WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 72 (SEQ ID NO: 193), revealed homology between the PR01244 amino acid sequence and the following Dayhoff sequences: AF008554 1, P485334, G02297, 35 HUMN33SI11, HUMN33S10 I. YOI3_CAEEL, GEN13255, S49758, E70107, and ERP5 MEDSA. <br><br> 156 <br><br> EXAMPLE 40 Isolation of cDNA Clones Encoding Human PRO 1246 DNA64885-1529 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster 5 sequence from the LIFESEQ® database, designated Incyte EST cluster no. 56853. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 (Altshul el al., Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score 10 of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as DNA56021. <br><br> In light of the sequence homology between the DNA56021 sequence and Incyte EST clone no. 2481345, Incyte EST no. 2481345 was purchased and the cDNA insert was obtained and sequenced. The sequence of this 15 cDNA insert is shown in Figure 73 (SEQ ID NO: 194) and is herein designated as DNA64885-1529. <br><br> The entire coding sequence of DNA64885-1529 is included in Figure 73 (SEQ ID NO: 194). Clone DNA64885-1529 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 119-121 and ending at the stop codon at nucleotide positions 1727-1729 (Figure 73). The predicted polypeptide precursor is 536 amino acids long (Figure 74; SEQ ID NO: 195). The full-length PRO 1246 protein 20 shown in Figure 74 has an estimated molecular weight of about 61,450 daltons and a pi of about 9.17. Analysis of the full-length PRO 1246 sequence shown in Figure 74 (SEQ ID NO: 195) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 1246 sequence shown in Figure 74 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 15; N-glycosylation sites 25 from about amino acid 108 to about amino acid 112, from about amino acid 166 to about amino acid 170. from | about amino acid 193 to about amino acid 197, from about amino acid 262 to about amino acid 266, from about amino acid 375 to about amino acid 379, from about amino acid 413 to about amino acid 417, and from about amino acid 498 to about amino acid 502; and sulfatase proteins homology blocks from about amino acid 286 to about amino acid 317, from about amino acid 359 to about amino acid 370, and from about amino acid 78 to about 30 amino acid 98. Clone DNA64885-1529 has been deposited with ATCC on November 3, 1998 and is assigned ATCC deposit no. 203457. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 74 (SEQ ID NO: 195), evidenced significant homology between the PR01246 amino acid sequence and the following Dayhoff sequences: P R51355. CELK09C41, 35 BCU44852J.IDS HUMAN,G65169,E64903,ARSA_HUMAN,GL6S_HUMAN,HSARSF_1,and GEN 12648. <br><br> 157 <br><br> EXAMPLE 41 Isolation of cDNA Clones Encoding Human PRQ1274 <br><br> A novel secreted molecule, DNA57700, was used to BLAST against Incyte's (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) proprietary database and GenBank's public database. Positive clones were 5 identified and used to generate assembly files by seqext program. The homology search was performed using the computer program BLAST or BLAST2 (Altshul et a!.. Methods in Enzvmology. 266:460-480(1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as 10 DNA59573. <br><br> Based on the DNA59573 consensus sequence and its relation to sequences identified in the assembly, one of the clones ( Incyte EST clone no. 2623992), including one of the sequences in the assembly was purchased and the . cDN A insert was obtained and sequenced. Incyte clone 2623992 came from a library constructed of RNA from epidermal breast keratinocytes. The sequence of this cDNA insert is shown in Figure 75 (SEQ ID NO: 196) and is 15 herein designated as DNA64889-1541. <br><br> The entire coding sequence of DNA64889-I541 is included in Figure 75 (SEQ ID NO: 196). Clone DNA64889-154I contains a single open reading frame with an apparent translational initiation site at nucleotide positions 24-26 and ending at the stop codon at nucleotide positions 354-356 (Figure 75). The predicted polypeptide precursor is 110 amino acids long (Figure 76; SEQ ID NO: 197). The full-length PROl274 protein 20 shown in Figure 76 has an estimated molecular weight of about 12,363 daltons and a pi of about 8.31. Analysis ofthe full-length PR01274 sequence shown in Figure 76 (SEQ ID NO: 197) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis ofthe full-length PRO 1274 sequence shown in Figure 76 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 24; an N-glycosylation site 25 from about amino acid 71 to about amino acid 75; and insulin family protein homology blocks from about amino acid 76 to about amino acid 96, and from about amino acid 42 to about amino acid 61. Clone DNA64889-1541 has been deposited with ATCC on September 9, 1998 and is assigned ATCC deposit no. 203250. <br><br> An analysis ofthe Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 76 (SEQ ID NO: 197), revealed sequence identity between 30 the PROl 274 amino acid sequence and the following Dayhoff sequences: CEW05B29, AF0169221 (insulin-like growth factor 1), B48151, A53640, BTIGF2REC1 (insulin-like growth factor 2), HSNF1GEN121, TXA3_RADMA (neurotoxin 3), CXMl_CONGE, P_P6130I, TXA4 RADMA (neurotoxin 4). <br><br> EXAMPLE 42 Isolation of cDNA Clones Encoding Human PRQ1286 35 DNA64903-1553 was identified by applying the proprietary signal sequence finding algorithm described in <br><br> Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster <br><br> 158 <br><br> sequence from the LIFESEQ® database, designated Incyte EST cluster no. 86809. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 5 (Altshul et al., Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). ESTs in the assembly included those identified from tumors, cell lines, or diseased tissue. One or more ofthe ESTs was obtained from a cDNA library constructed from RNA isolated from diseased colon tissue. The consensus 10 sequence obtained therefrom is herein designated as DNA58822. <br><br> In light of the sequence homology between the DNA58822 sequence and Incyte EST clone no. 1695434, Incyte EST no. 1695434 was purchased and the cDNA insert was obtained and sequenced. The sequence of this ^ cDNA insert is shown in Figure 77 (SEQ ID NO: 198) and is herein designated as DNA64903-1553. <br><br> The entire coding sequence of DNA64903-1553 is included in Figure 77 (SEQ ID NO: 198). Clone 15 DNA64903-1553 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 93-95 and ending at the stop codon at nucleotide positions 372-374 (Figure 77). The predicted polypeptide precursor is 93 amino acids long (Figure 78; SEQ ID NO: 199). The full-length PR01286 protein shown in Figure 78 has an estimated molecular weight of about 10,111 daltons and a pi of about 9.70. Analysis of the full-length PR01286 sequence shown in Figure 78 (SEQ ID NO: 199) evidences the presence of a variety 20 of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 1286 sequence shown in Figure 78 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 18; and N-myristoylation sites from about amino acid 15 to about amino acid 21, from about amino acid 17 to about amino acid 23, from about amino acid 19 to about amino acid 25, from about amino acid 83 to about amino acid 89, and from about 25 amino acid 86 to about amino acid 92. Clone DNA64903-1553 has been deposited with ATCC on September 15, | 1998 and is assigned ATCC deposit no. 203223. <br><br> An analysis ofthe Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 78 (SEQ ID NO: 199), revealed some homology between the PR01286 amino acid sequence and the following Dayhoff sequences: SR5C_ARATH, CELC17H1211, 30 MCPD ENTAE, JQ2283, INVO_LEMCA, P_R07309, ADEVBCAGN 4, AF020947J, CELT23H2J, and MDHSTRAR. <br><br> EXAMPLE 43 Isolation of cDNA Clones Encoding Human PRO 1294 DNA64905-1558 was identified by applying the proprietary signal sequence finding algorithm described in 35 Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster sequence from the LIFESEQ® database, designated Incyte EST cluster no. 10559. This EST cluster sequence was <br><br> 159 <br><br> then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ8, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 (Altshul et al., Methods in Enzvmology. 266:460-480 (1996)). Those comparisons resulting in a BLAST score 5 of 70 (or in some cases 90) or greater that did not encode known proteins w ere clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). The consensus sequence obtained therefrom is herein designated as DNA57203. <br><br> In light of the sequence homology between the DNA57203 sequence and Incyte EST clone no. 3037763, Incyte EST no. 3037763 was purchased and the cDNA insert was obtained and sequenced. The sequence of this 10 cDNA insert is shown in Figure 79 (SEQ ID N0:200) and is herein designated as DNA64905-1558. <br><br> The entire coding sequence of DNA64905-1558 is included in Figure 79 (SEQ ID N0:200). Clone DNA64905-1558 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 110-112 and ending at the stop codon at nucleotide positions 1328-1330 (Figure 79). The predicted polypeptide precursor is 406 amino acids long (Figure 80; SEQ ID N0:201). The full-length PRO 1294 protein 15 shown in Figure 80 has an estimated molecular weight of about 46,038 daltons and a pi of about 6.50. Analysis of the full-length PR01294 sequence shown in Figure 80 (SEQ ID N0:201) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 1294 sequence shown in Figure 80 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 21; N-glycosylation sites 20 from about amino acid 177 to about amino acid 181, and from about amino acid 248 to about amino acid 252; a cAMP- and cGMP-dependent protein kinase phosphorylation site from about amino acid 196 to about amino acid 200; a tyrosine kinase phosphorylation site from about amino acid 89 to about amino acid 97; N-myristoylation sites from about amino acid 115 to about amino acid 121, from about amino acid 152 to about amino acid 158, and from about amino acid 370 to about amino acid 376: and an amidation site from about amino acid 122 to about 25 aminoacid 126. Clone DNA64905-1558 has been deposited with ATCC on September 15,1998 and is assigned ATCC deposit no. 203233. <br><br> An analysis of the Dayhoff database (version 3 5.45 SwissProt 3 5), using a WU-BL AST2 sequence alignment analysis of the full-length sequence shown in Figure 80 (SEQ ID N0:201). evidenced significant homology between the PRO 1294 amino acid sequence and the following Dayhoff sequences: 173636, AF028740_I, 30 AB006686S31, P_R98225, RNU78105J, CELC48E7_4,CEF11C3_3, SCP1_MESAU, TPM3 HUMAN, and CELK05B23. <br><br> EXAMPLE 44 Isolation of cDNA Clones Encoding Human PROl 303 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 35 Example 1 above. This consensus sequence is designated herein as DNA47347. Based on the DNA47347 consensus sequence, and its homology to an Incyte EST within the assembly from which DNA47347 was derived, <br><br> 160 <br><br> # <br><br> Incyte clone 1430305 was purchased and sequenced in full. The sequence encoding PRO!303 was thereby identified. <br><br> The entire coding sequence of DNA65409-1566 is included in Figure 81 (SEQ ID N0:202). Clone DNA65409-1566 contains a single open reading frame with an apparent translational initiation site at nucleotide 5 positions 121-123, and an apparent stop codon at nucleotide positions 865-867. The predicted polypeptide precursor is 248 amino acids long, and has an estimated molecular weight of about 26,734 daltons and a pi of about 7.90. Analysis of the full-length PROl 303 sequence shown in Figure 82 (SEQ ID N0.203) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 1303 polypeptide shown in Figure 82 10 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 17; N-glycosylation sites from about amino acid 24 to about amino acid 28, and from about amino acid 163 to about amino acid 167; a serine proteases, trypsin family, histidine active site from about amino acid 58 to about amino acid 64; serine proteases, trypsin family, histidine protein domains from about amino acid 47 to about amino acid 64, from about amino acid 196 to about amino acid 207, and from about amino acid 218 to about amino acid 242; 15 kringle domain proteins homology blocks from about amino acid 194 to about amino acid 207, and from about amino acid 47 to about amino acid 65; and an apple domain from about amino acid 220 to about amino acid 248. Clone DN A65409-1566 has been deposited with the ATCC on September 15,1998 and is assigned ATCC deposit no. 203232. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment 20 analysis of the full-length sequence shown in Figure 82 (SEQ ID N0:203), revealed sequence identity between the PROl 303 amino acid sequence and the following Dayhoff sequences: AB009849 I, P W08475, AF024605 1, A420481, TRY3RAT, MMAE00066414, TRY 1 RAT, MMAE000663_4, MMAE000665_2. and MM AE00066412. <br><br> EXAMPLE 45 <br><br> 25 Isolation of cDNA Clones Encoding Human PRO 1304 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA35745. Based on the DNA35745 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PRO 1304. 30 PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (35745.fl): <br><br> 5'-GTGTTCTGCTGGAGCCGATGCC-3' (SEQ ID N0:206) <br><br> forward PCR primer (33745.P): <br><br> 5'-G ACATGG ACA ATG AC AGG-3' (SEQ ID N0:207) <br><br> 35 forward PCR primer (35745.f3): <br><br> 5'-CCTTTCAGGATGTAGGAG-3' (SEQ ID N0.208) <br><br> 161 <br><br> forward PCR primer G5745.f4): <br><br> 5-GATGTCTGCCACCCCAAG-3' (SEQ ID N0:209) reverse PCR primer (35745.ri): <br><br> 5-GCATCCTGATATG ACTTGTCACGTGGC-3' (SEQ ID NO:210) 5 reverse PCR primer (35745.r2): <br><br> 5'-TACAAGAGGGAAGAGGAGTTGCAC-3' (SEQ ID NQ:211) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA35745 sequence which had the following nucleotide sequence: <br><br> hybridization probe (35745.01): 10 S'-GCCCATTATGACGGCTACCTGGCTAAAGACGGCTCGAAATTCTACTGCAGCC-O'fSEQIDNO^) <br><br> In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened ^ by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PR01304 gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human ovary tissue. <br><br> 15 DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for <br><br> DNA65406-I567 [Figure 83, SEQ ID N0:204]; and the derived protein sequence for PR01304. <br><br> The entire coding sequence of DNA65406-1567 is included in Figure 83 (SEQ ID N0:204). Clone DNA65406-1567 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 23-25, and an apparent stop codon at nucleotide positions 689-691. The predicted polypeptide precursor 20 is 222 amino acids long, and has an estimated molecular weight of about 25,794 daltons and a pi of about 6.24. Analysis of the full-length PROl 304 sequence shown in Figure 84 (SEQ ID N0:205) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PRO 1304 polypeptide shown in Figure 84 evidences the presence of the following: an endoplasmic reticulum targeting sequence from about amino acid 219 25 to about amino acid 224; an N-glycosylation site from about amino acid 45 to about amino acid 49; FKBP-type peptidyl-prolyl cis-trans isomerase homology blocks from about amino acid 87 to about amino acid 124, and from about amino acid 129 to about amino acid 143; and an EF-hand calcium-binding domain protein homology block from about amino acid 202 to about amino acid 215. Clone DNA65406-1567 has been deposited with the ATCC on September 15, 1998 and is assigned ATCC deposit no. 203219. <br><br> 30 An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 84 (SEQ ID N0:205), evidenced significant homology between the PROl304 amino acid sequence and the following Dayhoff sequences: AF040252 1, P R28980, S71238, CELC05C8J, VFU52045J. S75144, FKB3_BOVIN, CELC50F2_6, CELB0511_12, and P_R4I781. <br><br> The DNA65406-1567 sequence was also obtained by isolating and sequencing the insert of Incyte EST clone 35 no. 2813577. <br><br> 162 <br><br> EXAMPLE 46 Isolation of cDNA Clones Encoding Human PRQ1312 DNA55773 was identified in a human fetal kidney cDNA library using a yeast screen, that preferentially represents the 5' ends of the primary cDNA clones. Based on the DNA55773 sequence, 5 oligonucleotides were synthesized for use as probes to isolate a clone of the full-length coding sequence for PRO 1312. <br><br> A full length clone [DNA61873-1574] was identified that contained a single open reading frame with an apparent translational initiation site at nucleotide positions 7-9, and a stop signal at nucleotide positions 643-645 (Figure 85; SEQ ID NO:213). The predicted polypeptide precursor is 212 amino acids long, and has a calculated 10 molecular weight of approximately 24,024 daltons and an estimated pi of approximately 6.26. Analysis of the full-length PR01312 sequence shown in Figure 86 (SEQ ID NO:214) evidences the presence of a variety of important polypeptide domains as shown in Figure 86, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR01312 polypeptide shown in Figure ' 86 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 14; a 15 transmembrane domain from about amino acid 141 to about amino acid 160; and N-glycosylation sites from about amino acid 76 to about amino acid 80. and from about amino acid 93 to about amino acid 97. Clone DNA6I873-1574 was deposited with the ATCC on August 18, 1998 , and is assigned ATCC deposit no. 203132. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 86 (SEQ ID NO:214) revealed some homology between the 20 PR01312 amino acid sequence and the following Dayhoff sequences: GCINTALPH_1, GIBMUC1A I, P_R96298, AF001406J, PVU88874J, P_R85151, AF041409J, CELC50F2 7, C45875, and AB009510_21. <br><br> EXAMPLE 47 Isolation of cDNA Clones Encoding Human PRQ1313 <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in 25 Example I above. This consensus sequence is designated herein as DNA64876. Based on the DNA64876 consensus sequence and upon a search for sequence homology with a proprietary Genentech EST sequence designated as DN A57711, a Merck'Washington University EST sequence (designated R80613) was found to have significant homology with DNA64876 and DNA57711. Therefore, the Merck/Washington University EST clone no. R80613 was purchased and the insert thereof obtained and sequenced, thereby giving rise to the DNA64966-30 1575 sequence shown in Figure 87 (SEQ ID NO:215), and the derived protein sequence for PR01313. <br><br> The entire coding sequence of DNA64966-1575 is included in Figure 87 (SEQ ID NO:215). Clone DNA64966-1575 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 115-117, and an apparent stop codon at nucleotide positions 1036-1038. The predicted polypeptide precursor is 307 amino acids long, and has an estimated molecular weight of about 35,098 daltons and a pi of about 35 8.11. Analysis of the full-length PRO 1313 sequence shown in Figure 88 (SEQ ID NO:216) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains <br><br> 163 <br><br> are approximate as described above. Analysts of the full-length PR01313 polypeptide shown in Figure 88 evidences the presence ofthe following: transmembrane domains from about amino acid 106 to about amino acid 121, from about amino acid 136 to about amino acid 152, from about amino acid 172 to about amino acid 188, from about amino acid 230 to about amino acid 245, and from about amino acid 272 to about amino acid 285; N-5 glycosylation sites from about amino acid 34 to about amino acid 38, from about amino acid 135 to about amino acid 139, and from about amino acid 203 to about amino acid 207; a tyrosine kinase phosphorylation site from about amino acid 59 to about amino acid 67; N-myristoylation sites from about amino acid 165 to about amino acid 171, from about amino acid 196 to about amino acid 202, from about amino acid 240 to about amino acid 246, and from about amino acid 247 to about amino acid 253; and an ATP/GTP-binding site motif A (P-loop) from about 10 amino acid 53 to about amino acid 61. Clone DNA64966-1575 has been deposited with the ATCC on January 12, 1999 and is assigned ATCC deposit no. 203575. