NZ531180A - Compositions and methods for effecting the levels of cholesterol using the LIPG polypeptide - Google Patents

Compositions and methods for effecting the levels of cholesterol using the LIPG polypeptide

Info

Publication number
NZ531180A
NZ531180A NZ531180A NZ53118000A NZ531180A NZ 531180 A NZ531180 A NZ 531180A NZ 531180 A NZ531180 A NZ 531180A NZ 53118000 A NZ53118000 A NZ 53118000A NZ 531180 A NZ531180 A NZ 531180A
Authority
NZ
New Zealand
Prior art keywords
lipg
leu
gly
ser
val
Prior art date
Application number
NZ531180A
Inventor
Michael Jaye
Original Assignee
Aventis Pharma 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
Priority claimed from US09/277,401 external-priority patent/US7008776B1/en
Application filed by Aventis Pharma Inc filed Critical Aventis Pharma Inc
Publication of NZ531180A publication Critical patent/NZ531180A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/18Carboxylic ester hydrolases (3.1.1)
    • C12N9/20Triglyceride splitting, e.g. by means of lipase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biophysics (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Microbiology (AREA)
  • Vascular Medicine (AREA)
  • Obesity (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

A composition capable of lowering the expression of LIPG in a patient comprising a ribozyme which cleaves mRNA which encodes LIPG polypeptide. (62) Divided out of 514350

Description

<div class="application article clearfix" id="description"> <p class="printTableText" lang="en">53 11 8 0 <br><br> NEW ZEALAND PATENTS ACT, 1953 <br><br> INTELLECTUAL PROPERTY <br><br> OFFICE OF N.z <br><br> 17 FEB 200^ RECEIVED <br><br> intellectual property office of n.Z. <br><br> , 16 FEB 2004 <br><br> {(A <br><br> RECEIVED <br><br> No: Date: <br><br> Divided out of No. 514350 Dated 24 March 2000 <br><br> COMPLETE SPECIFICATION <br><br> COMPOSITIONS AND METHODS FOR EFFECTING THE LEVELS OF CHOLESTEROL <br><br> We, AVENTIS PHARMACEUTICALS INC., of Route 202-206, P O Box 6800, Mail Stop EMCG1, Bridgewater, New Jersey 08807-0800, United States of America (formerly of 500 Areola Road, Collegeville, Pennsylvania 19426, United States of America); and THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA, of University of Pennsylvania, Suite 300, Office of Research Administration, 133 South 36th Street, Philadelphia, Pennsylvania 19104-3246, United States of America, 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 page la) <br><br> INTELLECTUAL PROPERTY <br><br> .... OFFICE OF N.Z <br><br> 17 FEB 2004 ■ ' RECEIVED <br><br> o <br><br> - la- <br><br> COMPOSITIONS AND METHODS FOR EFFECTING THE LEVELS OF HIGH DENSITY LIPOPROTEIN (HDL) CHOLESTEROL AND APOLIPOPROTEIN AI, VERY LOW DENSITY LIPOPROTEIN (VLDL) CHOLESTEROL AND LOW DENSITY LIPOPROTEIN (LDL) CHOLESTEROL <br><br> This is a divisional of NZ 514350. <br><br> FIELD OF THE INVENTION <br><br> This invention relates to methods and compositions for 15 increasing the level of high density lipoprotein (HDL) <br><br> cholesterol and apolipoprotein AI in a patient and to methods and compositions for lowering the levels of very low density lipoprotein (VLDL) cholesterol, and low density lipoprotein (LDL) cholesterol in a patient. The invention includes 20 within its scope methods and compositions which lower the expression of, or inhibit the activity of, a gene, LIPG, <br><br> which encodes a lipase enzyme that lowers the levels of HDL cholesterol and apolipoprotein AI. The invention additionally includes within its scope methods and 25 compositions to increase the expression of, or enhance the activity of, the lipase enzyme, resulting in lower levels of VLDL and LDL cholesterol. <br><br> - 2 - <br><br> Lipids <br><br> Lipids are water-insoluble organic biomolecules, which are essential components of diverse biological functions, 5 including the storage, transport, and metabolism of energy, and membrane structure and fluidity. Lipids are derived from two sources in man and other animals: some lipids are ingested as dietary fats and oils and other lipids are biosynthesized by the human or animal. In mammals, at least 10 10% of the body weight is lipid, the bulk of which is in the form of triacylglycerols. <br><br> Triacylglycerols, also known as triglycerides and triacylglycerides, are made up of three fatty acids esterified to glycerol. Dietary triacylglycerols are stored 15 in adipose tissues as a source of energy, or hydrolyzed in the digestive tract by triacylglycerol lipases, the most important of which is pancreatic lipase. Triacylglycerols are transported between tissues in the form of lipoproteins. <br><br> Lipoproteins are micelle-like assemblies found in plasma 20 which contain varying proportions of different types of lipids and proteins (called apoproteins). There are five main classes of plasma lipoproteins, the major function of which is lipid transport. These classes are, in order of increasing density: chylomicrons; very low density 25 lipoproteins (VLDL); intermediate-density lipoproteins (IDL); low density lipoproteins (LDL); and high density lipoproteins (HDL). Although many types of lipid are found associated with each lipoprotein class, each class transports predominantly one type of lipid: triacylglycerols described 30 above are transported in chylomicrons, VLDL, and IDL; whereas phospholipids and cholesterol esters are transported in HDL and LDL respectively. <br><br> Phospholipids are di-fatty acid esters of glycerol phosphate which contain a polar group coupled to the 35 phosphate. Phospholipids are important structural components of cellular membranes. Phospholipids are hydrolyzed by <br><br> i j <br><br> .r\ <br><br> l ) <br><br> - 3 - <br><br> enzymes called phospholipases. Phosphatidylcholine, an exemplary phospholipid, is a major component of most eukaryotic cell membranes. <br><br> Cholesterol is the metabolic precursor of steroid 5 hormones and bile acids as well as an essential constituent of cell membranes. In man and other animals, cholesterol is ingested in the diet and is synthesized also by the liver and other tissues. Dietary cholesterol is transported from the intestine to the liver by large lipoprotein molecules in the 10 blood. The liver secretes Very Low Density Lipoprotein <br><br> (VLDL) which transports cholesterol and cholesterol ester and various other compounds into the bloodstream. VLDL is partially converted in adipose tissue to Low Density Lipoprotein (LDL). LDL transports both free and esterified 15 cholesterol to body tissues. High Density Lipoprotein (HDL) transports cholesterol to the liver to be broken down and excreted. <br><br> Membranes surround every living cell and serve as a barrier between the intracellular and extracellular 20 compartments. Membranes also enclose the eukaryotic nucleus, make up the endoplasmic reticulum, and serve specialized functions such as in the myelin sheath that surrounds axons. A typical membrane contains about 40% lipid and 60% protein, but there is considerable variation. The major lipid 25 components are phospholipids, specifically phosphatidylcholine and phosphatidylethanplamine, and cholesterol. The physicochemical properties of membranes, such as fluidity, can be changed by modification of either the fatty acid profiles of the phospholipids or the 30 cholesterol content. Modulating the composition and organization of membrane lipids also modulates membrane-dependent cellular functions, such as receptor activity, endocytosis, and cholesterol flux. <br><br> Enzymes <br><br> 35 The triacylglycerol lipases are a family of enzymes which play several pivotal roles in the metabolism of lipids <br><br> 1 ) <br><br> V / <br><br> - 4 - <br><br> in the body. Three members of the human triacylglycerol lipase family have been described: pancreatic lipase, lipoprotein lipase, and hepatic lipase (Goldberg, I.J., Le, N.-A., Ginsberg, H.N., Krauss, R.M., and Lindgren, F.T. <br><br> 5 (1988) J". Clin. Invest. 81,561-568; Goldberg, I.J., Le, N., Paterniti J.R., Ginsberg, H.N., Lindgren, F.T., and Brown, W.V. (1982) J. Clin. Invest. 70,1184-1192; Hide, W.A., Chan, L., and Li, W.-H. (1992) J. Lipid. Res. 33,167-178). Pancreatic lipase is primarily responsible for the hydrolysis 10 of dietary lipids. Variants of pancreatic lipase have been described, but their physiological role has not been determined (Giller, T., Buchwald, P., Blum-Kaelin, D., and Hunziker, W. (1992) J. Biol. Chem. 267,16509-16516). Lipoprotein lipase is the major enzyme responsible for the 15 distribution and utilization of triglycerides in the body. Lipoprotein lipase hydrolyzes triglycerides in both chylomicrons and VLDL. Hepatic lipase hydrolyzes triglycerides in IDL and HDL and is responsible for lipoprotein remodeling. Hepatic lipase also functions as a 20 phospholipase and hydrolyzes phospholipids in HDL. <br><br> Phospholipases play important roles in the catabolism and remodeling of the phospholipid component of lipoproteins and the phospholipids of membranes. Phospholipases also play a role in the release of arachidonic acid and the subsequent 25 formation of prostaglandins, leukotrienes, and other lipids which are involved in a variety of inflammatory processes. <br><br> The aforementioned lipases are approximately 450 amino acids in length and have leader signal peptides to facilitate secretion. The lipases are comprised of two principal 30 domains (Winkler, K., D'Arcy, A., and Hunziker, W. (1990) <br><br> Nature 343, 771-774). The amino terminal domain contains the catalytic site while the carboxyl domain is believed to be responsible for substrate binding, cofactor association, and interaction with cell receptors (Wong, H., Davis, R.C., 35 Nikazy, J., Seebart, K.E., and Schotz, M.C. (1991) Proc. <br><br> Natl. Acad. Sci. USA 88,11290-11294; van Tilbeurgh, H., Roussel, A., Lalouel, J.-M., and Cambillau, C. (1994) J. <br><br> Biol. Chem. 269,4626-4633; Wong, H., Davis, R.C., Thuren, T., Goers, J.W., Nikazy, J., Waite, M., and Schotz, M.C. (1994) J. Biol. Chem. 269,10319-10323; Chappell, D.A., <br><br> Inoue, I., Fry, G.L., Pladet, M.W., Bowen, S.L., Iverius, P.-5 H., Lalouel, J.-M., and Strickland, D.K. (1994) J. Biol. Chem. 269, 18001-18006). The overall level of amino acid homology between members of the family is 22-65%, with local regions of high homology corresponding to structural homologies which are linked to enzymatic function. <br><br> 10 The naturally occurring lipoprotein lipase is glycosylated. Glycosylation is necessary for LPL enzymatic activity (Semenkovich, C.F., Luo, C.-C., Nakanishi, M.K., Chen, S.-H., Smith, L C., and Chan L. (1990) J. Biol. Chem. 265, 5429-5433). There are two sites for N-linked 15 glycosylation in hepatic and lipoprotein lipase and one in pancreatic lipase. Additionally, four sets of cysteines form disulfide bridges which are essential in maintaining structural integrity for enzymatic activity (Lo, J.-Y., <br><br> Smith, L.C., and Chan, L. (1995) Biochem. Biophys. Res. 20 Commun. 206, 266-271; Brady, L., Brzozowski, A.M., <br><br> Derewenda, Z.S., Dodson, E., Dodson G., Tolley, S., Turkenburg, J.P., Christiansen, L., Huge-Jensen B., Norskov, L., Thim, L., and Menge, U. (1990) Nature 343, 767-770). <br><br> Members of the triacylglycerol lipase family share a 25 number of conserved structural features, one such feature is the "GXSXG" motif, in which the central serine residue is one of the three residues comprising the "catalytic triad" (Winkler, K., D'Arcy, A., and Hunziker, W. (1990) Nature 343, 771-774; Faustinella, F., Smith, L.C., and Chan, L. (1992) 30 Biochemistry 31,7219-7223). Conserved aspartate and histidine residues make up the balance of the catalytic triad. A short span of 19-23 amino acids (the "lid region") forms an amphipathic helix structure and covers the catalytic pocket of the enzyme (Winkler, K., D'Arcy, A., and Hunziker, 35 W. (1990) Nature 343, 771-774). This region diverges significantly between members of the family. It has been determined recently that the span confers substrate <br><br> - 6 - <br><br> specificity to the enzymes (Dugi, K.A., Dichek H.L., and Santamarina-Fojo, S. (1995) J. Biol. Chem. 270, 25396-25401). Comparisons between hepatic and lipoprotein lipase have demonstrated that differences in triacylglycerol lipase and 5 phospholipase activities of the enzymes are in part mediated by this lid region (Dugi, K.A., Dichek H.L., and Santamarina-Fojo, S. (1995) J. Biol. Chem. 270, 25396-25401). <br><br> The triacylglycerol lipases possess varying degrees of heparin binding activity. Lipoprotein lipase has the highest 10 affinity for heparin. This binding activity has been mapped to stretches of positively charged residues in the amino terminal domain (Ma, Y., Henderson, H.E., Liu, M.