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 88 (SEQ ID NO:2l6), evidenced significant homology ^ between ihe PR01313 amino acid sequence and the following Dayhoff sequences: CELT27A13, CEF09C67, 15 U93688 9, H64896, YDCX_ECOLl. and RNU06101J. <br><br> EXAMPLE 48 Isolation of cDNA Clones Encoding Human PRO 1376 <br><br> A Merck/Washington University database was searched and a sequence was identified as Accession No. W39620 (identified as a Soares parathyroid tumor protein). This sequence was put into a database wherein it could 20 be compared to other sequences and aligned with other sequences. <br><br> A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in Example 1 above. This consensus sequence is designated herein as DNA6722I. <br><br> The clone encoding Merck W39620 was purchased and fully sequenced. DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for DNA67300-1605 [Figure 89, SEQ ID 25 NO:217];and the derived protein sequence for PR01376. <br><br> The entire coding sequence of DNA67300-1605 is included in Figure 89 (SEQ ID NO:217). Clone DNA67300-1605 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 107-109, and an apparent stop codon at nucleotide positions 623-625. The predicted polypeptide precursor is ! 72 amino acids long, and has an estimated molecular weight of about 19,206 daltons and a pi of about 30 5.36. Analysis of the full-length PRO 1376 sequence shown in Figure 90 (SEQ ID NO:218) evidences the presence of a variety of important polypeptide domaiiis, wherein the locations given forthose important polypeptide domains are approximate as described above. Analysis of the full-length PR01376 polypeptide shown in Figure 90 evidences the presence of the following: a signal peptide from about amino acid I to about amino acid 23; and a thioredoxin family proteins homology block from about amino acid 58 to about amino acid 75. Clone DNA67300-35 1605 has been deposited with the ATCC on August 25, 1998 and is assigned ATCC deposit no. 203163. <br><br> An analysis ofthe Dayhoff database (version 35.45 SwissProt 35). using a WU-BLAST2 sequence alignment <br><br> 164 <br><br> analysis of the full-length sequence shown in Figure 90 (SEQ ID NO:218), revealed sequence identity between the PRO 1376 amino acid sequence and the following Dayhoff sequences: XAG_XENLA, AF0254741, NP77 XENLA, D69100, S75124, ER60_SCHMA, H69466, A57254, AB002234J, and TATHIORDH I. <br><br> EXAMPLE 49 <br><br> 5 Isolation of cDNA Clones Encoding Human PROl 387 <br><br> DNA68872-1620 was identified by applying the proprietary signal sequence finding algorithm described in Example 3 above. Use of the above described signal sequence algorithm allowed identification of an EST cluster sequence from the LIFESEQ® database, designated Incyte EST cluster no. 10298. This EST cluster sequence was then compared to a variety of expressed sequence tag (EST) databases which included public EST databases (e.g., 10 GenBank) and a proprietary EST DNA database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA) to identify existing homologies. The homology search was performed using the computer program BLAST or BLAST2 . (Altshul et al.. Methods in Enzvmology. 266:460-480 (1996)"). Those comparisons resulting' in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into a consensus DNA sequence with the program "phrap" (Phil Green, University of Washington, Seattle. Washington). 15 The consensus sequence obtained therefrom is herein designated as DNA56259. A Genentech proprietary EST sequence was used in the assembly and is herein designated DNA39516. <br><br> In light of the sequence homology between the DNA56259 sequence and Incyte EST clone no. 3507924, Incyte EST no. 3507924 was purchased and the cDNA insert was obtained and sequenced. The sequence of this cDNA insert is shown in Figure 91 (SEQ ID NO:219) and is herein designated as DNA68872-1620. 20 The entire coding sequence of DNA68872-1620 is included in Figure 91 (SEQ ID NO:2l9). Clone <br><br> DNA68872-1620 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 85-87 and ending at the stop codon at nucleotide positions 1267-1269 (Figure 91). The predicted polypeptide precursor is 394 amino acids long (Figure 92; SEQ ID N0:220). The full-length PRO 1387 protein shown in Figure 92 has an estimated molecular weight of about 44.339 daltons and a pi of about 7.10. Analysis 25 of the full-length PR01387 sequence shown in Figure 92 (SEQ ID N0:220) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PROl 387 sequence shown in Figure 92 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 19; a transmembrane domain from about amino acid 275 to about amino acid 296; N-glycosylation sites from about amino acid 76 to 30 about amino acid 80, from about amino acid 231 to about amino acid 235, from about amino acid 302 to about amino acid 306. from about amino acid 307 to about amino acid 311, and from about amino acid 376 to about amino acid 380: and myelin P0 protein homology blocks from about amino acid 210 to about amino acid 240. and from about amino acid 92 to about amino acid 122. Clone DNA68872-1620 has been deposited with ATCC on August 25,1998 and is assigned ATCC deposit no. 203160. <br><br> 35 An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment analysis of the full-length sequence shown in Figure 92 (SEQ ID N0.220), evidenced significant homology <br><br> 165 <br><br> between the PROl 387 amino acid sequence and the following Dayhoff sequences: P_W36955, MYPO_HETFR, HS46KDA J ,AF049498J ,MYOO_HUM AN. AF030454J .A53268,SHPTCRA_1 ,P_W 14146,and GEN 12838. <br><br> EXAMPLE 50 Isolation of cDNA Clones Encoding Human PRQ1561 5 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described in <br><br> Example I above. This consensus sequence is designated herein as DNA40630. A proprietary Genentech EST sequence was employed in the consensus assembly and is herein designated as DNA40753. Based on the DNA40630 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for 10 PROl 561. <br><br> PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer (4063Q.flV. <br><br> 5-CTGCCTCCACTGCTCTGTGCTGGG-3' (SEQ IDNO:223) <br><br> reverse PCR primer (4Q630.rn: <br><br> 15 5-CAGAGCAGTGGATGTTCCCCTGGG-3' (SEQ ID NO:224) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA40630 sequence which had the following nucleotide sequence: <br><br> hybridization probe (40630.pl): <br><br> 5'-CTGAACAAGATGGTCAAGCAAGTGACTGGGAAAATGCCCATCCTC-3' (SEQ ID NO:225) <br><br> 20 In order to screen several libraries for a source of a full-length clone. DNA from the libraries was screened by PCR amplification with the PCR primer pairs identified above. A positive library was then used to isolate clones encoding the PR0156j gene using the probe oligonucleotide and one of the PCR primers. RNA for construction of the cDNA libraries was isolated from human breast tumor tissue. <br><br> DNA sequencing of the isolated clones isolated as described above gave the full-length DNA sequence for 25 DNA76538-1670 [Figure 93, SEQ ID NO:221]: and the derived protein sequence for PROl 561. <br><br> The entire coding sequence of DNA76538-1670 is included in Figure 93 (SEQ ID NO:22l). Clone DNA76538-1670 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 29-31, and an apparent stop codon at nucleotide positions 377-379. The predicted polypeptide precursor is 116 amino acids long, and has an estimated molecular weight of about 12,910 daltons and a pi of about 6.41. 30 Analysis of the full-length PROl 561 sequence shown in Figure 94 (SEQ ID NO:222) evidences the presence of a variety of important polypeptide domains, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PROl561 polypeptide shown in Figure 94 evidences the presence of the following: a signal peptide from about amino acid 1 to about amino acid 17; an N-glycosylation site from about amino acid 86 to about amino acid 90; N-myristoylation sites from about amino acid <br><br> 166 <br><br> 20 to about amino acid 26, and from about amino acid 45 to about amino acid 51; and a phospholipase A2 histidine active site from about amino acid 63 to about amino acid 71. Clone DNA76538-1670 has been deposited with the ATCC on October 6, 1998 and is assigned ATCC deposit no. 203313. <br><br> An analysis of the Dayhoff database (version 35.45 SwissProt 35), using a WU-BLAST2 sequence alignment 5 analysis of the full-length sequence shown in Figure 94 (SEQ ID NO:222), evidenced significant homology between the PR01561 amino acid sequence and the following Dayhoff sequences: P R63053, P R25416, P_R63055,P_P93363,P_R63046.PA2A_VIPAA,P_W58476,GEN 13747,PA2X_HUMAN,and PA2A_CRODU. <br><br> In addition to the above, a sequence homology search evidenced significant homology between the DNA40630 consensus sequence and Incyte EST clone no. 1921092. As such, Incyte EST clone no. 1921092 was 10 purchased and the insert obtained and sequenced, thereby giving rise to the DN A76538-1670 sequence shown in Figure 93 (SEQ lDNO:22I). <br><br> EXAMPLE 51 <br><br> " Isolation of cDNA Clones Encoding Human PRQ2I6 <br><br> The extracellular domain (ECD) sequences (including the secretion signal sequence, if any) from about 15 950 known secreted proteins from the Swiss-Prot public database were used to search EST databases. The EST databases included public EST databases (e.g., GenBank) and a proprietary EST database (LIFESEQ®, Incyte Pharmaceuticals, Palo Alto, CA). The search was performed using the computer program BLAST or BLAST2 [Altschul et a!., Methods in Enzvmologv. 266:460-480 (1996)] as a comparison of the ECD protein sequences to a 6 frame translation of the EST sequences. Those comparisons resulting in a BLAST score of 70 (or in some 20 cases, 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green, University of Washington, Seattle, Washington). <br><br> The complete cDN A sequence of DNA33087 isdisclosedinGenBank under accession numbers AB000114 1 and AB009589_1 (human osteomodulin). A related, but probably different protein, corneal keratin sulfate, is disclosed in Funderburgh et a!.. J. Biol. Chem.. 271:31431-31436 (1996). <br><br> I 25 A consensus DNA sequence was assembled relative to other EST sequences using phrap as described above. <br><br> This consensus sequence is herein designated DNA28754. In some cases, the consensus sequence derives from an intermediate consensus DNA sequence which was extended using repeated cycles of BLAST and phrap to extend that intermediate consensus sequence as far as possible using the sources of EST sequences discussed above. <br><br> Based on the DNA28754 consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a 30 cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0216. Forward and reverse PCR primers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In some cases, additional oligonucleotides are synthesized when the consensus sequence is greater than about 1-1.5 kbp. In order to screen several libraries for a full-length clone. DNA from the libraries was 35 screened by PCR amplification, as per Ausubel etal., Current Protocols in Molecular Biology, supra, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest using the probe <br><br> 167 <br><br> oligonucleotide and one of the primer pairs. <br><br> PCR primers (forward and reverse) were synthesized: <br><br> forward PCR primer: <br><br> 5-TCACGATGATCCTGACAATGC-3' (SEQ ID NO:228) <br><br> 5 reverse PCR primer: <br><br> 5'-AATAATGAAGGTCAAAGTGCCCTT-3' (SEQ IDNO:229) <br><br> Additionally, a synthetic oligonucleotide hybridization probe was constructed from the consensus DNA28754 sequence which had the following nucleotide sequence: <br><br> hybridization probe: <br><br> 10 S'-TGCTCCTTCTTGTTCTGGGCTCTCATG-S' (SEQ ID N0:230) <br><br> RNA for construction of the cDN A libraries was isolated from human fetal kidney tissue. The cDNA libraries used to isolate the cDNA clones were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA. The cDNA was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a 15 defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et at., Science. 253:1278-1280 (1991)) in the unique Xhol and Notl sites. <br><br> DNA sequencing of the clones isolated as described above gave the full-length DNA sequence for a full-length PR0216 polypeptide (designated herein as DNA33087 [Figure 95, SEQ ID NO:226]) and the derived protein sequence for that PR0216 polypeptide. <br><br> 20 The full length clone identified above contained a single open reading frame with an apparent translational initiation site at nucleotide positions 268-270 and a stop signal at nucleotide positions 1531-1533 (Figure 95, SEQ ID NO:226). The predicted polypeptide precursor is 421 amino acids long [Figure 96; (SEQ ID NO:227)] and has a calulated molecular weight of approximately 49,492 daltons and an estimated pi of about 5.51. Analysis of the full-length PR0216 sequence shown in Figure 96 (SEQ ID NO:227) evidences the presence of important 25 polypeptide domains as shown in Figure 96, wherein the locations given for those important polypeptide domains are approximate as described above. Analysis of the full-length PR02I6 sequence (Figure 96, SEQ ID NO:227) evidences the following: N-linked glycosylation sites from about amino acid 113 to about amino acid 117, from about amino acid 121 to about amino acid 125, from about amino acid 187 to about amino acid 191, from about amino acid 242 to about amino acid 246 and from about amino acid 316 to about amino acid 320; tyrosine kinase 30 phosphorylation sites from about amino acid 268 to about amino acid 275 and from about amino acid 300 to about amino acid 307; an N-myristoylation site from about amino acid 230 to about amino acid 236; and leucine zipper patterns from about amino acid 146 to about amino acid 168 and from about amino acid 217 to about amino acid 239. Clone DNA33087 has been deposited with ATCC on October 16, 1997 and is assigned ATCC deposit no. 209381. <br><br> 35 This clone is the same as human osteomodulin submitted by I. Ohno to GenBank on December 26, 1996 <br><br> (ABOOOI141) and submitted by I. Ohno et al., to GenBank on December 5, 1997 (AB009589-I). <br><br> 168 <br><br> EXAMPLE 52 Stimulation of Heart Neonatal Hypertrophy <br><br> This assay is designed to measure the ability of PRO polypeptides to stimulate hypertrophy of neonatal heart. Cardiac myocytes from 1-day old Harlan Sprague Dawley rats were obtained. Cells (180 /u\ at 7.5 x lOVml, serum 5 &lt;0.1%, freshly isolated) are added on day 1 to 96-well plates previously coated with DMEM/F12 + 4% FCS. Test samples containing the test PRO polypeptide are added directly to wells on day 2 in 20 fiL volumes. Cells are stained with crystal violet after an additional two days and scored visually by the next day. Incubator conditions are critical and require 5% C02. <br><br> Activity reference: phenylephrine at 1-100 /l/M, PGF-2 alpha at 0.1-1.0 //M, endothelin-1 at 1-10 nM, CT1 10 (LIF) at 1 -10 nM. No PBS is included, since Ca concentration is critical for assay response. Assay media included: DMEM/F12 (with 2.44 gm bicarbonate), 10 //g/ml transferrin, 1 /ug/ml insulin, 1 yug/ml aprotinin, 2 mmol/L glutamine, 100 U/ml penicillin G, 100 ^g/ml streptomycin. Protein buffer containing mannitol (4%) gave a positive signal (score 3.5) at 1/10 (0.4%) and 1/100 (0.04%), but not at 1/1000(0.004%). Therefore, the test sample buffer containing mannitol is not run. A secondary assay consists of measuring the ANP levels (ng/ml) by ELISA in 15 conditioned media from the cells. An increase in the ANP message can be measured by PCR from cells after a few hours. <br><br> Results are assessed by visually observing cell size: a score = 3.5 or greater is considered positive for conditioned media; a score of 3.0 or greater is considered positive for purified protein. <br><br> PR0231, PR0526, PR0713, PR0792, PRO 1246, and PRO 1312 purified polypeptides were observed to 20 stimulate neonatal heart hypertrophy and exhibited positive scores in this assay compared with positive controls as shown in TABLE 4 below: <br><br> TABLE 4 <br><br> Stimulation of Heart Neonatal Hypertrophy <br><br> PRO SAMPLE Concentration (or dilution) Score <br><br> 25 PR0231 12.0/uM 3.375 <br><br> PR0526 1% 3.50 <br><br> PR0713 10.0 nM 3.25 <br><br> PR0713 10.0 nM 3.25 <br><br> PR0792 1% 4.50 <br><br> 30 PR01246 1% 3.25 <br><br> PR01312 85.0 nM 3.375 <br><br> 169 <br><br> EXAMPLE 53 <br><br> Enhancement of Heart Neonatal Hypertrophy Induced by F2a This assay is designed to measure the ability of PRO polypeptides to stimulate hypertrophy of neonatal heart. Cardiac myocytes from l-day old Harlan Sprague Dawley rats were obtained. Cells (180 /ul at 7.5 x 104/ml, serum 5 &lt;0.1%, freshly isolated) are added on day 1 to 96-well plates previously coated with DMEM/F12 + 4% FCS. Test samples containing the test PRO polypeptide (20 //l/well) are added directly to the wells on day 1. PGF (20 /ul/well) is then added on day 2 at a final concentration of 10"6 M. The cells are then stained on day 4 and visually scored on day 5. Visual scores are based on cell size, wherein cells showing no increase in size as compared to negative controls are scored 0.0, cells showing a small to moderate increase in size as compared to negative controls 10 are scored 1.0 and cells showing a large increase in size as compared to negative controls are scored 2.0. A score of 1.0 or greater is considered positive. <br><br> No PBS is included, since Ca concentration is critical for assay response. Plates are coated with DMEM/F12 plus 4% FCS (200 /J/well). Assay media included: DMEM/F12 (with 2.44 gm bicarbonate), 10 ^g/ml transferrin, 1 ^g/ml insulin, 1 /^g/ml aprotinin. 2 mmol/L glutamine, 100 U/ml penicillin G, 100/^g/ml streptomycin. Protein 15 buffer containing mannitol (4%) gave a positive signal (score 3.5) at 1/10 (0.4%) and 1/100 (0.04%), but not at 1/1000 (0.004%). Therefore the test sample buffer containing mannitol is not run. <br><br> PRO 179, PRO 182, PRO 195, and PR0224 purified polypeptides showed positive results when assayed by the above method as shown in TABLE 5 below: <br><br> TABLE 5 <br><br> 20 Enhancement of Heart Neonatal Hypertrophy Induced bv F2a <br><br> PRO Concentration Score <br><br> PRO 179 0.01% 0.0 <br><br> PRO 179 0.10% 0.0 <br><br> PROl 79 1% 1.0 <br><br> 25 PRO 182 0.01% 0.0 <br><br> PRO 182 0.10% 0.0 <br><br> PROl 82 1% 1.0 <br><br> PR0195 0.01% 0.0 <br><br> PRO 195 0.1% 1.0 <br><br> 30 PRO 195 1% 1.0 <br><br> PR0224 0.01% 0.0 <br><br> PR0224 0.10% 0.0 <br><br> PR0224 1% 1.0 <br><br> EXAMPLE 54 <br><br> 35 Inhibition of Heart Adult Hypertrophy <br><br> This assay is designed to measure the inhibition of heart adult hypertrophy. Ventricular myocytes are freshly isolated from adult (250g) Harlan Sprague Dawley rats and the cells are plated at 2000/well in 180 //I volume. On <br><br> 170 <br><br> day two, test samples (20 /u 1) containing the test PRO polypeptide are added. On day Five, the cells are fixed and then stained. An increase in ANP message can also be measured by PCR from cells after a few hours. Results are based on a visual score of cell size: 0 = no inhibition, -1 =smal! inhibition, -2 = large inhibition. A score of less than 0 is considered positive. Activity reference corresponds to phenylephrin (PE) at 0.1 mM, as a positive control. A 5 score of 2 is considered very responsive. Assay media included: M199 (modified)-glutamine free, NaHCOj, phenol red, supplemented with 100 nM insulin, 0.2% BSA, 5 mM cretine, 2 mM L-carnitine, 5 mM taurine, 100 U/ml penicillin G, 100 ng/m\streptomycin (CCT medium). Only inner 60 wells are used in 96 well plates. Of these, 6 wells are reserved for negative and positive (PE) controls. Initially, quantitative PCR will be run in parallel to determine relative level of sensitivity to the visual scoring system. <br><br> 10 PR0269 and PROS 56 showed positive results in inhibition of heart adult hypertrophy in the above described assay. <br><br> EXAMPLE 55 Stimulation of Endothelial Cell Proliferation This assay is designed to determine whether PRO polypeptides show the ability to stimulate adrenal cortical capillary endothelial cell (ACE) growth. <br><br> Bovine adrenal cortical capillary'endothelial (ACE) cells (from primary culture, maximum of 12-14 passages) were plated in 96-well plates at 500 celisAvell per 100 microliter. Assay media included low glucose DMEM, 10% calf serum, 2 mM glutamine, and IX penicillin/streptomycin/fungizone. Control wells included the following: (1) no ACE cells added; (2) ACE cells alone; (3) ACE cells plus VEGF (5 ng/ml); and (4) ACE cells plus FGF (5ng/ml). The control or test sample, (in 100 microliter volumes), was then added to the wells (at dilutions of 1%, 0.1% and 0.01%, respectively). The cell cultures were incubated for 6-7 days at 37°C/5% C02. After the incubation, the media in the wells was aspirated, and the cells were washed IX with PBS. An acid phosphatase reaction mixture (100 microliter: 0.1M sodium acetate, pH 5.5, 0.1% Triton X-100, 10 mM p-nitrophenyl phosphate) was then added to each well. After a 2 hour incubation at 37°C, the reaction was stopped by addition 25 of 10 microliters IN NaOH. Optical density (OD) was measured on a microplate reader at 405 nm. <br><br> The activity of a PRO polypeptide was calculated