-S., Zhang, H., Forsythe, I.J., Clarke-Lewis, I., Hayden, M.R., and Brunzell, J.D. J. Lipid Res. 35, 2049-2059). The 15 localization of lipoprotein lipase to the endothelial surface (Cheng, C.F., Oosta, G.M., Bensadoun, A., and Rosenberg, R.D. (1981) J. Biol. Chem. 256, 12893-12896) is mediated primarily through binding to surface proteoglycans (Shimada K., Gill, P.J., Silbert, J.E., Douglas, W.H.J., and Fanburg, B.L. 20 (1981) J. Clin. Invest. 68, 995-1002; Saxena, U., Klein, M.G., and Goldberg, I.J. (1991) J. Biol. Chem. 266, 17516-17521; Eisenberg, S., Sehayek, E., Olivecrona, T., and Vlodavsky, I. (1992) J. Clin Invest. 90,2013-2021). It is this binding activity which allows the enzyme to accelerate 25 LDL uptake by acting as a bridge between LDL and the cell surface (Mulder, M., Lombardi, P., Jansen, H., vanBerkel T. J., Frants R.R., and Havekes, L.M. (1992) Biochem. <br><br> Biophys. Res. Comm. 185, 582-587; Rutledge, J.C., and Goldberg, I.J., (1994) J. Lipid Res. 35. 1152-1160; Tsuchiya, 30 S., Yamabe, M., Yamaguchi, T., Kobayashi, Y., Konno, T., and Tada, K. (1980) Int. J. Cancer 26,171-176). <br><br> Lipoprotein lipase and pancreatic lipase are both known to function in conjunction with co-activator proteins: apolipoprotein CII for lipoprotein lipase; and colipase for 35 pancreatic lipase. <br><br> The genetic sequences encoding human pancreatic lipase, <br><br> ^ <br><br> () <br><br> - 7 - <br><br> hepatic lipase and lipoprotein lipase have been reported (Genbank accession #M93285, #J03540, and #M15856 respectively). The messenger RNAs of human hepatic lipase and pancreatic lipase are approximately 1.7 and 1.8 kilobases 5 in length respectively. Two mRNA transcripts of 3.6 and 3.2 kilobases are produced from the human lipoprotein lipase gene. These two transcripts utilize alternate polyadenylation signals and differ in their translational efficiency (Ranganathan, G., Ong, J.M., Yukht, A., 10 Saghizadeh, M., Simsolo, R.B., Pauer, A., and Kern, P.A. (1995) J. Biol. Chem. 270, 7149-7155). <br><br> Physiological processes <br><br> The metabolism of lipids involves the interaction of lipids, apoproteins, lipoproteins, and enzymes. <br><br> 15 Hepatic lipase and lipoprotein lipase are multifunctional proteins which mediate the binding, uptake, catabolism, and remodeling of lipoproteins and phospholipids. Lipoprotein lipase and hepatic lipase function while bound to the luminal surface of endothelial cells in peripheral 20 tissues and the liver respectively. Both enzymes participate in reverse cholesterol transport, which is the movement of cholesterol from peripheral tissues to the liver either for excretion from the body or for recycling. Genetic defects in both hepatic lipase and lipoprotein lipase are known to be 25 the cause of familial disorders of lipoprotein metabolism. Defects in the metabolism of lipoproteins result in serious metabolic disorders, including hypercholesterolemia, hyperlipidemia, and atherosclerosis. <br><br> REPORTED DEVELOPMENTS <br><br> 30 Atherosclerosis is a complex, polygenic disease which is defined in histological terms by deposits (lipid or fibrolipid plaques) of lipids and of other blood derivatives in blood vessel walls, especially the large arteries (aorta, coronary arteries, carotid). These plaques, which are more 35 or less calcified according to the degree of progression of the atherosclerotic process, may be coupled with lesions and <br><br> - 8 - <br><br> are associated with the accumulation in the vessels of fatty deposits consisting essentially of cholesterol esters. These plagues are accompanied by a thickening of the vessel wall, hypertrophy of the smooth muscle, appearance of foam cells 5 (lipid-laden cells resulting from uncontrolled uptake of cholesterol by recruited macrophages) and accumulation of fibrous tissue. The atheromatous plaque protrudes markedly from the wall, endowing it with a stenosing character responsible for vascular occlusions by atheroma, thrombosis 10 or embolism, which occur in those patients who are most affected. These lesions can lead to serious cardiovascular pathologies such as infarction, sudden death, cardiac ^ insufficiency, and stroke. <br><br> High density lipoprotein (HDL) <br><br> 15 cholesterol levels and atherosclerotic diseases <br><br> High density lipoprotein (HDL) cholesterol levels are inversely associated with risk of atherosclerotic cardiovascular disease (Gordon et al., N. Engl. J. Med., 321, 1311-1316 (1989)). At least 50% of the variation in HDL 20 cholesterol levels is genetically determined (Breslow, J.L., The Metabolic Basis of Inhereited Disease, 2031-2052, McGraw-Hill, New York (1995); Heller et al., N. Engl. J. Med., 328, 1150-1156 (1993)), but the genes responsible for variation in HDL levels have not been fully elucidated. Lipoprotein |j25 lipase (LPL) and hepatic lipase (HL), two members of the triacylglycerol (TG) lipase family, both influence HDL metabolism (Breslow, supra; Murthy et al., Pharmacol. Ther., 70, 101-135 (1996); Goldberg, J.I., J. Lipid Res., 37, 693-707 (1996); Bensadoun et al., Curr. Opin. Lipidol., 7, 77-81 30 (1996)) and the HL (LIPC) locus has been associated with variation in HDL cholesterol levels in humans (Cohen et al., J. Clin. Invest., 94, 2377-2384 (1994); Guerra et al., Proc. Natl. Acad. Sci. USA, 94, 4532-4537 (1997)). The normal range for HDL cholesterol is about 35 to 65 mg/dL, and the 35 HDL level should account for more than 25% of the total cholesterol. <br><br> Very low density lipoprotein (VLDL) <br><br> - 9 - <br><br> and low density lipoprotein (LDL) <br><br> cholesterol levels and atherosclerotic diseases <br><br> High levels of circulating LDL and VLDL cholesterol are associated with increased risk of atherosclerosis. <br><br> 5 VLDL are the precursors of LDL. Therapeutic agents that lower plasma VLDL and LDL cholesterol levels are highly desirable because of the known strong association between these lipid parameters and coronary heart disease. <br><br> Epidemiologic studies have demonstrated a strong 10 relationship between elevated LDL cholesterol and coronary heart disease (CHD) and other atherosclerotic vascular diseases (Kannell, W.B., Am. J. Cardiol., 76, 69C-77C (1995)). Three major secondary prevention trials performed with statins have demonstrated that reduction of LDL 15 cholesterol levels result in significant reduction in CHD <br><br> events and total mortality (Scandinavian Simvastatin Survival Study Group, Lancet, 344, 1383-1389 (1994); Sacks et al., N. Engl. J. Med., 335, 1001-1009 (1996); Tonkin et al., N. Engl. J. Med., 339, 1349-1357 (1998); Grundy, S.M., Circulation, 20 1436-1439 (1998)). Two large primary prevention trials with statins have also demonstrated significant benefit of LDL cholesterol reduction with statins in reducing cardiovascular events (Grundy, supra; Shepherd et al., N. Engl. J. Med., 333, 1301-1307 (1995); Downs et al., JAMA, 279, 1615-1622 25 (1998)). However, current therapies do not adequately reduce LDL cholesterol levels in all persons. VLDL cholesterol levels have also been recognized to be associated with increased risk of CHD (Kannel, supra). Current therapies do not have as much effect in reducing VLDL cholesterol as LDL 30 cholesterol. Therefore, new approaches to reducing both LDL cholesterol and VLDL cholesterol are still needed. <br><br> Ideally, the range for VLDL cholesterol is about 1 to 30 mg/dL and the range for LDL cholesterol is about 60 to 160 mg/dL. The LDL to HDL ratio is ideally less than 3.5. <br><br> - 10 - <br><br> The Role of Triacylglycerol Lipases in Atherosclerotic Diseases <br><br> The role of triacylglycerol lipases in vascular pathologies such as atherosclerosis has been an area of 5 intense study (reviewed in Olivecrona, G., and Olivecrona, T. (1995) Curr. Opin. Lipid. 6,291-305). Generally, the action of the lipoprotein lipase is believed to be antiatherogenic because this enzyme lowers serum triacylglycerol levels and promote HDL formation. Transgenic 10 animals expressing human lipoprotein lipase have decreased levels of plasma triglycerides and an increased level of high density lipoprotein (HDL) (Shimada, M., Shimano, H., Gotoda, T., Yamamoto, K., Kawamura, M., Inaba, T., Yazaki, t., and Yamada, N. (1993) J. Biol. Chem. 268, 17924-17929; Liu, M.-15 S., Jirik, F. R., LeBoeuf, R. C., Henderson, H., <br><br> Castellani, L.W., Lusis, A. J., ma, Y., Forsythe, I.J., <br><br> Zhang, H., Kirk, E., Brunzell, J.D., and Hayden, M.R. (1994) J. Biol. Chem. 269, 11417-11424). Humans with genetic defects resulting in decreased levels of lipoprotein lipase 20 activity have been found to have hypertriglyceridemia, but no increased risk of coronary heart disease. This is reported to be due to the lack of production of intermediate-sized, atherogenic lipoproteins which could accumulate within the subendothelial space (Zilversmit, D.B. (1973) Circ. Res. 25 33,633-638). <br><br> In contrast to lipoprotein lipase (LPL), the physiologic function of HL appears to be related to the metabolism of lipoprotein remnants and HDL (Bensadoun et al., Curr. Opin. Lipidol., 7, 77-81 (1996)). Genetic deficiency of HL is 30 associated with modestly increased levels of remnants and HDL cholesterol in humans (Hegele et al., Arterioscler. Thromb., 13, 720-728 (1993)) and mutant mice (Homanics et al., J. <br><br> Biol. Chem., 270, 2974-2980 (3995)). Despite increased plasma cholesterol levels, HL deficiency is associated with 35 reduced atherosclerosis in apoE mutant mice (Mezdour et al., J. Biol. Chem., 272, 13570-13575 (1997)). Transgenic animals overexpressing HL have decreased HDL (Busch et al., J. Biol. Chem., 269, 16376-16382 (1994); Fan et al., Proc. Natl. Acad. <br><br> (.) <br><br> - 11 - <br><br> Sci. USA, 91, 8724-8728 (1994)). Increased HL activity in humans is associated with low HDL cholesterol. The HL locus on chromosome 15q21 has been associated with variation in plasma HDL cholesterol levels in humans (Cohen et al., J. <br><br> 5 Clin. Invest., 94, 2377-2384 (1994); Guerra et al. , Proc. Natl. Acad. Sci. USA, 94 4532-4537 (1997)), but accounts for only a portion of the genetic contribution to variation in HDL cholesterol levels. There is at least one major locus influencing HDL cholesterol levels in humans that is distinct 10 from the HL locus (Mahaney et al., Arterioscler. Thromb. <br><br> Vase. Biol., 15, 1730-1739 (.1995)). <br><br> In the localized area of an atherosclerotic lesion, the increased level of lipase activity is hypothesized to accelerate the atherogenic process (Zilversmit, D.B. (1995) 15 Clin. Chem. 41,153-158; Zambon, A., Torres, A., Bijvoet, S., Gagne, C., Moojani, S., Lupien, P.J., Hayden M.R. , and Brunzell, J.D. (1993) Lancet 341, 1119-1121). This may be due to an increase in the binding and uptake of lipoproteins by vascular tissue mediated by lipases (Eisenberg, S., 20 Sehayek, E., Olivecrona, T. Vlodavsky, I. (1992) J. Clin. Invest. 90,2013-2021; Tabas, I., Li, I., Brocia R.W., Xu, S.W., Swenson T.L. Williams, K.J. (1993) J. Biol. Chem. 268,20419-20432; Nordestgaard, B.G., and Nielsen, A.G. (1994) Curr. Opin. Lipid. 