as the fold increase in proliferation (as determined by the acid phosphatase activity, OD 405 nm) relative to (1) cell only background, and (2) relative to maximum stimulation by VEGF. VEGF (at 3-10 ng/ml) and FGF (at 1-5 ng/ml) were employed as an activity reference for maximum stimulation. Results of the assay were considered "positive" if the observed stimulation was &gt; 50% 30 increase over background. VEGF (5 ng/ml) control at 1% dilution gave 1.24 fold stimulation; FGB (5 ng/ml) control at 1% dilution gave 1.46 fold stimulation. <br><br> PRO 179, PR0212. PR01075. PROl 154, PR01244, PR01286, and PR01303 assayed "positive" as shown in TABLE 6 below: <br><br> 171 <br><br> 10 <br><br> 20 <br><br> TABLE 6 <br><br> Stimulation of Endothelial Cell Proliferation <br><br> PRO <br><br> Concentration <br><br> Fold Stimulation <br><br> PROl 79 <br><br> 0.01% <br><br> 1.16 <br><br> PRO 179 <br><br> 0.10% <br><br> 1.57 <br><br> PROl 79 <br><br> 1.0% <br><br> 1.38 <br><br> PR0212 <br><br> 0.01% <br><br> 4.08 <br><br> PR0212 <br><br> 0.10% <br><br> 4.73 <br><br> PR0212 <br><br> 1.0% <br><br> 4.75 <br><br> PRO 1075 <br><br> 0.01% <br><br> 4.21 <br><br> PRO 1075 <br><br> 0.10% <br><br> 4.52 <br><br> PRO 1075 <br><br> 1.0% <br><br> 3.39 <br><br> PROl 154 <br><br> 0.0017 <br><br> nM <br><br> 4.63 <br><br> PROl 154 <br><br> 0.017 <br><br> nM <br><br> 5.15 <br><br> PRO 1154 <br><br> 0.17 <br><br> nM <br><br> 5.53 <br><br> PRO 1244 <br><br> 0.67 <br><br> nM <br><br> 4.60 <br><br> PRO 1244 <br><br> 6.7 <br><br> nM <br><br> 4.78 <br><br> PRO 1244 <br><br> 67.0 <br><br> nM <br><br> 5.24 <br><br> PRO 1286 <br><br> 0.5 <br><br> nM <br><br> 4.61 <br><br> PRO 1286 <br><br> 5.0 <br><br> nM <br><br> 4.55 <br><br> PROl 303 <br><br> 0.38 <br><br> nM <br><br> 4.29 <br><br> PROl 303 <br><br> 3.80 <br><br> nM <br><br> 4.28 <br><br> PROl 303 <br><br> 38.0 <br><br> nM <br><br> 3.74 <br><br> EXAMPLE 56 <br><br> 25 Inhibition of Vascular Endothelial Growth Factor (VEGF") Stimulated Proliferation of Endothelial Cell Growth The ability of various PRO polypeptides to inhibit VEGF stimulated proliferation of endothelial cells was tested. Specifically, bovine adrenal cortical capillary endothelial (ACE) cells (from primary culture, maximum of 12-14 passages) were plated in 96-well plates at 500 cells/well per 100 microliter. Assay media included low glucose DMEM, 10% calf serum (not fetal calf), 2 mM glutamine, and IX penicillin/streptomycin/fungizone. <br><br> 30 Control wells included the following: (1) no ACE cells added; (2) ACE cells alone; (3) ACE cells plus 5 ng/ml FGF; (4) ACE cells plus 3 ng/ml VEGF; (5) ACE cells plus 3 ng/ml VEGF plus 1 ng/ml TGF-beta; and (6) ACE cells plus 3 ng/ml VEGF plus 5 ng/ml LIF. The test samples, poly-His tagged PRO polypeptides (in 100 microliter volumes), were then added to the wells (at dilutions of 1%, 0.1% and 0.01%, respectively). The cell cultures were incubated for 6-7 days at 37°C/5% CO,. After the incubation, the media in the wells was aspirated, and the cells <br><br> 35 were washed IX with PBS. An acid phosphatase reaction mixture (100 microliter; 0.1 M sodium acetate, pH 5.5, 0.1% Triton X-100, 10 mM p-nitrophenyl phosphate) was then added to each well. After a 2 hour incubation at 37°C, the reaction was stopped by addition of 10 microliters IN NaOH. Optical density (OD) was measured on a microplate reader at 405 nm. <br><br> 172 <br><br> The activity of the test PRO polypeptide was calculated as the percent inhibition of VEGF (3 ng/ml) stimulated proliferation (as determined by measuring acid phosphatase activity at OD 405 nm) relative to the cells without stimulation. TGF-beta was employed as an activity reference at 1 ng/ml, since TGF-beta blocks 70-90% of VEGF-stimulated ACE cell proliferation. The results, as shown in TABLE 7 below, are indicative of the utility 5 of the PRO polypeptides in cancer therapy and specifically in inhibiting tumor angiogenesis. The numerical values (relative inhibition) shown in TABLE 7 are determined by calculating the percent inhibition of VEGF stimulated proliferation by the PRO polypeptides relative to cells without stimulation and then dividing that percentage into the percent inhibition obtained by TGF-P at 1 ng/ml which is known to block 70-90% of VEGF stimulated cell proliferation. The results are considered positive if the PRO ploypeptide exhibits 30% or greater inhibition of 10 VEGF stimulation of endothelial cell growth (relative inhibition 30% or greater). <br><br> TABLE 7 <br><br> Inhibition of VEGF Stimilated Endothelial Cell Growth <br><br> PRO Name PRO Concentration Relative % Fnhibition <br><br> PRO 187 0.01% 91 <br><br> 15 PROl 87 0.10% 82 <br><br> PRO 187 1.0% 44 <br><br> PR0214 0.01% 68 <br><br> PR0214 0.10% 91 <br><br> PR0214 1.0% 94 <br><br> 20 PR0216 0.01% 96 <br><br> PR0216 0.10% 81 <br><br> PR0216 1.0% 31 <br><br> PR0216 0.01% 104 <br><br> PR0216 0.10% 93 <br><br> 25 PR0216 1.0% 50 <br><br> PR0323 0.01% 59 <br><br> PR0323 0.10% 93 <br><br> PR0323 1.0% 91 <br><br> PR0812 0.025 nM 101 <br><br> 30 PR0812 0.25 nM 101 <br><br> PR0812 2.5 nM 96 <br><br> PRO 1246 0.007 nM 90 <br><br> PRO 1246 0.07 nM 87 <br><br> PRO 1246 0.70 nM 60 <br><br> 35 PRO1246 0.007 nM 99 <br><br> PRO 1246 0.07 nM 94 <br><br> PRO 1246 0.70 nM 73 <br><br> 173 <br><br> EXAMPLE 57 Induction of c-fos in Endothelial Cells This assay is designed to determine whether PRO polypeptides show the ability to induce c-fos in endothelial cells. <br><br> 5 Human venous umbilical vein endothelial cells (HUVEC, Cell Systems) in growth media (50% Ham's FI2 <br><br> w/o GHT: low glucose, and 50% DMEM without glycine: with NaHC03, l%glutamine, 10 mM HEPES, 10% FBS, 10 ng/ml bFGF) were plated on 96-well microtiter plates at a cell density of IxlO4 cells/well. The day after plating, the cells were starved by removing the growth media and treating the cells with 100 /^I/well test samples and controls (positive control = growth media; negative control = Protein 32 buffer = 10 mM HEPES, 140 mM 10 NaCl, 4% (w/v) mannitol, pH 6.8). The cells were incubated for 30 minutes at 37°C, in 5% C02. The samples were removed, and the first part of the bDNA kit protocol (Chiron Diagnostics, cat. #6005-037) was followed, where each capitalized reagent/buffer listed below was available from the kit. <br><br> ^ Briefly, the amounts of the TM Lysis Buffer and Probes needed for the tests were calculated based on information provided by the manufacturer. The appropriate amounts of thawed Probes were added to the TM Lysis 15 Buffer. The Capture Hybridization Buffer was warmed to room temperature. The bDNA strips were set up in the metal strip holders, and 100 //I of Capture Hybridization Buffer was added to each b-DNA well needed, followed by incubation for at least 30 minutes. The test plates with the cells were removed from the incubator, and the media was gently removed using the vacuum manifold. 100 tA of Lysis Hybridization Buffer with Probes were quickly pipetted into each well of the microtiter plates. The plates were then incubated at 55 °C for 15 minutes. Upon 20 removal from the incubator, the plates were placed on the vortex mixer with the microtiter adapter head and vortexed on the #2 setting for one minute. 80 fA of the lysate was removed and added to the bDNA wells containing the Capture Hybridization Buffer, and pipetted up and down to mix. The plates were incubated at 53 °C for at least 16 hours. <br><br> On the next day, the second part of the bDNA kit protocol was followed. Specifically, the plates were 25 removed from the incubator and placed on the bench to cool for 10 minutes. The volumes of additions needed were calculated based upon information provided by the manufacturer. An Amplifier Working Solution was prepared by making a 1:100 dilution of the Amplifier Concentrate (20 fm///l) in AL Hybridization Buffer. The hybridization mixture was removed from the plates and washed twice with Wash A. 50 jA of Amplifier Working Solution was added to each well and the wells were incubated at 53 °C for 30 minutes. The plates were then removed from the 30 incubator and allowed to cool for 10 minutes. The Label Probe Working Solution was prepared by making a 1:100 dilution of Label Concentrate (40 pmoles/^1) in AL Hybridization Buffer. After the 10-minute cool-down period, the amplifier hybridization mixture w as removed and the plates were washed twice with Wash A. 50 )A of Label Probe Working Solution was added to each well and the wells were incubated at 53 °C for 15 minutes. After cooling for 10 minutes, the Substrate was wanned to room temperature. Upon addition of 3 ix\ of Substrate 35 Enhancer to each ml of Substrate needed for the assay, the plates were allowed to cool for 10 minutes, the label hybridization mixture was removed, and the plates were washed twice with Wash A and three times with Wash D. 50 tA of the Substrate Solution with Enhancer was added to each well. The plates were incubated for 30 minutes <br><br> 174 <br><br> at 37°C and RLU was read in an appropriate luminometer. <br><br> The replicates were averaged and the coefficient of variation was determined. The measure of activity of the fold increase over the negative control (Protein 32/HEPES buffer described above) value was indicated by chemiluminescence units (RLU). The results are shown in TABLE 8 below, and are considered positive if the PRO polypeptide exhibits at least a two-fold value over the negative buffer control. Negative control = 1.00 RLU at 1.00% dilution. Positive control = 8.39 RLU at 1.00% dilution. <br><br> 175 <br><br> PRO Name <br><br> PR0287 PR0287 PR0287 <br><br> TABLE 8 <br><br> Induction of c-fos in Endothelial Cells <br><br> PRO Concentration <br><br> 0.018 nM 0.18 nM 1.8 nM <br><br> RLU values <br><br> 2.06 1.89 1.73 <br><br> PR0287 PR0287 <br><br> 0.18 1.8 <br><br> nM nM <br><br> 1.64 2.14 <br><br> PR0538 10 PR0538 PR0538 <br><br> 0.2149 nM 2.149 nM 21.49 nM <br><br> 1.95 2,15 1.46 <br><br> PR0538 PR0538 PR0538 <br><br> 0.2149 nM 2.149 nM 21.49 nM <br><br> 2.38 2.42 2.84 <br><br> PR0713 PR0713 PR0713 <br><br> 0.022 0.22 o "&gt; <br><br> nM nM nM <br><br> 1.95 2.17 2.13 <br><br> PR0713 PR0713 20 PR0713 <br><br> 0.022 <br><br> 0.22 <br><br> 2.2 <br><br> nM nM nM <br><br> 2.66 2.52 2.69 <br><br> PR0788 PR0788 PR0788 <br><br> 0.29 nM 2.9 nM 29.0 nM <br><br> 1.23 2.65 0.93 <br><br> 25 <br><br> PR0865 PR0865 PR0865 <br><br> 0.027 <br><br> 0.27 <br><br> 2.7 <br><br> nM nM nM <br><br> 1.98 3.09 2.90 <br><br> PR0865 PR0865 PR0865 <br><br> 0.027 nM 0.27 nM 2.7 nM <br><br> 2.88 2.19 2.64 <br><br> 30 <br><br> PROl 126 PROl 126 PROl 126 <br><br> 0.029 nM 0.29 nM 2.9 nM <br><br> 1.94 2.33 1.81 <br><br> 35 <br><br> PROl 130 PROl 130 PROl 130 <br><br> 0.6 nM 6.0 nM 60.0 nM <br><br> 1.89 2.04 2.06 <br><br> PRO 1274 PRO 1274 PRO 1274 <br><br> 0.365 nM 3.65 nM 36.5 nM <br><br> 0.86 1.14 2.11 <br><br> 176 <br><br> 10 <br><br> ,15 <br><br> 20 <br><br> PRO 1274 <br><br> 0.365 <br><br> nM <br><br> 2.04 <br><br> PRO 1274 <br><br> 3.65 <br><br> nM <br><br> 1.86 <br><br> PROl 274 <br><br> 36.5 <br><br> nM <br><br> 2.41 <br><br> PRO 1294 <br><br> 0.13 <br><br> nM <br><br> 2.38 <br><br> PRO 1294 <br><br> 1.3 <br><br> nM <br><br> 2.23 <br><br> PRO 1294 <br><br> 13.0 <br><br> nM <br><br> 1.39 <br><br> PROl 294 <br><br> 0.13 <br><br> nM <br><br> 2.78 <br><br> PRO 1294 <br><br> 1.3 <br><br> nM <br><br> 2.53 <br><br> PRO 1294 <br><br> 13.0 <br><br> nM <br><br> 1.39 <br><br> PRO 1304 <br><br> 0.074 <br><br> nM <br><br> 1.72 <br><br> PROl 304 <br><br> 0.74 <br><br> nM <br><br> 2.08 <br><br> PRO 1304 <br><br> 7.4 <br><br> nM <br><br> 1.41 <br><br> PRO 1304 <br><br> 0.074 <br><br> nM <br><br> 2.66 <br><br> PRO 1304 <br><br> 0.74 <br><br> nM <br><br> 1.75 <br><br> PRO 1304 <br><br> 7.4 <br><br> nM <br><br> 1.45 <br><br> PRO 1376 <br><br> 0.86 <br><br> nM <br><br> 1.41 <br><br> PR01376 <br><br> 8.6 <br><br> nM <br><br> 2.20 <br><br> PR01376 <br><br> 86.0 <br><br> nM <br><br> 2.59 <br><br> PR01376 <br><br> 0.86 <br><br> nM <br><br> 1.43 <br><br> PRO 1376 <br><br> 8.6 <br><br> nM <br><br> 2.28 <br><br> PR01376 <br><br> 86.0 <br><br> nM <br><br> 1.49 <br><br> PROl 387 <br><br> 0.1 <br><br> nM <br><br> 2.19 <br><br> PR01387 <br><br> 1.0 <br><br> nM <br><br> 1.81 <br><br> PR01387 <br><br> 10.0 <br><br> nM <br><br> 2.45 <br><br> 25 <br><br> 30 <br><br> 35 <br><br> EXAMPLE 58 Human Venous Endothelial CeH Ca Flux Assay This assay is designed to determine whether PRO polypeptides show the ability to stimulate calcium flux in human umbilical vein endothelial cells (HUVEC, Cell Systems). Ca influx is a well documented response upon binding of certain ligands to their receptors. A test compound that results in a positive response in the present Ca influx assay can be said to bind to a specific receptor and activate a biological signaling pathway in human endothelial cells. This could ultimately lead, for example to cell division, inhibition of cell proliferation, endothelial tube formation, cell migration, apoptosis, etc. <br><br> Human venous umbilical vein endothelial cells (HUVEC, Cell Systems) in growth media (50:50 without glycine, l%glutamine, lOmMHepes, 10%FBS, 10 ng/ml bFGF), were plated on %-well microtiter ViewPlates-96 (Packard Instrument Company Part #6005182) microtiter plates at a cell density of 2 x 10* cells/well. The day after plating, the cells were washed three times with buffer (HBSS plus lOmM Hepes), leaving 100 ^1/well. Then 100 ^l/well of 8 uM Fluo-3 (2x) was added. The cells were incubated for 1.5 hours at 37°C/5% C02. After incubation, the cells were then washed 3x with buffer (described above) leaving 100 ^l/well. Test samples of the PRO polypeptides were prepared on different 96-well plates at 5x concentration in buffer. The positive control <br><br> 177 <br><br> corresponded to 50 /zM ionomycin (5x); the negative control corresponded to Protein 32. Cell plate and sample plates were run on a FLIPR (Molecular Devices) machine. The FLIPR machine added 25 yt/l of test sample to the cells, and readings were taken even.' second for one minute, then every 3 seconds for the next three minutes. <br><br> The fluorescence change from baseline to the maximum rise of the curve (A change) was calculated, and replicates averaged. The rate of fluorescence increase was monitored, and only those samples which had a A change greater than 1000 and a rise within 60 seconds, were considered positive. In the following TABLE 9, the results are expressed relative to the positive control. <br><br> 10 <br><br> 15 <br><br> 20 <br><br> 25 <br><br> 30 <br><br> 35 <br><br> TABLE 9 <br><br> Human Venous Endothelial Cell Ca Flux Assay <br><br> PRO Name <br><br> PRO Concentration <br><br> Relative <br><br> PRO 179 <br><br> 0.01% <br><br> 1.0 <br><br> PRO 179 <br><br> 0.10% <br><br> 1.0 <br><br> PROl 79 <br><br> 1.0% <br><br> 3.0 <br><br> PROl 79 <br><br> 0.01% <br><br> 1.0 <br><br> PRO 179 <br><br> 0.10% <br><br> 1.0 <br><br> PRO 179 <br><br> 1.0% <br><br> 3.0 <br><br> PR0245 <br><br> 0.093 <br><br> nM <br><br> 1.0 <br><br> PR0245 <br><br> 0.93 <br><br> nM <br><br> 1.0 <br><br> PR0245 <br><br> 9.3 <br><br> nM <br><br> 2.0 <br><br> PR0771 <br><br> 0.075 <br><br> nM <br><br> 1.0 <br><br> PR0771 <br><br> 0.75 <br><br> nM <br><br> 1.0 <br><br> PR0771 <br><br> 7.5 <br><br> nM <br><br> 2.0 <br><br> PR013I3 <br><br> 0.611 <br><br> nM <br><br> 1.0 <br><br> PR01313 <br><br> 6.11 <br><br> nM <br><br> 1.0 <br><br> PROI313 <br><br> 61.1 <br><br> nM <br><br> 4.0 <br><br> PR013I3 <br><br> 0.611 <br><br> nM <br><br> 1.0 <br><br> PR01313 <br><br> 6.11 <br><br> nM <br><br> 1.0 <br><br> PR01313 <br><br> 61.1 <br><br> nM <br><br> 3.0 <br><br> PROl 376 <br><br> 0.86 <br><br> nM <br><br> 1.0 <br><br> PRO 1376 <br><br> 8.6 <br><br> nM <br><br> 1.0 <br><br> PRO 1376 <br><br> 86.0 <br><br> nM <br><br> 2.0 <br><br> PRO 1376 <br><br> 0.86 <br><br> nM <br><br> 1.0 <br><br> PR01376 <br><br> 8.6 <br><br> nM <br><br> 1.0 <br><br> PR01376 <br><br> 86.0 <br><br> nM <br><br> 2.0 <br><br> PRO 1561 <br><br> 0.023 <br><br> nM <br><br> 1.0 <br><br> PRO 1561 <br><br> 0.23 <br><br> nM <br><br> 1.0 <br><br> PROl 561 <br><br> 23.0 <br><br> nM <br><br> 2.0 <br><br> 178 <br><br> EXAMPLE 59 Induction of Endothelial Cell Apootosis The ability of PRO polypeptides to induce apoptosis in endothelial cells was tested in human venous umbilical vein endothelial cells (HUVEC, Cell Systems). <br><br> 5 Thecellswereplatedon 96-well microtiter plates (Amersham Life Science, cytostar-T scintillating microplate, <br><br> RPNQ160, sterile, tissue-culture treated, individually wrapped), in 10% serum (CSG-medium, Cell Systems), at a density of 2 x 104 cells per well in a total volume of 100 jul On day 2, test samples containing the PRO polypeptide were added in triplicate at dilutions of 1 %, 0.33% and 0.11 %. Wells without cells were used as a blank and wells with cells only were used as a negative control. As a positive control 1:3 serial dilutions of 50 ^1 of a 10 3x stock of staurosporine were used. The ability of the PRO polypeptide to induce apoptosis was determined by processing of the 96 well plates for detection of Annex in V, a member of the calcium and phospholipid binding proteins, to detect apoptosis. <br><br> 0.2 ml Annexin V - Biotin stock solution (100 Mg/ml) was diluted in 4.6 ml 2 x Ca2* binding buffer and 2.5% BSA (1:25 dilution). 50 of the diluted Annexin V - Biotin solution was added to each well (except controls) to 15 a final concentration of 1.0 /zg/ml. The samples were incubated for 10-15 minutes with Annexin-Biotin prior to direct addition of 35S-Streptavidin. 3,S-Streptavidin was diluted in 2x Ca24 Binding buffer, 2.5% BSA and was added to all wells at a final concentration of 3 x IO4 cpm/well. The plates were then sealed, centrifuged at 1000 rpm for 15 minutes and placed on orbital shaker for 2 hours. The analysis was performed on a 1450 Microbeta Trilux (Wallac). Percent above background represents the percentage amount of counts per minute above the 20 negative controls. Percents greater than or equal to 30% above background are considered positive. PROl78, PROl 79, PROl 88, PR0217. PR0261, PR0301, PR0538 and PR0719 gave positive results (induced endothelial cell apoptosis) in the above described assay. <br><br> EXAMPLE 60 Induction of Endothelial Cell Apoptosis (ELISA) <br><br> 25 The ability of PRO polypeptides to induce apoptosis in endothelial cells was tested in human venous umbilical vein endothelial cells (HUVEC. Cell Systems) using a 96-well format, in 0% serum media supplemented with 100 ng/ml VEGF, 0.1% BSA, IX penn/strep. The 96-well plates used were manufactured by Falcon (No. 3072). Coating of 96 well plates were prepared by allowing gelatinization to occur for &gt;30 minutes with 100 /il of 0.2% gelatin in PBS solution. The gelatin mix was aspirated thoroughly before plating HUVE cells at a final concentration of 2 30 x 10"' cells/ml in 10% serum containing medium - 100 tA volume per well. The cells were grown for 24 hours before adding test samples containing the PRO polypeptide of interest. <br><br> To all wells, 100 ^1 of 0% serum media (Cell Systems) complemented with 100 ng/ml VEGF, 0.1% BSA, IX penn/strep. was added. Test samples containing PRO polypeptides were added in triplicate at dilutions of 1%, 0.33% and 0.11%. Wells without cells were used as a blank and wells with cells only were used as a negative 35 control. As a positive control 1:3 serial dilutions of 50 of a 3x stock of staurosporine were used. The cells were incubated for 24 to 35 hours prior to ELISA. <br><br> 179 <br><br> ELISA was used to determine levels of apoptosis preparing solutions according to the Boehringer Manual [Boehringer, Cell Death Detection ELISA plus, Cat No. 1 920 685]. Sample preparations: 96 well plates were spun down at I krpm for 10 minutes (200g); the supernatant was removed by fast inversion, placing the plate upside down on a paper towel to remove residual liquid. To each well, 200 /ul of IX Lysis buffer was added and 5 incubation allowed at room temperature for 30 minutes without shaking. The plates were spun down for 10 minutes at 1 krpm, and 20 /il of the lysate (c\toplasmic fraction) was transferred into streptavidin coated MTP. 80 /il of immunoreagent mix was added to the 20 /il lystate in each well. The MTP was covered with adhesive foil and incubated at room tempearature for 2 hours by placing it on an orbital shaker (200 rpm). After two hours, the supernatant was removed by suction and the wells rinsed three times with 250 /il of 1X incubation buffer per well 10 (removed by suction). Substrate solution was added (100 /il) into each well and incubated on an orbital shaker at room temperature at 250 rpm until color development was sufficient for a