5,252-257; Williams, K.J., and Tabas, I. 25 (1995) Art. Thromb. and Vase. Biol. 15,551-561). <br><br> Additionally, a high local level of lipase activity may result in cytotoxic levels of fatty acids and lysophosphatidylcholine being produced in precursors of atherosclerotic lesions. <br><br> 30 Despite the understanding that has evolved regarding the role of lipase enzyme activity in regulating the levels of lipids and the various plasma lipoproteins, there is a need to identify and develop therapies which can increase the levels of HDL cholesterol, as well as lower the levels of 35 VLDL and LDL cholesterol to reduce the risk of developing atherosclerotic cardiovascular diseases. <br><br> 12 <br><br> SUMMARY OF THE INVENTION <br><br> In one aspect the invention is a composition capable of lowering the expression of LIPG in a patient comprising a ribozyme which cleaves mRNA which encodes LIPG polypeptide. <br><br> In another aspect the invention is a composition capable of lowering the expression of LIPG in a patient comprising an expression vector which includes a DNA sequence encoding a ribozyme which cleaves mRNA which encodes LIPG polypeptide. <br><br> In one alternative the expression vector is selected from the group consisting of retroviral vectors, adenoviral vectors, adeno-associated viral vectors, herpesviral vectors, and naked DNA vectors. <br><br> In another alternative the ribozyme is a hammerhead ribozyme. <br><br> In another aspect the invention is the use of a ribozyme which cleaves mRNA which encodes LIPG polypeptide in the manufacture of a medicament for raising the level of high density lipoprotein (HDL) cholesterol and apolipoprotein AI in a patient. <br><br> In one alternative the expression vector is selected from the group consisting of retroviral vectors, adenoviral vectors and adeno-associated viral vectors, herpesviral vectors, and naked DNA vectors. <br><br> 134795_1.DOC <br><br> 13 <br><br> / <br><br> INTELLECTUAL PROPERTY office of nz 1 2 APR 2005 <br><br> RECEIVED <br><br> This page is intentionally blank <br><br> 134795J.DOC <br><br> 14 <br><br> ,/ <br><br> INTELLECTUAL PROPERTY Of: <br><br> 1 2 APR 2005 RECEIVED <br><br> This page is intentionally blank <br><br> 134795J.DOC <br><br> 15 <br><br> INTELLECTUAL PRO!: Of- N.Z, <br><br> I 2 APR 2005 <br><br> This page is intentionally blank <br><br> I34795_1.DOC <br><br> 16 <br><br> BRIEF DESCRIPTION OF THE DRAWINGS <br><br> Figure 1 shows the sequences (SEQ ID Nos: 17-31) of the primers used in the exemplified PCR amplifications. <br><br> Figure 2 shows the nucleic acid sequence (SEQ ID NO: 1) and the deduced amino acid sequence (SEQ ID NO: 2) of the differential display RT-PCR product containing the LIPG gene cDNA. The sequences corresponding to the two primers used in the amplification are underlined. The termination codon and polyadenylation signal are boxed. The GAATTC motifs and flanking sequence are from the pCRII vector into which the product was cloned. <br><br> Figure 3 shows the nucleic acid sequence (SEQ ID NO: 3) and the deduced amino acid sequence (SEQ ID NO: 4) of the 5*RACE extension of the LIPG cDNA. The sequences <br><br> INTELLECTUAL PROPERTY QKspcl OF N.Z. " | <br><br> ' 2 APR 2005 I RECEIVFn I <br><br> 134795_1.DOC <br><br> - 17 - <br><br> corresponding to the two primers used in the amplification are underlined. The GAATTC motifs and flanking sequence are from the pCRII vector into which the product was cloned. <br><br> Figure 4 shows the sequence (SEQ ID NO: 7) of the cDNA 5 containing the complete open reading frame of the LIPG gene, LLGXL. The start codon (ATG) and termination codon (TGA) are boxed. The Dral site (TTTAAA) and Srfl site (GCCCGGGC) used in the construction of the expression vectors are underlined. <br><br> Figure 5 shows the deduced amino acid sequence (SEQ ID 10 NO: 8) of the LLGXL protein. The predicted signal sequence is underlined. <br><br> Figure 6 shows a protein sequence alignment of the members of the triacylglycerol lipase gene family (SEQ ID Nos: 13-15) . Shaded residues are identical to the LLGXL 15 protein (SEQ ID NO: 8) . The deduced amino acid sequence of human LIPG (EL) is provided on the top line and is compared with the other major members of the TG lipase family, LPL, HL and PL. EL residues identical to those in at least one other member of the family are shaded as well as the corresponding 20 residue in the other family member. Amino acids are numbered according to convention beginning with the initial residue of the secreted protein. The predicted sites of signal peptide cleavage are marked with a solid line between amino acid residues. The GXSXG lipase motif containing the active 25 serine is boxed. The amino acids of the catalytic triad are marked with an asterisk. The conserved cysteines are marked with filled circles. Potential N-linked glycosylation sites are marked with arrowheads. The lid region is indicated by a bold line. Gaps were introduced into the sequences to 30 maximize the alignment values using the CLUSTAL program. <br><br> Figure 7 shows a northern analysis of LIPG mRNA in THP-1 cells. Cells were stimulated with either PMA or PMA and oxidized LDL (PMA + oxLDL) . Numbers at the left indicate the positions of RNA standards (in kilobases). <br><br> t ) <br><br> i ) <br><br> - 18 - <br><br> Figure 8 shows a northern-blot analysis of expression of LIPG mRNA compared with LPL in human tissues. A blot containing mRNA from the indicated human tissues was incubated with radiolabelled LPL and (5-actin (ACTB) probes as 5 described. <br><br> Figure 9 shows a Northern-blot analysis of cultured cell lines. The panel on the left (lanes 1-6) was hybridized with the LIPG (EL) probe and that on the right (lanes 7-12) with the LPL probe. Lanes 1, 7, unstimulated HUVEC; lanes 2, 8, 10 HUVEC stimulated with PMA; lanes 3, 9, HUVEC stimulated with thrombin; lanes 4, 10, unstimulated HCAEC; lanes 5, 11, HCAEC stimulated with PMA; lanes 6, 12, THP-1 stimulated with PMA. <br><br> Figure 10 shows the sequence of the immunizing peptide (SEQ ID NO: 16) and its relation to the LLGXL protein 15 sequence. The peptide is shown in the shaded box. The terminal cysteine was introduced to aid coupling of the peptide to the carrier protein. <br><br> Figure 11 shows the results obtained when conditioned media from HUVEC and HCAEC were subjected to immunoblot 20 analysis with rabbit anti-EL peptide antiserum. Lane l, unconditioned media; lane 2, unstimulated HUVEC; lane 3, <br><br> HUVEC stimulated with PMA; lane 4, unstimulated HCAEC; lane 5, HCAEC stimulated with PMA. <br><br> Figure 12 shows a western analysis of heparin-Sepharose 25 bound proteins in conditioned medium from COS-7 cells transiently transfected with an expression vector containing a cDNA for LLGN or LLGXL or no DNA (Mock). Proteins from PMA-stimulated endothelial cells (HCAEC + PMA) were included for size reference. Numbers to the left indicate the 30 apparent molecular weight of the major immunoreactive proteins as determined by a comparison to protein standards. <br><br> Figure 13 shows the sequence of the rabbit LIPG PCR product (RLLG.SEQ, SEQ ID NO: 12) and the sequence alignment between the rabbit LIPG PCR product and the corresponding <br><br> - 19 - <br><br> sequence in the human cDNA (LLG7742A) . Identical nucleotides are shaded. <br><br> Figure 14 shows the phospholipase A activity of human EL-AS, EL and LPL using a phosphatidylcholine substrate. To 5 perform the assay 700/il of conditioned medium harvested from COS—7 cells transiently transfected with either pcDNA3.0/LIPG-AS, LIPG, or LPL expression constructs were assayed in triplicate for phospholipase activities as described below. Following a two hour incubation at 37°C, 10 reactions were terminated, and 14C labeled free fatty-acid was extracted, and counted to determine the amount of free fatty-acid produced. <br><br> Figure 15 shows the triacylglyceride lipase activity of human EL-AS, EL and LPL using a triolein substrate. To 15 perform the assay 700/il of conditioned medium harvested from COS—7 cells transiently transfected with either pcDNA3.0/LIPG-AS, LIPG, or LPL expression constructs was assayed in triplicate for triglyceride activities described below. Following a two hour incubation at 37°C, reactions 20 were terminated, and 14C labeled free fatty-acid was extracted, and counted to determine the amount of free fatty-acid produced. <br><br> Figure 16 shows the hybridization of LIPG and LPL probes to genomic DNAs from different species. <br><br> 25 Figure 17 shows expression of LIPG in the liver of a wild-type mouse 5 days after AdhEL injection. Lane 1, liver from mouse injected with Adnuli; lane 2, liver from mouse injected with AdhEL. <br><br> Figure 18 shows plasma levels of HDL cholesterol in 30 AdhEL- and Adnull-injected wild-type mice. <br><br> Figure 19 shows lipoprotein profiles in wild-type mice injected with AdhEL and Adnuli at baseline before injection (left) and 14 days after injection (right) . <br><br> ) <br><br> - 20 - <br><br> Figure 20 shows HDL cholesterol levels in human apoA-I transgenic mice after injection with Adnull or AdhEL. <br><br> Figure 21 shows ApoA-I levels in human apoA-1 transgenic mice after injectiori with Adnull or AdhEL. <br><br> 5 Figure 22 shows the effect of injection of AdhEL in LDL <br><br> receptor-deficient mice on VLDL/LDL cholesterol levels. <br><br> Figure 23 shows the effect of AdhEL on HDL receptor-deficient mice on HDL cholesterol levels. <br><br> Figure 24 is a western blot analysis-of LIPG (EL) 10 demonstrating the effect of glycosidase and tunicamycin on EL. Moving from left to right, the lanes are: untreated EL control; EndoF-treated EL; EndoH-treated EL; neuraminidase-treated EL; marker, plasma sample; EL untreated lysate; EL tunicamycin-treated lysate; EL untreated pellet; EL 15 tunicamycin-treated pellet; EL untreated medium; EL tunicamycin-treated medium, marker; plasma. <br><br> Figure 25 is a western blot illustrating the effect on EL of heparin administration. "Pre*! lanes are blood samples taken from mice before heparin injection; "post" lanes are 20 blood samples taken after heparin injection. "Control virus" * did not include EL; "EL virus" was an adenoviral vector expressing EL. <br><br> Figures 26A and 26B are bar graphs illustrating the triglyceridase activity (Fig. 26A) and the phospholipase 25 activity (Fig. 26B) of LPL (lipoprotein lipase), HL (hepatic lipase) and EL (endothelial lipase). <br><br> Figures 27A and 27b are graphs illustrating the effect of human serum on lipoprotein lipase triglyceridase activity (Fig. 27A) and endothelial lipase triglyceridase activity 30 (Fig. 27B). <br><br> Figures 28A and 28B illustrate expression of EL at different virus doses. Figure 28A is a western blot illustrating the EL present at the different viral doses. Figure 28B illustrates phospholipase activity at the 5 different viral dosages. <br><br> DETAILED DESCRIPTION OF THE INVENTION <br><br> The detailed description which follows sets forth the basis for the present invention, followed by a definitions section. Following the definitions section, the various 10 compositions useful in the practice of the invention are discussed, followed by a discussion of the methods used to lower or raise the levels of LIPG activity. <br><br> The Enzvmatic Activity of the LIPG Gene Product <br><br> The present invention relates to methods for regulating 15 the levels of HDL cholesterol and apolipoprotein AI, VLDL cholesterol and LDL cholesterol utilizing methods and compositions which lower or raise the activity of the LIPG lipase enzyme. In particular, the present invention is based in part on the discovery of the enzymatic activity of the 20 polypeptide products of the LIPG gene on HDL cholesterol and apolipoprotein AI, VLDL cholesterol and LDL cholesterol. The polypeptide products of LIPG are members of the triacylglycerol lipase family and comprise an approximately 39 kD catalytic domain of the triacylglycerol lipase family, 25 e,g., having the sequence SEQ ID NO: 10. Because this newly discovered lipase was found to be synthesized by endothelial cells and this is a unique feature compared with other members of the triacylglycerol lipase family, this lipase has been named "endothelial lipase" (EL). Because the LIPG gene 30 will be discussed extensively in the sections which follow, EL will be hereinafter referred to as LIPG polypeptide, for the purposes of clarity. In general, the LIPG polypeptide is found in two major forms, referred to hereinafter as "the LLGN polypeptide" and "the LLGXL polypeptide." The LLGN 35 polypeptide, has 354 amino acids. The LLGXL polypeptide has 500 amino acids and exhibits 43% similarity to human lipoprotein lipase and 37% similarity to human hepatic lipase. As used herein, the term "LIPG polypeptide" or "LIPG <br><br> - 22 - <br><br> protein" encompasses both LLGN and LLGXL. <br><br> The sequence of the LIPG polypeptide contains the characteristic GXSXG lipase motif, a conserved catalytic triad, a 19-residue lid region, conserved heparin and 5 lipoprotein binding sites and 5 potential N-linked glycosylation sites. The region