photometric analysis (approx. after 10-20 minutes). A 96 well reader was used to read the plates at 405 nm, reference wavelength, 492 nm. The levels . obtained for PIN 32 (control buffer) was set to 100%. Samples with levels &gt;130% were considered positive for ' induction of apoptosis. PROl 72 and PR0235 gave positive results as measured by the ELISA assay described 15 above. <br><br> EXAMPLE 61 Detection of Endothelial Cell Apoptosis ( FACS1 <br><br> The ability of PRO polypeptides to induce apoptosis in endothelial cells was tested in human venous umbilical vein endothelial cells (HUVEC, Cell Systems) in gelatinized T175 flasks using HUVE cells below 20 passage 10. On day one, the cells were split [420,000 cells per gelatinized 6 cm dishes-(II x IO3 cells/cm2 Falcon, Primaria)] and grown in media containing serum (CS-C, Cell System) overnight or for 16 hours to 24 hours. <br><br> On day 2, the cells were washed I x with 5 ml PBS ; 3 ml of 0% serum medium was added with VEGF (100 ng/ml); and 30 //I of the PRO test compound (final dilution 1%) was added. The mixture containing cells, medium and test PRO compound were incubated for 48 hours before harvesting. <br><br> ^.25 The cells were then harvested for FACS analysis. The medium was aspired and the cells washed once with <br><br> PBS. 5 ml of 1 x trypsin was added to the cells in a T-175 flask, and the cells were allowed to stand until they were released from the plate (about 5-10 minutes). Trypsinization was stopped by adding 5 ml of growth media. The cells were spun at 1000 rpm for 5 minutes at 4°C. The media was aspirated and the cells were resuspended in 10 ml of 10% serum complemented medium (Cell Systems), 5 /il of Annexin-FITC (BioVison) added and chilled tubes 30 were submitted for FACS. <br><br> PR0331 and PR0364 gave positive results by the above described assay. <br><br> EXAMPLE 62 In situ Hybridization <br><br> In situ hybridization is a powerful and versatile technique for the detection and localization of nucleic acid 35 sequences within cell or tissue preparations. It may be useful, for example, to identify sites of gene expression, <br><br> 180 <br><br> analyze the tissue distribution of transcription, identify and localize viral infection, follow changes in specific mRNA synthesis, and aid in chromosome mapping. <br><br> In situ hybridization was performed following an optimized version of the protocol by Lu and Gillett, Cell Vision, h 169-176 (1994), using PCR-generated 33P-labeled riboprobes. Briefly, formalin-fixed, paraffin-5 embedded human tissues were sectioned, deparaffinized, deproteinated in proteinase K (20 g/ml) for 15 minutes at 37°C, and further processed for in situ hybridization as described by Lu and Gillett, supra. A (33-P)UTP-labeled antisense riboprobe was generated from a PCR product and hybridized at 55°C overnight. The slides were dipped in Kodak NTB2™ nuclear track emulsion and exposed for 4 weeks. <br><br> —P-Riboprobe synthesis <br><br> 10 6.0 mI (125 mCi) of 33P-UTP (Amersham BF 1002, SA&lt;2000 Ci/mmol) were speed-vacuum dried. To each tube containing dried "P-UTP, the following ingredients were added: <br><br> 2.0 fil 5x transcription buffer 1.0 //IDTT (100 mM) <br><br> 2.0 /d NTP mix (2.5 mM: 10 pi\ each of 10 mM GTP, CTP&amp; ATP + I0/^IH20) <br><br> 15 1.0 /J UTP (50/iM) <br><br> 1.0 fil RNAsin 1.0 /il DNA template (1 pig) <br><br> 1.0 &gt;ul H:0 <br><br> 1.0 /il RNA polymerase (for PCR products T3 = AS. T7 = S, usually) <br><br> 20 The tubes were incubated at 37°C for one hour. A total of 1.0 jwl RQ1 DNase was added, followed by incubation at 37 °C for 15 minutes. A total of 90 pi\ TE (10 mM Tris pH 7.6/1 mM EDTA pH 8.0) was added, and the mixture was pipetted onto DE81 paper. The remaining solution was loaded in a MICROCON-50™ ultrafiltration unit, and spun using program 10 (6 minutes). The filtration unit was inverted over a second tube and spun using program 2 (3 minutes). After the final recovery spin, a total of 100 TE was added, then 1 m! of the 25 final product was pipetted on DE81 paper and counted in 6 ml of BIOFLUOR II™. <br><br> The probe was run on a TBE/urea gel. A total of 1-3 ^1 of the probe or 5 tu\ of RNA Mrk III was added to 3 fA of loading buffer. After heating on a 95 °C heat block for three minutes, the gel was immediately placed on ice. The wells of gel were flushed, and the sample was loaded and run at 180-250 volts for 45 minutes. The gel was wrapped in plastic wrap (SARAN™ brand) and exposed to XAR film with an intensifying screen in a -70°C 30 freezer one hour to overnight. <br><br> -P-Hvbridization <br><br> A. Pretreatment of frozen sections <br><br> The slides were removed from the freezer, placed on aluminum trays, and thawed at room temperature for 5 minutes. The trays were placed in a 55 °C incubator for five minutes to reduce condensation. The slides were fixed 35 for 10 minutes in 4% paraformaldehyde on ice in the fume hood, and washed in 0.5 x SSC for 5 minutes, at room temperature (25 ml 20 x SSC + 975 ml SQ H20). After deproteination in 0.5 yug/'ml proteinase K for 10 minutes at 37°C (12.5 ^1 of 10 mg/ml stock in 250 ml prewarmed RNAse-free RNAse buffer), the sections were washed <br><br> 181 <br><br> in 0.5 x SSC for 10 minutes at room temperature. The sections were dehydrated in 70%. 95%, and 100% ethanol, 2 minutes each. <br><br> B. Pretreatment of paraffin-em bedded sections <br><br> The slides were deparaffinized, placed in SQ H:0, and rinsed twice in 2 x SSC at room temperature, for 5 5 minutes each time. The sections were deproteinated in 20 tug/ml proteinase K (500 p. 1 of 10 mg/ml in 250 ml RNase-free RNase buffer; 37°C, 15 minutes) for human embryo tissue, or 8 x proteinase K (100 ^1 in 250 ml Rnase buffer, 37°C, 30 minutes) for formalin tissues. Subsequent rinsing in 0.5 x SSC and dehydration were performed as described above. <br><br> C. Prehybridization <br><br> 10 The slides were laid out in a plastic box lined with Box buffer (4 x SSC, 50% formamide) - saturated filter paper. The tissue was covered with 50 ofhybridization buffer (3.75 g dextran sulfate + 6 ml SQ H,0), vortexed, and heated in the microwave for2 minutes with the cap loosened. After cooling on ice, 18.75 ml formamide, 3.75 ml 20 x SSC, and 9 ml SQ H;,0 were added, and the tissue was vortexed well and incubated at 42°C for 1-4 hours. <br><br> D. Hybridization <br><br> 15 1.0 x 106 cpm probe and 1.0 //I tRNA (50 riig/ml stock) per slide were heated at 95 "C for 3 minutes. The slides were cooled on ice, and 48 nI hybridization buffer was added per slide. After vortexing, 50 &gt;ul "P mix was added to 50 pi prehybridization on the slide. The slides were incubated overnight at 55°C. <br><br> E. Washes <br><br> Washing was done for 2x 10 minutes with 2xSSC, EDTA at room temperature (400 ml 20 x SSC + 16 ml 0.25 20 M EDTA, V,=4L), followed by RNAseA treatment at 37°C for 30 minutes (500 /xt of 10 mg/ml in 250 ml Rnase buffer = 20 Mg/ml), The slides were washed 2x10 minutes with 2 x SSC, EDTA at room temperature. The stringency wash conditions were as follows: 2 hours at 55°C, 0.1 x SSC, EDTA (20 ml20x SSC + 16 ml EDTA, Vr=4L). <br><br> F. Oligonucleotides <br><br> 25 in situ analysis was performed on twelve of the DNA sequences disclosed herein. The oligonucleotides employed for these analyses are as follows: <br><br> (1) DNA23339-1130 (PROl 78) <br><br> pi: <br><br> 5'-GGA TTC TAA TAC G AC TCA CTA TAG GGC CAC GGG CGC TGT GTG CTG GAG-3' (SEQ ID NO:231) <br><br> 30 p2: <br><br> 5'-CTA TG A A AT TAA CCC TCA CTA AAG GG A TGG TGG GG A CCG C AG GGT G AC-3'(SEQ ID N0.232) p3: <br><br> 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC CCG CCA CGA GGA GCT GTT ACG-3' (SEQIDNO:233) p4: <br><br> 35 5'-CTA TG A A AT TAA CCC TCA CTA AAG GG A TG A CCT GCA GGC ATG GGA G A A-3'(SEQ ID NO:234) p5: <br><br> 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC GGC CGC CAC GAG GAG CTG TTA-3' (SEQ IDNO:235) <br><br> 182 <br><br> P6: <br><br> 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA GGG GCT CTG GGG CTG GGT C-3' (SEQ ID NO:236) <br><br> (2) DNA28497-1130 (PROl m <br><br> 5 pi: <br><br> 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC CAA CAC CAA GGG GCA AGA TG-3' (SEQ ID NO:237) p2: <br><br> 5'-CTA TGA AAT TAA CCC TCA CTA AAG GGA GGG CTT TTG GTG GGA G AA GTT-3'(SEQ ID NO:238) <br><br> (3) DNA30942-1134 (PRQ212) <br><br> 10 pi: <br><br> 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC TCG CTG CTG TGC CTG GTG TTG-3' (SEQ ID NO:239) <br><br> 5-CTA TGA AAT TAA CCC TCA CTA A AG GGA CCG CTG CAG CCT CTT GAT GG A-3' (SEQ ID N0:240) <br><br> (4) DNA32286-1191 (PRQ2I4) <br><br> 15 pi: <br><br> 5-GGA TTC TAA TAC GAC TCA CTA TAG GGC CCC TCC TGC CTT CCC TGT CC-3'(SEQ ID NO:241) p2- <br><br> 5-CTA TGA AAT TAA CCC TCA CTA A AG GGA GTG GTG GCC GCG ATT ATC TGC-3' (SEQ 1D NO:242) <br><br> (5) DNA33094-1131 (PR0217&gt; <br><br> 20 pi: <br><br> 5-GGA TTC TAA TAC GAC TCA CTA TAG GGC TCA G A A AAG CGC AAC AGA GAA-3'(SEQ ID NO:243) <br><br> 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA TGT CTT CCA TGC CAA CCT TC-3' (SEQ ID NO:244) (6) DNA33221-1133 (PRQ224) <br><br> 25 pi: <br><br> 5-GGA TTC TAA TAC GAC TCA CTA TAG GGC GCA GCG ATG GCA GCG ATG AGG-3'(SEQ ID NO:245) <br><br> 5'-CTA TGA AAT TAA CCC TCA CTA AAG GGA CAG ACG GGG CAG CAG GGAGTG-3'(SEQ ID NO:246) (7) DNA35638-1141 (PR0245) <br><br> 30 pi: <br><br> 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC GGG AAG ATG GCG AGG AGG AG-3" (SEQ ID NO:247) <br><br> p2: <br><br> 183 <br><br> P2: <br><br> 5'-CTA TGA AAT TAA CCC TCA CTA AAG GGA CCA AGG CCA CAA ACG GA A ATC-3'(SEQ ID NO.248) <br><br> (8) DNA33473-1176 (PRQ26n pi: <br><br> 5 5 -GGA TTC TAA TAC GAC TCA CTA TAG GGC GCG AGG ACG GCG GCT TCA-3' (SEQ ID NO:249) P2: <br><br> 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA AGA GTC GCG GCC GCC CTT TTT-3' (SEQ ID N0:250) <br><br> (9) DNA40628-1216 fPRQ301) <br><br> Pi: <br><br> 10 5-GGA TTC TAA TAC GAC TCA CTA TAG GGC GAG TCC TTC GGC GGC TGT T-3' (SEQ ID NO:251) <br><br> } p2: <br><br> w 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA CGG GTG CTT TTG GGA TTC GTA-3' (SEQ ID NO:252) <br><br> (10) DNA47365-1206 (PRQ364) <br><br> pi: <br><br> 15 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC AAC CCG AGC ATG GCA CAG CAC-3' (SEQ lDNO:253) p2: <br><br> 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA TCT CCC AGC CGC CCC TTC TC-3' (SEQ ID NO:254) <br><br> (11) PNA29101-1122 (PRQ7I3) <br><br> pi: <br><br> 20 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC GGC GGA ATC CAA CCT GAG TAG-31 (SEQ ID NO:255) p2: <br><br> | 5'-CTA TGA AAT TAA CCC TCA CTA AAG GGA GCG GCT ATC CTC CTG TGC TC-3' (SEQ ID NO:256) <br><br> (12) DNA33087 (PRQ2I6^ <br><br> pi: <br><br> 25 5'-GGA TTC TAA TAC GAC TCA CTA TAG GGC CCC GAG TGT TTT CCA AGA-3" (SEQ ID NO:257) p2: <br><br> 5'-CTA TGA AAT TAA CCC TCA CTA AAG GGA CAA GTT TAC TAG CCC ATC CAT-3' (SEQ ID NO:258) <br><br> pj: <br><br> 5'-GG A TTC TAA TAC GAC TCA CTA TAG GGC TGG ATG GGC TAG TAA ACT TG A-3' (SEQ ID NO:259) 30 p4: <br><br> 5-CTA TGA AAT TAA CCC TCA CTA AAG GGA CCC TTC TGC TCC TTC TTG TT -3' (SEQ ID N0:260) <br><br> 184 <br><br> G. Results <br><br> In situ analysis was performed on the above twelve DNA sequences disclosed herein. The results from these analyses are as follows: <br><br> (1) DNA2339-1130 (PRO! 78) <br><br> In fetal lower limb, a distinctive expression pattern was observed at the connective tissue interface between skeletal muscle and bone (primitive periosteum). Expression was also seen adjacent to vascular tissue, thereby indicating a possible link with angiogenesis. In the body wall, a similar expression pattern to lower limb expression was observed. Expression was seen in the smooth muscle of the trachea. Also, expression was seen in the small intestine and stomach in smooth muscle/connective tissue of lamina propria. Cord vascular smooth muscle was also positive. The highly organized expression pattern in the developing limb, intestine and body wall suggests a distinctive role at these sites. Possibilities would include angiogenesis and patterning. <br><br> Possible increased expression appeared in the medulla of the fetal thymus as well as in fetal brain cerebral cortex (cortical neurones). The following fetal tissues did not show positive results: spinal cord, thyroid, adrenals, liver and placenta. <br><br> All adult tissues examined were negative. <br><br> (2) DNA28497-1130 (PRO 188) <br><br> In fetal lower limb, high expression was observed at sites of enchondral bone formation, in osteocytes and in periosteum/perichondrium of developing bones. This distribution suggests a role in bone formation/differentiation. A faint increase in expression was seen over thyroid epithelial cells. In the body wall, high expression was observed in osteocytes as well as in the periosteum/perichondrium of developing bones. This distribution suggests a role in bone formation and differentiation. No expression was seen in the following fetal tissues: thymus, trachea, brain (cerebral cortex), spinal cord, small intestine, adrenals, liver, stomach, placenta and cord. <br><br> In adult tissues, expression was seen over benign breast epithelium, areas of apocrine metaplasia and sclerosing adenosis. In addition, expression was seen over infiltrating breast ductal carcinoma eel Is. No expression was observed in adult liver, heart or hepatocellular carcinoma. <br><br> Possible expression appeared in adult squamous epithelium of skin and in adult adrenal cortex. All other tissues were negative. Fetal multiblock tissues included: liver, kidney, adrenals, thyroid, lungs, heart, great vessels, small intestine, spleen, thymus, pancreas, brain, spinal cord, body wall, pelvis and lower limb. Adult multiblock tissues included: liver, kidney, adrenal, myocardium, aorta, spleen, lymph node, pancreas, lung and skin. <br><br> (3) DNA30942-1134 (PRQ212) <br><br> Expression in Lung Carcinomas, Tumor Block, and Breast Carcinomas <br><br> Expression was observed in mononuclear phagocytes in the normal chimp thymus, as well as in one out of one gastric carcinomas, one out of one colorectal cancers, two out of five breast cancers and one out of four lung cancers examined. Expression was observed by malignant cells in an osteosarcoma and a poorly differentiated <br><br> 185 <br><br> liposarcoma. A possible signal was seen in the malignant cells of a testicular teratoma and one out of five breast cancers examined. In one of the lung cancers, a scattered signal was seen over a high endothelial venule within pulmonary lymphoid tissue. <br><br> Fetal tissues examined (E12-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, thyroid, 5 lung, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal cord, body wall, pelvis and lower limb. Adult human tissues examined included: liver, kidney, adrenals, myocardium, aorta, spleen, lung, skin, chondrosarcoma, eye, stomach, gastric carcinoma, colon, colonic carcinoma, renal cell carcinoma, prostate, bladder mucosaand gall bladder, and acetominophen induced liver injury and hepatic cirrhosis. Rhesus tissues examined included cerebral cortex (rm) and hippocampus (rm). Chimp tissues examined included: 10 thyroid, parathyroid, ovary, nerve, tongue, thymus, adrenal gastric mucosa and salivary gland. <br><br> Expression in Lung Adenocarcinoma and Squamous Carcinomas <br><br> Eight adenocarcinomas and seven squamous lung carcinomas were examined. Actins were strongly positive in all tumors, indicating that all were suitable for in situ hybridization analysis. Expression was observed in six of the tumors as follows: <br><br> 15 6727-95 squamous carcinoma - strongly expressed over neoplastic epithelium 9558-95 squamous carcinoma - expression over neoplastic epithelium 12235-95 adenocarcinoma - expression over in situ and infiltrating tumor cells <br><br> 6545-95 &amp; 4187-97 squamous carcinomas <br><br> - expression over cells in tumor stroma, no expression seen over tumor cells 20 12954-94 squamous carcinoma - possible weak expression over stromal cells <br><br> (4) PNA32286-1191 (PRQ2141 <br><br> In fetal tissue, low level expression was seen throughout the mesenchyme. Moderate expression was observed in placenta) stromal cells in membraneous tissues and in the thyroid. Low level expression was also seen in cortical neurones. Adult tissues were all negative. <br><br> 25 Fetal tissues examined (E12-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, thyroid, <br><br> lungs, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal cord, body wall, pelvis and lower limb. Adult tissues examined included: liver, kidney, adrenal, myocardium, aorta, spleen, lymph node, pancreas, lung and skin. <br><br> (5) DNA33Q94-1131 (PRQ217) <br><br> 30 A highly distinctive expression pattern was observed as follows: in the human embryo, expression was observed in the outer smooth muscle layer of the G1 tract, respiratory cartilage, branching respiratory epithelium, osteoblasts, tendons, gonad, in the optic nerve head and developing dermis. In the adult, expression was observed in the epidermal pegs of the chimp tongue, the basal epithelial/myoepithelial cells of the prostate and urinary bladder. Also, expression was seen in the alveolar lining cells of the adult lung, mesenchymal cells juxtaposed to 35 erectile tissue in the penis and the cerebral cortex (probably glial cells). In the kidney, expression was only seen <br><br> 186 <br><br> in disease, in cells surrounding thyroidized renal tubules. <br><br> Human fetal tissues examined (E12-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, thyroid, lung, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal cord, body wall, pelvis and lower limb. Adult human tissues examined included: kidney (normal and end stage), 5 adrenals, myocardium, aorta, spleen, lymph node, gall bladder, pancreas, lung, skin, eye (including retina), prostate, bladder, and liver (normal, cirrhotic, acute failure). <br><br> Non-human primate tissues examined included: Chimp tissues (salivary gland, stomach, thyroid, parathyroid, skin, thymus, ovary, lymph node): Rhesus Monkey tissues (cerebral cortex, hippocampus, cerebellum, penis). <br><br> (6) DNA3322I-1133 (PRQ224) <br><br> 10 Expression was limited to vascular endothelium in fetal spleen, adult spleen, fetal liver, adult thyroid and adult lymph node (chimp). Additional site of expression was seen in the developing spinal ganglia. All other tissues k were negative. <br><br> " Human fetal tissues examined (EI2-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, <br><br> thyroid, lung, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal 15 cord, body wall, pelvis and lower limb. Adult human tissues examined included: kidney (normal and end-stage), adrenals, myocardium, aorta, spleen, lymph node, pancreas, lung, skin, eye (including retina), bladder, liver, (normal, cirrhotic, acute failure). Non-human primate tissues examined included: Chimp tissues (salivary gland, stomach, thyroid, parathyroid, skin, thymus, ovary, lymph node); Rhesus Monkey tissues (cerebral cortex, hippocampus, cerebellum, penis). <br><br> 20 (7) DNA35638-1141 (PRQ24S^ <br><br> Expression Pattern in Human Adult and Fetal Tissues <br><br> Expression was observed in the endothelium lining a subset of fetal and placental vessels. Endothelial expression was confined to these tissue blocks. Expression was also observed over intermediate trophoblast cells k of the placenta. All other tissues w ere negative. <br><br> 25 Fetal tissues examined (E12-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, thyroid, <br><br> lung, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal cord, body wall, pelvis and lower limb. Adult tissues examined included: liver, kidney, adrenals, myocardium, aorta, spleen, lymph node, pancreas, lung, skin, cerebral cortex (rm), hippocampus (rm), cerebellum (rm), penis, eye, bladder,stomach,gastriccarcinoma.colon, colonic carcinoma, thyroid (chimp), parathyroid (chimp), ovary (chimp) 30 and chondrosarcoma, acetominophen induced liver injury and hepatic cirrhosis. <br><br> Expression in Inflamed Tissues (Psoriasis, 1BD, Inflamed Kidney, Inflamed Lung. Hepatitis (Liver Block), Normal Tonsil, Adult and Chimp Multiblocksi <br><br> This molecule has been shown to be immunostimulatory (enhances T lymphocyte proliferation in the MLR and costimulation) and has proinflammatory properties (induces a neutrophil infiltrate in vivo). As indicated above, 35 this molecule has been shown to be expressed on a subset of fetal vessels in the endothelium/intima and in the <br><br> 187 <br><br> placenta but was not found to be expressed in a variety of normal adult human tissues or vessels in those tissues. An evaluation was performed for the expression of this molecule in vessels of inflamed human tissues as compared to non-inflammed tissues. In summary, expression was seen in the endothelium/intima of large vessels in the lung afflicted with chronic inflammation, in the superficial dermal vessels of psoriatic skin, in arterioles in a specimen 5 of chronic sclerosing nephritis and in capillaries including the perifollicular sinuses of tonsil. <br><br> Expression was not observed in normal skin (human foreskin specimens), normal lung, inflamed (eight IBD specimens) or normal large bowel, chronically inflamed or cirrhotic liver, normal adult cardiac tissue, or adrenal gland. <br><br> (8) DNA33473-1176 (PRQ26H <br><br> 10 Expression Pattern in Human Adult and Fetal Tissues <br><br> Strong expression was observed in dermal fibroblasts in normal adult skin. Strong expression was also seen in two cirrhotic livers, at sites of active hepatic fibrosis. In addition, moderate expression was seen over fasiculata W cells of the adrenal cortex. Localization of expression supports a role for this molecule in extracellular matrix formation/turnover. <br><br> 15 Expression in Human Breast Carcinoma and Normal Breast Tissue, and in Lung Carcinoma <br><br> A weak diffuse signal was seen in two breast tumors examined. No expression was seen on benign and malignant epithelial cells, but specific hybridization was observed in mesenchymal cells, particularly in areas of tissue repair including dystrophic ossification. The signal appears to be localized to the same cell population, however in some areas (breast tumor 02), the signal was significantly strong. Most positive cells have the <br><br> 20 morphology of fibroblasts, but smooth muscle cells appear to be negative. The signal was less intense in lung tumor tissue, however, this section showed less tissue repair compared with the breast tumor slides. Normal lung and kidney tissue were essentially negative. <br><br> In summary, this study showed expression in mesenchymal cells involved in tissue repair and/or collagen deposition. The signal was particularly strong in benign fibroblast-like cells adjacent to either infiltrating breast . 