with the greatest sequence divergence in the triacylglycerol lipase family is the lid domain, which forms an amphipathic helix covering the catalytic pocket of the enzyme (Winkler et al., Nature, 343, 10 771-774 (1990); van Tilbeurgh et al., J. Biol. Chem., 269, 4626-4633 (1994)) and confers substrate specificity to the enzymes of this family (Dugi et al., J. Biol. Chem., 270, 25396-25401 (1995)). The 19-residue lid region of LIPG is I three residues shorter and less amphipathic than those found 15 in lipoprotein lipase and hepatic lipase, consistent with a different enzymatic profile. The predicted molecule weight of the mature protein is approximately 55 kD; a 68 kD form is likely to be a glycosylated form, whereas a 40 kD form may be the product of a specific proteolytic cleavage. <br><br> 20 The LIPG polypeptide has the ability to lower the levels of HDL cholesterol and apolipoprotein AI as well as the levels of VLDL cholesterol and LDL cholesterol. It is well established that lowered HDL cholesterol levels result in increased susceptibility to atherosclerosis and increased 25 levels of HDL cholesterol can dramatically reduce " susceptibility to atherosclerosis. <br><br> One physiologic role of LIPG may be to hydrolyse HDL phospholipid in peripheral tissues and in liver to facilitate selective uptake of HDL cholesteryl ester via the HDL 30 receptor SR-BI (Kozarsky et al., Nature, 387, 414-417 <br><br> (1997)). Another possible role is the facilitation of apoB-containing remnant lipoprotein uptake, similar to the role of hepatic lipase (Mahley et al., J. Lipid Res., 40, 1-16 (1999)). In addition, LIPG is abundantly expressed in the 35 placenta, and a role for this enzyme in development is possible, given the importance of lipid transport in fetal development (Farese et al., Trends Genet., 14, 115-120 <br><br> - 23 - <br><br> (1998)) . <br><br> Based on HDL cholesterol•s beneficial properties, it is desirable to raise HDL cholesterol levels by lowering the enzymatic activity of LIPG. Accordingly, the present 5 invention is directed to methods and compositions which lower the activity of LIPG in the body by lowering the expression of the LIPG gene or lowering the enzymatic activity of the LIPG polypeptide. <br><br> Given the ability of the LIPG polypeptide to reduce the 10 levels of VLDL cholesterol and LDL cholesterol and the studies demonstrating the correlation between high levels of these compounds and atherosclerotic diseases, it is desirable to lower the level of these compounds in a patient. Accordingly, the present invention additionally provides 15 methods and compositions for increasing the expression of the LIPG gene and increasing the enzymatic activity of the LIPG polypeptides. <br><br> There are set forth hereafter definitions of terms used herein and descriptions of preferred embodiments of the 20 present invention. <br><br> Definitions <br><br> The following defined terms are used throughout the present specification and should be helpful in understanding the scope and practice of the present invention. <br><br> 25 A "polypeptide" is a polymeric compound comprised of covalently linked amino acid residues. Amino acids are classified into seven groups on the basis of the side chain: (1) aliphatic side chains, (2) side chains containing a hydroxylic (OH) group, (3) side chains containing sulfur 30 atoms, (4) side chains containing an acidic or amide group, (5) side chains containing a basic group, (6) side chains containing an aromatic ring, and (7) proline, an imino acid in which the side chain is fused to the amino group. <br><br> - 24 - <br><br> A "protein" is a polypeptide which plays a structural or functional role in a living cell. <br><br> The polypeptides and proteins of the invention may be glycosylated or ungiycosylated. <br><br> 5 "Homology" means similarity of sequence reflecting a common evolutionary origin. Polypeptides or proteins are said to have homology, or similarity, if a substantial number of their amino acids are either (1) identical, or (2) have a chemically similar side chain. Nucleic acids are said to 10 have homology if a substantial number of their nucleotides are identical. <br><br> "Isolated polypeptide" or "isolated protein" is a polypeptide or protein which is substantially free of those compounds that are normally associated therewith in its 15 natural state (e.g., other proteins or polypeptides, nucleic acids, carbohydrates, lipids). "Isolated" is not meant to exclude artificial or synthetic mixtures with other compounds, or the presence of impurities which do not interfere with biological activity, and which may be present, 20 for example, due to incomplete purification, addition of stabilizers, or compounding into a pharmaceutical^ <br><br> acceptable preparation. <br><br> A molecule is "antigenic" when it is capable of specifically interacting with an antigen recognition molecule 25 of the immune system, such as an immunoglobulin (antibody) or T cell antigen receptor. An antigenic polypeptide contains at least about 5, and preferably at least about 10, amino acids. An antigenic portion of a molecule can be that portion that is immunodominant for antibody or T cell 30 receptor recognition, or it can be a portion used to generate an antibody to the molecule by conjugating the antigenic portion to a carrier molecule for immunization. A molecule that is antigenic need not be itself immunogenic, i.e., capable of eliciting an immune response without a carrier. <br><br> - 25 - <br><br> "LLGN polypeptide" and "LLGN protein" mean a polypeptide including the sequence SEQ ID NO: 6, said polypeptide being glycosylated or non-glycosylated. <br><br> "LLGXL polypeptide" and "LLGXL protein" mean a 5 polypeptide including the sequence SEQ ID NO: 8, said polypeptide being glycosylated or non-glycosylated. <br><br> "LIPG polypeptide" and "LIPG protein" describe the lipase enzyme encoded by the LIPG gene and generically describes both the LLGN polypeptide and the LLGXL 10 polypeptide. <br><br> "Endothelial lipase," or "EL", refer to the lipase enzyme encoded by the LIPG gene and is equivalent to the term LIPG polypeptide. <br><br> The LIPG polypeptide or protein of the invention 15 includes any analogue, fragment, derivative, or mutant which is derived from an LIPG polypeptide and which retains at least one biological property of the LIPG polypeptide. Different variants of the LIPG polypeptide exist in nature. These variants may be allelic variations characterized by 20 differences in the nucleotide sequences of the structural gene coding for the protein, or may involve differential splicing or post-translational modification. The skilled artisan can produce variants having single or multiple amino acid substitutions, deletions, additions, or replacements. 