25 carcinoma cells or tissue destruction due to benign, inflammatory conditions (duct rupture). Of note, is the fact that deposition of benign osteoid seems to correlate with strong expression of RNA. <br><br> Expression in Normal Human Colon and Colon Carcinoma <br><br> None of the tissue sections showed a positive hybridization signal in tumor cells. Positive signals of variable intensity were observed in mesenchymal cells of either fibroblast or smooth muscle differentiation. Fibroblasts <br><br> 30 with a positive signal was observed adjacent to invasive tumor, if this tumor elicits a so called desmoplastic response (fibroblast proliferation with deposition of collagenous fibrosis). Positive fibroblasts were also seen in areas of tissue repair (granulation tissue or granulomatous response). Positive smooth muscle cells were seen in mostly arterial vessels of medium size. <br><br> (9) DNA40628-12I6 (PRQ30H <br><br> 35 Expression in Inflamed Human Tissues (Psoriasis, IBD, Inflamed Kidney, Inflamed Lung, Hepatitis, Normal Tonsil, <br><br> 188 <br><br> Adult and Chimp Multiblocks) <br><br> Expression was evaluated in predominantly inflamed human tissue with few normal human and non-human primate tissues. Expression was seen in every epithelial structure evaluated including the mucosal epithelium of the colon, bronchial large airway epithelium, oral mucosa (tongue), tonsillar crypt mucosa, placental mucosa, 5 prostatic mucosa, glandular stomach mucosa, epithelial cells of thymic Hassall's corpuscles, hepatocytes, biliary epithelium, and placental epithelium. The only evidence for expression outside of an epithelial structure was weak low, inconsistent expression in the germinal centers of follicles in a tonsil with reactive hyperplasia. <br><br> In non-human primate tissues (chimp): weak diffuse expression was seen in the epidermis of tongue epithelium; weak expression was seen in thymic epithelium of Hassall's corpuscles; and in the stomach, mild 10 diffuse expression in the epithelium of the glandular mucosa was observed. <br><br> In human tissues: <br><br> (1) in liver (multiblock including: chronic cholangitis, lobular hyperplasia, acetominophen toxicity), there was a ^ diffuse, low to moderate, expression in hepatocytes and biliary epithelium. Expression was most prominent in perilobular/periportal hepatocytes. It was most prominent in biliary epithelium in sections of liver with chronic 15 sclerosing cholangitis. <br><br> (2) Weak expression was observed in psoriasis samples in the epidermis. <br><br> (3) In lung with chronic interstitial pneumonia or chronic bronchitis, low diffuse expression was seen in the mucosal epithelium of large airways: weak diffuse expression was also seen in alveolar epithelium. There was no expression in the epithelium of the submucosal glands of bronchi/bronchioles. <br><br> 20 (4) Moderate diffuse expression was observed in both placental epithelium and in the mucosal epithelium of the gall bladder. <br><br> (5) In prostate epithelium, low diffuse expression was seen. <br><br> (6) High diffuse expression was observed in the epithelium of the tonsillar mucosa and crypts; the signal was highest in the mucosal cells which line the tonsillar crypts. There was weak inconsistent diffuse expression in the <br><br> 25 germinal centers of cortical folicles (B lymphocyte areas); however, in no other tissue evaluated with lymphoid | structures or lymphocytic inflammation was there any expression in B lymphocytes. <br><br> (7) In colon with inflammatory bowel disease and polyp/adenomatous change: low expression in the mucosal epithelium was seen with expression greatest in the villi tips. In one specimen with a polyp, there was no evidence of increased expression in the dysplastic epithelium ofthe polyp as compared to the adjacent mucosa. There was <br><br> 30 no apparent expression in reactive mucosal lympohid tissue that was present in many of the sections. <br><br> Tissues with no expression included: heart, peripheral nerve, and muscle (cardiac, smooth). <br><br> (10) DNA47365-1206 (PR(Ti64) <br><br> Expression was observed in the fetus, in the fascia lining the anterior surface of the vertebral body. Expression was also seen over the fetal retina. Low level expression occurred over fetal neurones. All other tissues 35 were negative. <br><br> 189 <br><br> Ml) DNA29101-1122 fPRQ713&gt; <br><br> In fetal tissues: Expression was observed in developing lower limb bones at the edge of the cartilagenous anlage (i.e. around the outer edge): in developing tendons, in vascular smooth muscle and in cells embracing developing skeletal muscle myocytes and myotubes. Expression was also observed at the epiphyseal growth plate. 5 In fetal lymph node, expression was seen in the marginal sinus of developing lymph nodes. Expression was observed in the subcapsular region ofthe thymic cortex, possibly representing either the subcapsular epithelial cells or the proliferating thymocytes that are found in this region. Expression was seen additionally in the following tissues: expression in the smooth muscle of the trachea; focal expression in cortical neurones in the brain cerebral cortex; expression in the smooth muscle of the small intestine; generalized expression over the thyroid epithelium; 10 expression in ductal plate cells of liver; expression in mural smooth muscle ofthe stomach; expression in the basal layer of squamous epithelium in fetal skin; expression in interstitial cells in trophoblastic villi in the placenta; and expression in wall of arteries and veins in the umbilical cord. Expression patterns suggest that this molecule may . be involved in cell differentiation/proliferation. <br><br> No expression was observed in fetal spleen, spinal cord, or adrenals. <br><br> 15 High expression was observed in the following sites: <br><br> (1) chimp ovary - granulosa cells of maturing follicles, lower intensity signal was observed over thecal cells; <br><br> (2) Chimp parathyroid - high expression was seen over chief cells; <br><br> (3) Human fetal testis - moderate expression was seen over stromal cells surrounding developing tubules; <br><br> (4) Human fetal lung - high expression was seen over chondrocytes in developing bronchial tree, and low level 20 expression was seen over branching bronchial epithelium. <br><br> Specific expression was not observed over the renal cell, gastric and colonic carcinomas. <br><br> Fetal tissues examined (E12-E16 weeks) included: placenta, umbilical cord, liver, kidney, adrenals, thyroid, lungs, heart, great vessels, oesophagus, stomach, small intestine, spleen, thymus, pancreas, brain, eye, spinal cord, body wall, pelvis and lower limb. Adult tissues examined included: liver, kidney, adrenals, myocardium, aorta, 25 spleen, lymph node, pancreas, lung, skin, cerebral cortex (rm), hippocampus (rm), cerebellum (rm), penis, eye, bladder, stomach, gastric carcinoma, colon, colonic carcinoma and chondrosarcoma, acetominophen induced liver injury and hepatic cirrhosis. <br><br> (12) DNA33087 (PRQ2I6) <br><br> Strong specific expression was seen in osteoblasts at all sites of enchondral and periosteal new bone 30 formation. Additional sites of expression included the developing pulmonary arterial and aortic trunks. Otherwise, all fetal tissues were negative. Tissues examined included: placenta, umbilical cord, brain, spinal cord, eye, optic nerve, trachea, lung, heart, thymus, liver, spleen, esophagus, small intestine, pancreas, adrenals, thyroid, body wall and lower limb. <br><br> Adult tissues examined were all negative and included: liver, kidney, adrenals, myocardium, aorta, spleen, 35 lymph node, pancreas, lung and skin. All adult tissues in the multiblock were positive for beta-actin. <br><br> This molecule's probable role is in control of bone matrix deposition and/or osteoblast growth. <br><br> 190 <br><br> EXAMPLE 63 Use of PRO Polypeptides as a Hybridization Probe The following method describes use of a nucleotide sequence encoding PRO polypeptides as a hybridization probe. <br><br> 5 DNA comprising the coding sequence of full-length or mature PRO polypeptides (as shown in Figures I, 3, <br><br> 5, 7, 9, I I, 13, 15, 17, 19,21,23,25.27,29,31, 33,35,37,39,41,43,45,47,49,51,53, 55, 57, 59,61,63,65, 67,69, 71, 73,75,77, 79,81, 83. 85. 87, 89, 91,93, and 95, respectively, SEQ ID NOS: 3, 8, 13, 15,20,25, 30, 35,40,45,50,55,61,66,71,76,84,89,97,106,111,116,126,131,136,142, 147,152, 154, 159, 161,169,180, 182, 190,192, 194, 196, 198,200.202,204,213,215,217,219,221 and 226, respectively) or a fragment thereof 10 is employed as a probe to screen for homologous DNAs (such as those encoding naturally-occurring variants of PRO) in human tissue cDNA libraries or human tissue genomic libraries. <br><br> Hybridization and washing of filters containing either library DNAs is performed under the following high-stringency conditions. Hybridization of radiolabeled probe derived from the gene encoding a PRO polypeptide to ^ the filters is performed in a solution of 50% formamide, 5xSSC, 0.1% SDS, 0.1% sodium pyrophosphate, 50 mM 15 sodium phosphate, pH 6.8,2x Denhardt's solution, and 10% dextran sulfate at 42°C for 20 hours. Washing of the filters is performed in an aqueous solution of 0.1 x SSC and 0.1 % SDS at 42°C. <br><br> DNAs having a desired sequence identity with the DNA encoding full-length native sequence can then be identified using standard techniques known in the art. <br><br> EXAMPLE 64 <br><br> 20 Expression of Nucleic Acid Encoding PRO Polypeptides in E. coli <br><br> This Example illustrates preparation of an unglycosylated form of PRO polypeptides by recombinant expression in E. coli. <br><br> The DNA sequence encoding PRO 187, PRO 195, PR0224, PR0301, PR0364, PR0713, PR078 8, PRO 1274, PRO 1286, PROl 303, PRO 1312. PRO 1313, and PRO 1376 (SEQ ID NOS: 20,30.50.89,116,136,152,196,198, 25 202, 213, 215, and 217, respectively) was initially amplified using selected PCR primers. The primers should contain restriction enzyme sites that correspond to the restriction enzyme sites on the selected expression vector. A variety of expression vectors may be employed. An example of a suitable vector is pBR322 (derived from E. coli; see Bolivar et ai, Gene. 2: 95 (1977)), which contains genes for ampicillin and tetracycline resistance. The vector was digested with restriction enzyme and dephosphorylated. The PCR-amplified sequences were then 30 ligated into the vector. The vector will preferably include sequences that encode an antibiotic-resistance gene, a trp promoter, a poly-His leader (including the first six STII codons, poly-His sequence, and enterokinase cleavage site), the region encoding PR01S7. PR0195, PR0224, PR0301, PR0364. PR07I3, PR0788, PR01274, PR01286, PR01303, PROD 12. PR01313, and PR01376. lambda transcriptional terminator, and an argU gene. The ligation mixture was then used to transform a selected E. coli strain using the methods described in 35 Sambrook et al., supra. Transformants were identified by their ability to grow on LB plates and antibiotic-resistant colonies were then selected. Plasmid DNA was isolated and confirmed by restriction analysis and DNA <br><br> 191 <br><br> sequencing. <br><br> Selected clones were grown overnight in liquid culture medium such as LB broth supplemented with antibiotics. The overnight culture may subsequently be used to inoculate a larger-scale culture. The cells were then grown to a desired optical density, during which the expression promoter was turned on. <br><br> 5 After culturing the cells for several more hours, the cells were harvested by centrifugation. The cell pellet obtained by the centrifugation was solubilized using various agents known in the art, and the solubilized PRO 187, PR0195,PR0224, PR0301,PR0364.PR0713,PR0788. PR01274, PR01286, PROl 303, PROl 312, PR01313, and PRO 1376 polypeptides were then purified using a metal-chelating column under conditions that allow tight binding of the polypeptide. <br><br> 10 EXAMPLE 65 <br><br> Expression of Nucleic Acid Encoding PRO Polypeptides in Mammalian Cells <br><br> This Example illustrates preparation of a potentially glycosylated form of PRO polypeptides by recombinant expression in mammalian cells. <br><br> The vector, pRK5 {see. EP 307.247, published March 15, 1989), is employed as the expression vector. 15 Optionally, the PRO DNA is ligated into pRK5 with selected restriction enzymes to allow insertion of the DNA encoding PRO polypeptides using ligation methods such as described in Sambrook et ai, supra. The resulting vector is called pRK5-(DNA encoding PRO). <br><br> In one embodiment, the selected host cells are 293 cells. Human 293 cells (ATCC CCL 1573) are grown to confluence in tissue culture plates in medium such as DMEM supplemented with fetal calf serum and optionally, 20 nutrient components and'or antibiotics. About 10 fig DNA of pRK5-(DNA encoding PRO) is mixed with about 1 fig DNA encoding the VA RNA gene (Thimmappaya et a!.. Cell. 31: 543 (1982)) and dissolved in 500 fA of 1 mM Tris-HCl. 0.1 mM EDTA, 0.227 M CaCk. To this mixture is added, dropwise, 500 pi\ of 50 mM HEPES (pl l 7.35), 280 mM NaCl, 1.5 mM NaPOj. and a precipitate is allowed to form for 10 minutes at 25°C. The precipitate is suspended and added to the 293 cells and allowed to settle for about four hours at 37"C. The culture medium is <br><br> • 25 aspirated off and 2 ml of 20% glycerol in PBS is added for 30 seconds. The 293 cells are then washed with serum-free medium, fresh medium is added, and the cells are incubated for about 5 days. <br><br> Approximately 24 hours after the transfections, the culture medium is removed and replaced with culture medium (alone) or culture medium containing 200 ftCi/ml 35S-cysteine and 200 ^Ci/ml 35S-methionine. After a 12-hour incubation, the conditioned medium is collected, concentrated on a spin filter, and loaded onto a 15% SDS 30 gel. The processed gel may be dried and exposed to film for a selected period of time to reveal the presence of the PRO polypeptide. The cultures containing transfected cells may undergo further incubation (in serum-freemedium) and the medium is tested in selected bioassays. <br><br> In an alternative technique, the gene encoding the PRO polypeptide may be introduced into 293 cells transiently using the dextran sulfate method described by Somparyrac et al.. Proc. Natl. Acad. Sci.. 12:7575 (198IV 35 293 cells are grown to maximal density in a spinner flask and 700 /ug pRK5-(DNA encoding PRO) is added. The cells are first concentrated from the spinner flask by centrifugation and washed with PBS. The DNA-dextran <br><br> 192 <br><br> precipitate is incubated on the cell pellet for four hours. The cells are treated with 20% glycerol for 90 seconds, washed with tissue culture medium, and re-introduced into the spinner flask containing tissue culture medium, 5 ^g/ml bovine insulin, and 0.1 Mg/ml bovine transferrin. After about four days, the conditioned media is centrifuged and filtered to remove cells and debris. The sample containing the expressed gene encoding the PRO polypeptide 5 can then be concentrated and purified by any selected method, such as dialysis and/or column chromatography. <br><br> In another embodiment, the gene encoding the PRO polypeptide can be expressed in CHO cells. The pRK.5-(DNA encoding PRO) nucleic acid can be transfected into CHO cells using known reagents such as CaP04 or DEAE-dextran. As described above, the cell cultures can be incubated, and the medium replaced with culture medium (alone) or medium containing a radiolabel such as 35S-methionine. After determining the presence of the 10 PRO polypeptide, the culture medium may be replaced with serum-free medium. Preferably, the cultures are incubated for about 6 days, and then the conditioned medium is harvested. The medium containing the expressed PRO polypeptide can then be concentrated and purified by any selected method. <br><br> Epitope-tagged gene encoding the PRO polypeptide may also be expressed in host CHO cells. The gene ^ encoding the PRO polypeptide may be subcloned out of the pRK5 vector. The subclone insert can undergo PCR 15 amplification to fuse in frame with a selected epitope tag such as a poly-His tag into a baculovims expression vector. The gene insert encoding the poly-His-tagged-PRO polypeptide can then be subcloned into a SV40- driven vector containing a selection marker such as DHFR for selection of stable clones. Finally, the CHO cells can be transfected (as described above) with the SV40-driven vector. Labeling may be performed, as described above, to verify expression. The culture medium containing the expressed gene encoding the poly-His-tagged-PRO 20 polypeptide can then be concentrated and purified by any selected method, such as by Np-chelate affinity chromatography. <br><br> PROl 72, PROl 78, PRO 179, PR0182, PR0188, PR0212, PR0214, PR0216, PR0217, PR0224, PR0245, PR0261, PR0301, PR0356, PR0364, PR0713, PR0788, PR0792, PR0865, PROl 126, PROl 130, PR01244, PROl 246, PRO 1274, PRO 1286, PRO 1294, PRO 1303, PRO 1304, PROl 376, and PRO 1387 were stably expressed 25 in CHO cells by the above described method. In addition, PROI72, PROl78, PROI82, PR023I, PR0245, PR0301, PR0356 and PR0364 were expressed in CHO cells by a transient procedure. <br><br> EXAMPLE 66 <br><br> Expression ofNucleic Acid Encoding PRO Polypeptides in Yeast <br><br> The following method describes recombinant expression of the gene encoding PRO polypeptides in yeast. 30 First, yeast expression vectors are constructed for intracellular production or secretion of the PRO polypeptide from the ADH2/GAPDH promoter. DNA encoding the PRO polypeptide and the promoter is inserted into suitable restriction enzyme sites in the selected plasmid to direct intracellular expression of the gene encoding the PRO polypeptide. For secretion, DNA encoding the PRO polypeptide can be cloned into the selected plasmid, together with DNA encoding the ADH2/GAPDH promoter, a native PRO polypeptide signal peptide or other mammalian 35 signal peptide, or, for example, a yeast alpha-factor or invertase secretory signal/leader sequence, and linker sequences (if needed) for expression of the gene encoding the PRO polypeptide. <br><br> 193 <br><br> Yeast cells, such as yeast strain AB110. can then be transformed with the expression plasmids described above and cultured in selected fermentation media. The transformed yeast supernatants can be analyzed by precipitation with 10% trichloroacetic acid and separation by SDS-PAGE, followed by staining of the gels with Coomassie Blue stain. <br><br> 5 Recombinant PRO polypeptides can subsequently be isolated and purified by removing the yeast cells from the fermentation medium by centrifugation and then concentrating the medium using selected cartridge filters. The concentrate containing the PRO polypeptide may further be purified using selected column-chromatography resins. <br><br> EXAMPLE 67 <br><br> Expression of Nucleic Acid Encoding PRO Polypeptides in Baculovirus-lnfected Insect Cells 10 The following method describes recombinant expression in Baculovirus-infected insect cells. <br><br> The sequence coding for the PRO polypeptide is fused upstream of an epitope tag contained within a baculovirus expression vector. Such epitope tags include poly-His tags and immunoglobulin tags (like Fc regions " of IgG). A variety of plasmids may be employed, including plasmids derived from commercially available plasmids such as pVL1393 (Novagen). Briefly, the sequence encoding the PRO polypeptide or the desired portion 15 of the coding sequence of the PRO polypeptide [such as the sequence encoding the extracellular domain of a transmembrane protein or the sequence encoding the mature protein if the protein is extracellular] is amplified by PCR with primers complementary to the 5' and 3' regions. The 5' primer may incorporate flanking (selected) restriction enzyme sites. The product is then digested with those selected restriction enzymes and subcloned into the expression vector. <br><br> 20 Recombinant baculovirus is generated by co-transfecting the above plasmid and BacuIoGoldTM virus DNA <br><br> (Pharmingen) intoSpodopterafrugiperda ("Sf9") cells (ATCCCRL171 l)usinglipofectin (commercially available from GIBCO-BRL). After 4-5 days of incubation at 28 °C, the released viruses are harvested and used for further amplifications. Viral infection and protein expression are performed as described by O'Reilley et a!., Baculovirus Expression Vectors: A Laboratory Manual (Oxford: Oxford University Press (1994)). <br><br> 25 Expressed poly-His tagged-PRO polypeptides can then be purified, for example, by Ni ^'-chelate affinity chromatography as follows. Extracts are prepared from recombinant virus-infected Sf9 cells as described by Rupert et al.. Nature. 362:175-179 (1993). Briefly, Sf9 cells are washed, resuspended in sonication buffer (25 ml Hepes, pH 7.9; 12.5 mM MgCU; 0.1 mM EDTA; 10% glycerol; 0.1% NP-40; 0.4 M K.CI), and sonicated twice for 20 seconds on ice. The sonicates are cleared by centrifugation, and the supernatant is diluted 50-fold in loading buffer 30 (50 mM phosphate, 300 mM NaCl. 10% glycerol, pH 7.8) and filtered through a 0.45 p.m filter. A Ni 2*-NTA agarose column (commercially available from Qiagen) is prepared with a bed volume of 5 ml, washed with 25 ml of water and equilibrated with 25 ml of loading buffer. The filtered cell extract is loaded onto the column at 0.5 ml per minute. The column is washed to baseline A,g0 with loading buffer, at which point fraction collection is started. Next, the column is washed with a secondary wash buffer (50 mM phosphate; 300 mM NaCl, 10% 35 glycerol, pH 6.0), which elutes non-specifically-bound protein. After reaching an A2g0 baseline again, the column is developed with a 0 to 500 mM imidazole gradient in the secondary wash buffer. One ml fractions are collected <br><br> 194 <br><br> and analyzed by SDS-PAGE and silver staining or Western blot with Ni 2~-NTA-cdnjugated to alkaline phosphatase (Qiagen). Fractions containing the eluted His,0-tagged-PRO polypeptide are pooled and dialyzed against loading buffer. <br><br> Alternatively, purification of the JgG-tagged (or Fc tagged)-PRO polypeptide can be performed using known 5 chromatography techniques, including for instance, Protein A or protein G column chromatography. <br><br> PRO 172, PRO 178, PR0214. PR0216, PR0231, PR0235, PR0269, PR0301, PR0356, PR0538, PR0719, and PR01376 were expressed in baculovirus infected Sf9 insect cells. While expression was actually performed in a 0.5-2 L scale, it can be readily scaled up for larger (e.g., 8 L) preparations. The proteins are expressed as an IgG construct (immunoadhesin), in which the protein extracellular region is fused to an IgGl constant region 10 sequence containing the hinge, CH2 and CH3 domains and/or in poly-His tagged forms. <br><br> Following PCR amplification, the respective coding sequences are subcloned into a baculovirus expression vector (pb.PH.IgG for IgG fusions and pb.PH.His.c for poly-His tagged proteins), and the vector and Baculogold® baculovirus DNA (Pharmingen) are co-transfected into 105 Spodoptera frugiperda ("Sf9") cells (ATCC CRL " 1711), using Lipofectin (Gibco BRL). pb.PH.IgG and pb.PH.His are modifications of the commercially available 15 baculovirus expression vector pVL 1393 (Pharmingen), with modified polylinker regions to include the His or Fc tag sequences. The cells are grown in Hink's TNM-FH medium supplemented with 10% FBS (Hyclone). Cells are incubated for 5 days at 28 °C. The supernatant is harvested and subsequently used for the first viral amplification by infecting Sf9 cells in Hink's TNM-FH medium supplemented with 10% FBS at an approximate multiplicity of infection (MOI) of 10. Cells are incubated for 3 days at 28 °C. The supernatant is harvested and 20 the expression of the constructs in the baculovirus expression vector is detemiined by batch binding of 1 ml of supernatant to 25 ml of Ni :~-NTA beads (QIAGEN) for histidine tagged proteins or Protein-A Sepharose CL-4B beads (Pharmacia) for IgG tagged proteins followed by SDS-PAGE analysis comparing to a known concentration of protein standard by Coomassie blue staining. <br><br> The first viral amplification supernatant is used to infect a spinner culture (500 ml) of Sf9 cells grown in 25 ESF-921 medium (Expression Systems LLC) at an approximate MOI of 0.1. Cells are incubated for 3 days at I 28 °C. The supernatant is harvested and filtered. Batch binding and SDS-PAGE analysis is repeated, as necessary, until expression of the spinner culture is confirmed. <br><br> The conditioned medium from the transfected cells (0.5 to 3 L) is harvested by centrifugation to remove the cells and filtered through 0.22 micron filters. For the poly-His tagged constructs, the protein construct is purified 30 using a Ni 2"-NTA column (Qiagen). Before purification, imidazole is added to the conditioned media to a concentration of 5 mM. The conditioned media is pumped onto a 6 ml Ni 2~-NTA column equilibrated in 20 mM Hepes, pH 7.4, buffer containing 0.3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml/min. at 4°C. After loading, the column is washed with additional equilibration buffer and the protein eluted with equilibration buffer containing 0.25 M imidazole. The highly purified protein is subsequently desalted into a storage buffer containing 35 10 mM Hepes, 0.14 M NaCl and 4% mannitol, pH 6.8, with a 25 ml G25 Superfine (Pharmacia) column and stored at -80°C. <br><br> Immunoadhesin (Fc containing) constructs of proteins are purified from the conditioned media as follows. <br><br> 195 <br><br> The conditioned media is pumped onto a 5 ml Protein A column (Pharmacia) which has been equilibrated in 20 mM Na phosphate buffer, pH 6.8. After loading, the column is washed extensively with equilibration buffer before elution with 100 mM citric acid, pH 3.5. The eluted protein is immediately neutralized by collecting 1 ml fractions into tubes containing 275 ml of 1 M Tris buffer, pH 9. The highly purified protein is subsequently desalted into 5 storage buffer as described above for the poly-His tagged proteins. The homogeneity of the proteins is verified by SDS polyacrylamide gel (PEG) electrophoresis and N-terminal amino acid sequencing by Edman degradation. <br><br> Alternatively, a modified baculovirus procedure may be used incorporating high-5 cells. In this procedure, the DNA encoding the desired sequence is amplified with suitable systems, such as Pfu (Stratagene), or fused upstream (5'-of) of an epitope tag contained with a baculovirus expression vector. Such epitope tags include 10 poly-His tags and immunoglobulin tags (like Fc regions of IgG). A variety of plasmids may be employed, including plasmids derived from commercially available plasmids such as pIE I -1 (Novagen). The pIE I -1 and pIE 1-2 vectors are designed for constitutive expression of recombinant proteins from the baculovirus iel promoter in stably-transformed insect cells. The plasmids differ only in the orientation of the multiple cloning sites and contain I all promoter sequences known to be important for ie 1 -mediated gene expression in uninfected insect cells as well 15 as the hr5 enhancer element. pIEI-l and pi El-2 include the translation initiation site and can be used to produce fusion proteins. Briefly, the desired sequence or the desired portion of the sequence (such as the sequence encoding the extracellular domain of a transmembrane protein) is amplified by PCR with primers complementary to the 5' and 3' regions. The 5' primer may incorporate flanking (selected) restriction enzyme sites. The product is then digested with those selected restriction enzymes and subcloned into the expression vector. For example, derivatives 20 of plEl -1 can include the Fc region of human IgG (pb.PH.IgG) or an 8 histidine (pb.PH.His) tag downstream (3'-of the desired sequence). Preferably, the vector construct is sequenced for confirmation. <br><br> High-5 cells are grown to a confluency of 50% under the conditions of, 27 °C, no C02, NO pen/strep. For each 150 mm plate, 30 fx% of pIE based vector containing the sequence is mixed with 1 ml Ex-Cell medium [Media: Ex-Cell 401 + 1/100 L-Glu JRH Biosciences #14401-78P (note: this media is light sensitive)], and in a separate 25 tube, 100 fu\ ofCellFectin [CellFECTIN (GibcoBRL #10362-010) (vortexed to mix)] ismixedwith 1 mlofEx-Cell medium. The two solutions are combined and allowed to incubate at room temperature for 15 minutes. 8 ml of Ex-Cell media is added to the 2 ml of DNA/CellFECTIN mix and this is layered on high-5 cells that have been washed once with Ex-Cell media. The plate is then incubated in darkness for 1 hour at room temperature. The DNA/CellFECTIN mix is then aspirated, and the cells are washed once with Ex-Cell to remove excess 30 CellFECTIN, 30 ml of fresh Ex-Cell media is added and the cells are incubated for 3 days at28°C. The supernatant is harvested and the expression of the sequence in the baculovirus expression vector is determined by batch binding of 1 ml of supernatant to 25 ml ofNi :"-NT A beads (Q1AGEN) for histidine tagged proteins or Protein-A Sepharose CL-4B beads (Pharmacia) for IgG tagged proteins followed by SDS-PAGE analysis comparing to a known concentration of protein standard by Coomassie blue staining. <br><br> 35 The conditioned media from the transfected cells (0.5 to 3 L) is harvested by centrifugation to remove the cells and filtered through 0.22 micron filters. For the poly-His tagged constructs, the protein comprising the sequence is purified using a Ni 2~-NTA column (Qiagen). Before purification, imidazole is added to the conditioned media <br><br> 196 <br><br> to a concentration of 5 mM. The conditioned media is pumped onto a 6 ml Ni 2+-NTA column equilibrated in 20 mM Hepes, pH 7.4, buffer containing 0.3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml/min. at 48°C. After loading, the column is washed with additional equilibration buffer and the protein eluted with equilibration buffer containing 0.25 M imidazole. The highly purified protein is then subsequently desalted into a storage buffer 5 containing 10 mM Hepes, 0.14 M NaCl and 4% mannitol, pH 6.8, with a 25 ml G25 Superfine (Pharmacia) column and stored at -80°C. <br><br> Immunoadhesin (Fc containing) constructs of proteins are purified from the conditioned media as follows. The conditioned media is pumped onto a 5 ml Protein A column (Pharmacia) which has been equilibrated in 20 mM Na phosphate buffer, pH 6.8. After loading, the column is washed extensively with equilibration buffer before 10 elution with 100 mM citric acid, pH 3.5. The eluted protein is immediately neutralized by collecting I ml fractions into tubes containing 275 ml of 1 M Tris buffer, pH 9. The highly purified protein is subsequently desalted into storage buffer as described above for the poly-His tagged proteins. The homogeneity of the sequence is assessed by SDS polyacrylamide gels and by N-terminal amino acid sequencing by Edman degradation and other analytical 0) procedures as desired or necessary. <br><br> 15 PROl72, PRO 178, PR0179, PR0182, PR0187, PROI88, PR0212, PR0216, PR0217, PR0224,PR0231, <br><br> PR0235, PR0269, PR0287, PR0301, PR0323, PR0331, PR0356, PR0364, PR0526, PR0538, PR0713, PR0771, PR0788, PR0792, PR0812, PR0865, PRO 1075, PRO 1154, PRO 1244, PRO 1286, PRO 1304, PRO 1312, PR01376, PROI387, and PR01561 were successfully expressed by the above modified baculovirus procedure incorporating high-5 cells. <br><br> 20 EXAMPLE 68 <br><br> Preparation of Antibodies that Bind PRO Polypeptides <br><br> This Example illustrates preparation of monoclonal antibodies that can specifically bind PRO polypeptides. <br><br> Techniques for producing the monoclonal antibodies are known in the art and are described, for instance, in Goding, supra. Immunogens that may be employed include purified PRO fusion proteins containing PRO 25 polypeptides, and cells expressing the gene encoding the PRO polypeptide on the cell surface. Selection of the immunogen can be made by the skilled artisan without undue experimentation. <br><br> Mice, such as Balb/c, are immunized with the PRO polypeptide immunogen emulsified in complete Freund's adjuvant and injected subcutaneously or intraperitoneally in an amount from 1 to 100 micrograms. Alternatively, the immunogen is emulsified in MPL-TDM adjuvant (Ribi Immunochemical Research, Hamilton, MT)and injected 30 into the animal's hind foot pads. The immunized mice are then boosted 10 to 12 days later with additional immunogen emulsified in the selected adjuvant. Thereafter, for several weeks, the mice may also be boosted With additional immunization injections. Serum samples may be periodically obtained from the mice by retro-orbital bleeding for testing in ELISA assays to detect anti-PRO antibodies. <br><br> After a suitable antibody titer has been detected, the animals "positive" for antibodies can be injected with a 35 final intravenous injection of the PRO polypeptide. Three to four days later, the mice are sacrificed and the spleen cells are harvested. The spleen cells are then fused (using 35% polyethylene glycol) to a selected murine myeloma <br><br> 197 <br><br> 10 <br><br> cell line such as P3X63AgU. I, available from ATCC, No. CRL 1597. The fusions generate hybridoma cells that can then be plated in 96-well tissue culture plates containing HAT medium to inhibit proliferation of non-fused cells, myeloma hybrids, and spleen cell hybrids. <br><br> The hybridoma cells will be screened in an ELISA for reactivity against the PRO polypeptide. Determination of "positive" hybridoma cells secreting the desired monoclonal antibodies against the PRO polypeptide is within the skill in the art. <br><br> The positive hybridoma cells can be injected intraperitoneally into syngeneic Balb/c mice to produce ascites containing the anti-PRO monoclonal antibodies. Alternatively, the hybridoma cells can be grown in tissue-culture flasks or roller bottles. Purification of the monoclonal antibodies produced in the ascites can be accomplished using ammonium-sulfate precipitation, followed by gel-exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can be employed. <br><br> Deposit of Material <br><br> The following material(s) has-'have been deposited with the American Type Culture Collection, 10801 University Blvd., Manassas, VA 20110-2209, USA (ATCC): <br><br> 15 <br><br> Material <br><br> ATCC Deo. No. <br><br> Deposit Date <br><br> DNA35916-1161 <br><br> 209419 <br><br> October 28, 1997 <br><br> DNA23339-1130 <br><br> 209282 <br><br> September 18,1997 <br><br> DNA1645I-I388 <br><br> 209776 <br><br> April 14, 1998 <br><br> DNA27865-1091 <br><br> 209296 <br><br> September 23, 1997 <br><br> 20 <br><br> DNA27864-1155 <br><br> 209375 <br><br> October 16, 1997 <br><br> DNA28497-1130 <br><br> 209279 <br><br> September 18, 1997 <br><br> DN A26847-1395 <br><br> 209772 <br><br> April 14, 1998 <br><br> DNA30942-1134 <br><br> 209254 <br><br> September 16, 1997 <br><br> DM A32286-1191 <br><br> 209385 <br><br> October 16, 1997 <br><br> 25 <br><br> DNA33094-1131 <br><br> 209256 <br><br> September 16, 1997 <br><br> DNA33221-1133 <br><br> 209263 <br><br> September 16, 1997 <br><br> DNA34434-1139 <br><br> 209252 <br><br> September 16, 1998 <br><br> DNA35558-1167 <br><br> 209374 <br><br> October 16, 1997 <br><br> DNA35638-1141 <br><br> 209265 <br><br> September 16, 1997 <br><br> 30 <br><br> DNA33473-1176 <br><br> 209391 <br><br> October 17, 1997 <br><br> DNA3 8260-1180 <br><br> 209397 <br><br> October 17, 1997 <br><br> DNA39969-1185 <br><br> 209400 <br><br> October 17, 1997 <br><br> DNA40628-1216 <br><br> 209432 <br><br> November 7, 1997 <br><br> DNA35595-I228 <br><br> 209528 <br><br> December 10, 1997 <br><br> 35 <br><br> DNA4098M234 <br><br> 209439 <br><br> November 7, 1997 <br><br> DNA47470-1130-P1 <br><br> 209422 <br><br> October 28, 1997 <br><br> 198 <br><br> DNA47365-1206 <br><br> 209436 <br><br> November 7,1997 <br><br> DNA44184-1319 <br><br> 209704 <br><br> March 26,1998 <br><br> DNA48613-1268 <br><br> 209752 <br><br> April 7,1998 <br><br> DNA29101-1122 <br><br> 209653 <br><br> March 5,1998 <br><br> DNA49646-1327 <br><br> 209705 <br><br> March 26,1998 <br><br> DNA49829-1346 <br><br> 209749 <br><br> April 7,1998 <br><br> DNA56405-1357 <br><br> 209849 <br><br> May 6,1998 <br><br> DNA56352-1358 <br><br> 209846 <br><br> May 6,1998 <br><br> DNA59205-1421 <br><br> 203009 <br><br> June 23,1998 <br><br> DNA53974-1401 <br><br> 209774 <br><br> April 14,1998 <br><br> DNA57689-1385 <br><br> 209869 <br><br> May 14,1998 <br><br> DNA60615-1483 <br><br> 209980 <br><br> June 16,1998 <br><br> DNA59814-1486 <br><br> 203359 <br><br> October 20,1998 <br><br> DNA59846-1503 <br><br> 209978 <br><br> June 16,1998 <br><br> DNA64883-1526 <br><br> 203253 <br><br> September 9,1998 <br><br> DNA64885-1529 <br><br> 203457 <br><br> November 3,1998 <br><br> DNA64889-1541 <br><br> 203250 <br><br> September 9,1998 <br><br> DNA64903-1553 <br><br> 203223 <br><br> September 15, 1998 <br><br> DNA64905-1558 <br><br> 203233 <br><br> September 15, 1998 <br><br> DNA65409-1566 <br><br> 203232 <br><br> September 15,1998 <br><br> DNA65406-1567 <br><br> 203219 <br><br> September 15,1998 <br><br> DNA61873-1574 <br><br> 203132 <br><br> August 18,1998 <br><br> DNA64966-1575 <br><br> 203575 <br><br> January 12,1999 <br><br> DNA67300-1605 <br><br> 203163 <br><br> August 25,1998 <br><br> DNA68872-1620 <br><br> 203160 <br><br> August 25,1998 <br><br> DNA76538-1670 <br><br> 203313 <br><br> October 6,1998 <br><br> DNA33087 <br><br> 203381 <br><br> October 16,1997 <br><br> This deposit was made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty). 30 This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. Hie deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Genentech, Inc., and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by 35 the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 USC §122 and the Commissioner's rules pursuant thereto (including 37 CFR §1.14 with particular reference to 886 OG 638). <br><br> 199 <br><br> The assignee of the present application has agreed that if a culture of the material(s) on deposit should die or be lost or destroyed when cultivated under suitable conditions, the material(s) will be promptly replaced on notification with another ofthe same. Availability of the deposited material(s) is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws. <br><br> The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by the construct(s) deposited, since the deposited embodiment(s) is/are intended as single illustration(s) of certain aspects of the invention and any constructs that are functionally equivalent are within the scope of this invention. The deposit of material(s) herein does not constitute an admission that the written description herein contained is inadequate to enable the practice of any aspect of the invention, including the best mode thereof, nor is it to be construed as limiting the scope of the claims to the specific illustrations that it represents. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. <br><br> 200 <br><br></p> </div>

Claims (96)

<div class="application article clearfix printTableText" id="claims"> <p lang="en"> WHAT WE CLAIM IS:<br><br>
1. An isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence that encodes a polypeptide comprising an amino acid sequence shown in Figure 88 (SEQ ID NO:216).<br><br>
2. An isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence shown in Figure 87 (SEQ ID NO:215).<br><br>
3. An isolated nucleic acid having at least 80% nucleic acid sequence identity to a nucleotide sequence comprising the full length coding sequence of the nucleotide sequence shown in Figure 87 (SEQ ID NO:215).<br><br>
4. An isolated nucleic acid having at least 80% nucleic acid sequence identity to the full-length coding sequence of the DNA deposited under ATCC accession number 203575.<br><br>
5. An isolated nucleic acid having a nucleotide sequence that encodes a polypeptide comprising the amino acid sequence shown in Figure 88 (SEQ ID NO: 216).<br><br>
6. The nucleic acid of claim 5 wherein said nucleotide sequence comprises the nucleotide sequence shown in figure 87 (SEQ ID NO: 215).<br><br>
7. The nucleic acid of claim 5 or claim 6 wherein said nucleotide sequence comprises the full length coding sequence from within the sequence shown in Figure 87 (SEQ ID NO: 215).<br><br>
8. The nucleic acid of claim 5 which comprises the full length coding sequence of the DNA deposited under accession number ATCC 203575.<br><br>
9. An isolated nucleic acid molecule consisting of an at least 20 nucleotides fragment of the nucleic acid sequence of SEQ ID NO:215, or a complement thereof, that hybridizes under stringent conditions to:<br><br> (a) the nucleic acid sequence of SEQ ID NO:215 or a complement thereof;<br><br> (b) the full length coding sequence of the cDNA deposited under ATCC<br><br> accession number 203575 or a complement thereof; and<br><br> rjpjTELLECTUAL-PROPERT* OFFICE Or Ni<br><br> 1 5 JI$ 2008<br><br> (c) wherein, said stringent conditions use 50% formamide, 5 x SSC, 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5x Denhardt's solution, sonicated salmon sperm DNA (50 |ig/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC and 50% formamide at 55°C, followed by a wash comprising of 0.1 x SSC containing<br><br>
10. The isolated nucleic acid molecule of Claim 9 that is at least 50 nucleotides.<br><br>
11. The isolated nucleic acid molecule of Claim 9 that is at least 60 nucleotides.<br><br>
12. The isolated nucleic acid molecule of Claim 9 that is at least 70 nucleotides.<br><br>
13. The isolated nucleic acid molecule of Claim 9 that is at least 80 nucleotides.<br><br>
14. The isolated nucleic acid molecule of Claim 9 that is at least 90 nucleotides.<br><br>
15. The isolated nucleic acid molecule of Claim 9 that is at least 100 nucleotides.<br><br>
16. Isolated nucleic acid having at least 80% nucleic acid sequence identity to:<br><br> (a) a nucleotide sequence encoding the polypeptide shown in Figure 88 (SEQ ID NO:216) lacking its associated signal peptide;<br><br> (b) a nucleotide sequence encoding an extraceullalar domain of the polypeptide shown in Figure 88 (SEQ ID NO:216), with its associated signal peptide; or<br><br> (c) a nucleotide sequence encoding an extraceullular domain of the polypeptide shown in Figure 88 (SEQ ID NO:216) lacking its associated signal peptide.<br><br>
17. A vector comprising the nucleic acid of any one of claims 1 to 16.<br><br>
18. The vector of claim 17 operably linked to control sequence recognized by a host cell transformed with the vector.<br><br>
19. An isolated host cell comprising the vector of claim 18.<br><br> EDTA at 55°C.<br><br> 553996 I.DOC<br><br>
20. The host cell of claim 19, wherein said cell is a CHO cell.<br><br> 202<br><br>
21. The host cell of claim 20, wherein said cell is an E. coli.<br><br>
22. The host cell of claim 21, wherein said cell is a yeast cell.<br><br>
23. The host cell of claim 21, wherein said cell is a Baculovirus-infected insect cell.<br><br>
24. An isolated polypeptide having at least 80% amino acid sequence identity to an amino acid sequence shown in Figure 88 (SEQ ID NO:216).<br><br>
25. An isolated polypeptide scoring at least 80% positives when compared to an amino acid sequence consisting of the amino acid sequence shown in Figure 88 (SEQ ID NO:216).<br><br>
26. An isolated polypeptide having at least 80% amino acid sequence identity to an amino acid sequence encoded by the full-length coding sequence of the DNA deposited under ATCC accession number 203575.<br><br>
27. An isolated polypeptide comprising a sequence having the amino acid sequence shown in figure 88 (SEQ ID NO: 216).<br><br>
28. An isolated polypeptide comprising a sequence having the amino acid sequence encoded by the nucleotide deposited under ATCC accession number 203575.<br><br>
29. An isolated polypeptide having at least 80% amino acid sequence identity to:<br><br> (a) the polypeptide shown in<br><br> Figure 88 (SEQ ID NO:216) lacking its associated signal peptide;<br><br> (b) an extracellular domain of the polypeptide shown in Figure 88 (SEQ ID NO:216) with its associated signal peptide; or<br><br> (c) an extraceullular domain of the polypeptide shown in Figure 88 (SEQ ID NO:216) lacking its associated signal peptide.<br><br>
30. A process for producing a polypeptide as claimed in any one of claims 24 to 29 comprising culturing the host cell of any one of claims 19 to 23 under conditions suitable for expression of said polypeptide and recovering said polypeptide from the cell culture.<br><br> 553996 I. DOC 203<br><br> intellectual PROPERTY office<br><br> Or N.2<br><br> 1 5 JUM<br><br> s ¥ el y' i<br><br>
31.<br><br> A chimeric molecule comprising a polypeptide according to any one of claims 24 to 29 fused to a heterologous amino acid sequence.<br><br>
32. The chimeric molecule of claim 31, wherein said heterologous amino acid sequence is an epitope tag sequence.<br><br>
33. The chimeric molecule of claim 32, wherein said heterologous amino acid sequence is a Fc region of an immunoglobulin.<br><br>
34. An antibody which specifically binds to a polypeptide according to any one of claims 24 to<br><br>
35. The antibody of claim 34, wherein said antibody is a monoclonal antibody, a humanized antibody or a single-chain antibody.<br><br>
36. A polypeptide according to any one of claims 24 to 29 for use in a method of medical treatment.<br><br>
37. A pharmaceutical composition comprising a polypeptide of any one of claims 24 to 29 and a pharmaceuticaliy acceptable carrier, excipient or stabiliser.<br><br>
38. A composition comprising an antibody according to claim 34 or claim 35 in admixture with a pharmaceuticaliy acceptable carrier.<br><br>
39. The composition of claim 37 further comprising a cardiovascular, endothelial, angiogenic or angiostatic agent.<br><br>
40. A method of preparing the composition of claim 37 comprising admixing said polypeptide or antibody with a pharmaceuticaliy acceptable carrier, excipient or stabiliser.<br><br>
41. A method of preparing the composition of claim 38 comprising admixing said antibody with a pharmaceuticaliy acceptable carrier, excipient or stabiliser.<br><br> 29.<br><br> INTELLECTUAL KMO"<br><br> iCTUAL PROWr OmCt Or ivz'<br><br> 1 5 JUN ®<br><br>
42. An article of manufacture comprising:<br><br> (1) a polypeptide having at least 80% sequence identity to an amino acid sequence shown in Figure 88 (SEQ ID NO: 216), or an antibody which specifically binds to said polypeptide in admixture with a pharmaceuticaliy acceptable carrier;<br><br> (2) a container containing said composition; and<br><br> (3) a label affixed to said container, or a package insert included in said container referring to the use of said composition in the treatment of a cardiovascular, endothelial, and angiogenic disorder.<br><br>
43. The article of manufacture of claim 42, wherein said composition comprises a therapeutically effective amount of said polypeptide or antibody in admixture with said pharmaceuticaliy acceptable carrier.<br><br>
44. A method for identifying an agonist of a polypeptide as defined in claim 24 comprising:<br><br> (a) contacting cells ex vivo and a test compound to be screened under conditions suitable for the induction of a cellular response normally induced by said polypeptide; and<br><br> (b) determining the induction of said cellular response to determine if the test compound is an effective agonist, wherein the induction of said cellular response is indicative of said test compound being an effective agonist.<br><br>
45. The method of claim 44, wherein the cellular response normally induced by said polypeptide is stimulation of cell proliferation.<br><br>
46. A method for identifying a compound that inhibits an activity of a polypeptide as defined in claim 24 comprising contacting a test compound with said polypeptide ex vivo under conditions and for a time sufficient to allow the test compound and polypeptide to interact and determining whether the activity of said polypeptide is inhibited.<br><br>
47. A method for identifying a compound that inhibits an activity of a polypeptide as defined in claim 24 comprising the steps of:<br><br> (a) contacting cells ex vivo and a test compound to be screened in the presence of said polypeptide under conditions suitable for the induction of a cellular response normally induced by said polypeptide; and<br><br> (b) determining the induction of said cellular response to determine i|<br><br> is an effective antagonist.<br><br> 15 JUN 2006<br><br> 553996 I.DOC<br><br> 205<br><br> &lt; office<br><br> * p r r<br><br>
48.<br><br> The method of claim 47, wherein the cellular response normally induced by said polypeptide is stimulation of cell proliferation.<br><br>
49. A method for identifying a compound that inhibits the expression of a polypeptide as defined in claim 24 in cells that normally expresses the polypeptide, wherein the method comprises contacting the cells ex vivo with a test compound under conditions suitable for allowing expression of said polypeptide and determining whether the expression of said polypeptide is inhibited.<br><br>
50. An oligonucleotide anti-sense to a nucleic acid molecule encoding a polypeptide as defined in claim 24, which inhibits the expression of said polypeptide in a mammalian cell which expresses said polypeptide.<br><br>
51. A method for diagnosing a disease or susceptibility to a disease which is related to a mutation in a nucleic acid sequence encoding a polypeptide as defined in claim 24, comprising determining the presence or absence of said mutation in said polypeptide-encoding nucleic acid sequence, wherein the presence or absence of said mutation is indicative of the presence of said disease or susceptibility to said disease.<br><br>
52. A method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal which comprises analyzing the level of expression of a gene encoding a polypeptide as defined in claim 24 (a) in a test sample of tissue cells obtained from said mammal, and (b) in a control sample of known normal tissue cells of the same cell type, wherein a higher or lower expression level in the test sample as compared to the control sample is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in said mammal.<br><br>
53. A method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal which comprises detecting the presence or absence of a polypeptide as defined in claim 24 in a test sample of tissue cells obtained from said mammal, wherein the presence or absence of said polypeptide in said test sample is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in said mammal.<br><br>
54. A method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal comprising (a) contacting an antibody to the polypeptide as defined ir<br><br> 553996 I. DOC<br><br> 206<br><br> Or h.l<br><br> 1 5 JUil 2006 RECEIVED<br><br> sample of tissue cells obtained from the mammal, and (b) detecting the formulation of a complex between said antibody and said polypeptide in the test sample, wherein the formulation of said complex is indicative of the presence of a cardiovascular, endothelial or angiogenic disorder in the mammal.<br><br>
55. A method for determining the presence of a polypeptide as defined in claim 24 in a sample comprising contacting a sample suspected of containing said polypeptide with an antibody thereto and determining binding of said antibody to a compound of said sample.<br><br>
56. A cardiovascular, endothelial or angiogenic disorder diagnostic kit comprising an antibody to the polypeptide as defined in claim 24 and a carrier in suitable packaging.<br><br>
57. A use of a polypeptide as defined in claim 24 or an antibody which specifically binds to said polypeptide, in the manufacture of a medicament for treating a cardiovascular, endothelial or angiogenic disorder in a mammal.<br><br>
58. The use according to claim 57, wherein the mammal is human.<br><br>
59. The use of claim 58, wherein the human has suffered myocardial infarction.<br><br>
60. The use of claim 58, wherein the human has cardiac hypertrophy, trauma, a cancer, or age-related macular degeneration.<br><br>
61. The use of claim 60, wherein the cardiac hypertrophy is characterized by the presence of an elevated level of PGFiu.<br><br>
62. The use of claim 57, wherein the medicament further comprises a cardiovascular, endothelial or angiogenic agent.<br><br>
63. A use of a polypeptide as defined in claim 24 or an antibody which specifically binds to said polypeptide; and a cardiovascular, endothelial or angiogenic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal.<br><br> 5539% I. DOC<br><br> 207<br><br> i?iil£CTHwOPE^^<br><br> OF Nl<br><br> 1 5 JUN 2006 ncr*ci\/pn<br><br>
64.<br><br> The use of any one of claims 57 to 59, wherein the mammal has undergone primary angioplasty.<br><br>
65. The use of claim 57, wherein the cardiovascular, endothelial or angiogenic disorder is cancer.<br><br>
66. The use of claim 65, wherein the medicament further comprises a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent.<br><br>
67. A use of a polypeptide as defined in claim 24 or an antibody which specifically binds to said polypeptide, and a chemotherapeutic agent, a growth inhibitory agent or a cytotoxic agent in the manufacture of a medicament for simultaneous, separate or sequential use for treating a cardiovascular, endothelial or angiogenic disorder in a mammal.<br><br>
68. A use of a nucleic acid molecule that encodes a polypeptide as defined in claim 24 or an antibody thereto in the manufacture of a medicament for treating a cardiovascular, endothelial or angiogenic disorder in a mammal.<br><br>
69. A use of claim 68, wherein the mammal is human.<br><br>
70. The use of claim 68, wherein the nucleic acid molecule is formulated for administration via ex vivo gene therapy.<br><br>
71. A recombinant retroviral particle comprising a retroviral vector consisting essentially of (1) a promoter, (2) nucleic acid encoding a polypeptide as defined in claim 24, an antibody thereto, and (3) a signal sequence for cellular secretion of the polypeptide, wherein the retroviral vector is in association with retroviral structure proteins.<br><br>
72. An ex vivo producer cell comprising a nucleic acid construct that expresses retroviral structural proteins and also comprises a retroviral vector consisting essentially of (1) a promoter, (2) nucleic acid encoding a polypeptide as defined in claim 24, or an antibody thereto, and (3) a signal sequence for cellular secretion of the polypeptide, wherein said producer cell packages the retroviral vector in association with the structural proteins to produce recombinant retroviral particles.<br><br> 553996 I.DOC<br><br> 208<br><br>
73. A use of a polypeptide as defined in claim 24 or an antibody thereto, in the manufacture of a medicament for inhibiting endothelial cell growth in a mammal.<br><br>
74 A use of an antibody which specifically binds a polypeptide as defined in claim 24, in the manufacture of a medicament for stimulating endothelial cell growth in a mammal.<br><br>
75. Isolated nucleic acid as claimed in any one of claims 1 to 16 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
76. A vector as defined in claim 17 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
77. An isolated host cell as defined in claim 19 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
78. An isolated polypeptide as claimed in any one of claims 24 to 29 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
79. A process as claimed in claim 30 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
80. A chimeric molecule as defined in claim 31 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
81. An antibody as defined in claim 34 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
82. A pharmaceutical composition in claim 37 or claim 38 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
83. A method as claimed in claim 40 substantially as herein described with reference to any<br><br> 553996 I. DOC<br><br> example thereof and with or without reference to the accompanying figiuj^j.^ | pr.ti jarp^op^rtv office or n.z.<br><br> 15 JUN 2006 QPrtFIVFR<br><br> 209<br><br>
84. A method as claimed in claim 41 substantially as herein described with reference to any example thereof and with or without reference to the accompanying figures.<br><br>
85. An article of manufacture as defined in claim 42 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
86. A method as defined in claim 44 for identifying an agonist substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
87. A method as claimed in any one of claims 46 to 48 for identifying a compound that inhibits an activity of a polypeptide as defined in claim 24 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
88. A method as claimed in claim 49 for identifying a compound that inhibits the expression of a polypeptide as defined in claim 1 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
89. An oligonucleotide as claimed in claim 50 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
90. A method for diagnosing a disease or susceptibility to a disease as defined in claim 51 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
91. A method of diagnosing a cardiovascular, endothelial or angiogenic disorder in a mammal as claimed in any one of claims 52 to 54 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
92. A method as claimed in claim 55 for determining the present of a polypeptide as defined in claim 1 substantially as herein described with reference to any example thereof and wither<br><br> without reference to the accompanying drawings. INTELLECTUAi^U ER&gt; &gt; 0.<br><br> • JM<br><br> 553996 I.DOC<br><br> 210<br><br> 15 m r-<br><br> sr p r'<br><br>
93. A cardiovascular, endothelial or angiogenic disorder kit as claimed in claim 56 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
94. A use as claimed in any one of claims 57 to 70, 73 or 74 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
95. A recombinant retroviral particle as claimed in claim 71 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br>
96. An ex vivo producer cell as defined in claim 72 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.<br><br> 553996 I.DOC<br><br> 211<br><br> Intellectual property office of n.z.<br><br> \ 5 JUH 2005 I nrPClV/PH<br><br> </p> </div>
NZ54075499A 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization NZ540754A (en)

Applications Claiming Priority (18)

Application Number Priority Date Filing Date Title
US82510898A 1998-12-01 1998-12-01
US11285098P 1998-12-16 1998-12-16
US11555499P 1999-01-12 1999-01-12
US90502899A 1999-03-08 1999-03-08
US12395799P 1999-03-12 1999-03-12
US13144599P 1999-04-28 1999-04-28
US13428799P 1999-05-14 1999-05-14
US91225299A 1999-06-02 1999-06-02
US14103799P 1999-06-23 1999-06-23
US14475899P 1999-07-20 1999-07-20
US14569899P 1999-07-26 1999-07-26
US92011199A 1999-09-01 1999-09-01
US92059499A 1999-09-08 1999-09-08
US92094499A 1999-09-13 1999-09-13
US92154799A 1999-09-15 1999-09-15
US92109099A 1999-09-15 1999-09-15
US92308999A 1999-10-05 1999-10-05
US16250699P 1999-10-29 1999-10-29

Publications (1)

Publication Number Publication Date
NZ540754A true NZ540754A (en) 2006-11-30

Family

ID=31999971

Family Applications (1)

Application Number Title Priority Date Filing Date
NZ54075499A NZ540754A (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization

Country Status (1)

Country Link
NZ (1) NZ540754A (en)

Similar Documents

Publication Publication Date Title
AU771751B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
WO2000053753A2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
AU768694B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
JP2003531811A5 (en)
WO2000053752A2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
US6800604B2 (en) Polypeptides that inhibit human serum-induced cleavage of hepatocyte growth factor
EP1212417B1 (en) Promotion or inhibition of angiogenesis and cardiovascularization
WO2001040464A1 (en) Interleukin-1-receptor associated kinase-3 (irak3) and its use in promotion or inhibition of angiogenesis and cardiovascularization
WO2000015792A2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
AU2003259607B2 (en) Promotion or inhibition of angiogenesis and cardiovascularization
EP1734051A2 (en) Composition and methods for the diagnosis of tumours
NZ540754A (en) Promotion or inhibition of angiogenesis and cardiovascularization
NZ545534A (en) Promotion or inhibition of angiogenesis and cardiovascularization
WO2001019987A1 (en) Promotion or inhibition of angiogenesis and cardiovascularization
NZ535590A (en) Promotion or inhibition of angiogenesis and cardiovascularization
ZA200103707B (en) Promotion or inhibition of angiogenesis and cardio-vascularization.
NZ532803A (en) Promotion or inhibition of angiogenesis and cardiovascularization
ZA200105990B (en) Promotion or inhibition of angiogenesis and cardiovascularization.

Legal Events

Date Code Title Description
PSEA Patent sealed
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
EXPY Patent expired