25 These variants may include, inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids, (b) variants in which one or more amino acids are added to the LIPG polypeptide, (c) variants in which one or more of the amino 30 acids includes a substituent group, and (d) variants in which the LIPG polypeptide is fused with another polypeptide such as serum albumin. Other LIPG polypeptides of the invention include variants in which amino acid residues from one species are substituted for the corresponding residue in 35 another species, either at conserved or non-conserved <br><br> - 26 - <br><br> positions. In another embodiment, amino acid residues at non-conserved positions are substituted with conservative or non-conservative residues. The techniques for obtaining these variants, including genetic (suppressions, deletions, 5 mutations, etc.), chemical, and enzymatic techniques, are known to persons having ordinary skill in the art. <br><br> If such allelic variations, analogues, fragments, derivatives, mutants, and modifications, including alternative mRNA splicing forms and alternative post-10 translational modification forms result in derivatives of the LIPG polypeptide which retain any of the biological properties of the LIPG polypeptide, they are included within the scope of this invention. <br><br> A "nucleic acid" is a polymeric compound comprised of 15 covalently linked subunits called nucleotides. Nucleic acid includes polyribonucleic acid (RNA) and polydeoxyribonucleic acid (DNA), both of which may be single-stranded or double-stranded. DNA includes cDNA, genomic DNA, synthetic DNA, and semi-synthetic DNA. The sequence of nucleotides that encodes 20 a protein is called the sense sequence. <br><br> An "antisense nucleic acid" is a sequence of nucleotides that is complementary to the sense sequence. Antisense nucleic acids can be used to down regulate or block the expression of the polypeptide encoded by the sense strand. <br><br> 25 "Isolated nucleic acid" means a nucleic acid which is substantially free of those compounds that are normally associated therewith in its natural state. "Isolated" is not meant to exclude artificial or synthetic mixtures with other compounds, or the presence of impurities which do not 30 interfere with biological activity, and which may be present, for example, due to incomplete purification, addition of stabilizers, or compounding into a pharmaceutically acceptable preparation. <br><br> The phrase "a nucleic acid vhicfc hybridizes at high <br><br> o <br><br> - 27 - <br><br> stringency" means that the hybridized nucleic acids are able to withstand a washing under high stringency conditions. An example of high stringency washing conditions for DNA-DNA hybrids is 0.1X SSC, 0.5% SDS at 68°c. other conditions of 5 high stringency washing are known to persons having ordinary skill in the art. <br><br> "Regulatory region** means a nucleic acid sequence which regulates the expression of a nucleic acid. A regulatory region may include sequences which are naturally responsible 10 for expressing a particular nucleic acid (a homologous region) or may include sequences of a different origin (responsible for expressing different proteins or even synthetic proteins). In particular, the sequences can be sequences of eukaryotic or viral genes or derived sequences 15 which stimulate or repress transcription of a gene in a specific or non-specific manner and in an inducible or non-inducible manner. Regulatory regions include origins of replication, RNA splice sites, enhancers, transcriptional termination sequences, signal sequences which direct the 20 polypeptide into the secretory pathways of the target cell, and promoters. <br><br> A regulatory region from a "heterologous source" is a regulatory region which is not naturally associated with the expressed nucleic acid. Included among the heterologous 25 regulatory regions are regulatory regions from a different species, regulatory regions from a different gene, hybrid regulatory sequences, and regulatory sequences which do not occur in nature, but which are designed by one having ordinary skill in the art. <br><br> 30 A "vector" is any means for the transfer of a nucleic acid according to the invention into a host cell. The term "vector" includes both viral and nonviral means for introducing the nucleic acid into a prokaryotic or eukaryotic cell in vitro, ex vivo or in vivo. Non-viral vectors include 35 plasmids, liposomes, electrically charged lipids <br><br> (cytofectins), DNA-protein complexes, and biopolymers. Viral <br><br> 9 <br><br> - 28 - <br><br> vectors include retrovirus, adeno-associated virus, pox, baculovirus, vaccinia, herpes simplex, Epstein-Barr and adenovirus vectors. In addition to nucleic acid according to the invention, a vector may also contain one or more 5 regulatory regions, and/or selectable markers useful in selecting, measuring, and monitoring nucleic acid transfer results (transfer to which tissues, duration of expression, etc.). <br><br> A "recombinant cell" is a cell which contains a nucleic 10 acid which is not naturally present in the cell. <br><br> "Recombinant cell" includes higher eukaryotic cells such as mammalian cells, lower eukaryotic cells such as yeast cells, prokaryotic cells, and archaebacterial cells. <br><br> "Pharmaceutically acceptable carrier" includes diluents 15 and fillers which are pharmaceutically acceptable for methods of administration, are sterile, and may be aqueous or oleaginous suspensions formulated using suitable dispersing or wetting agents and suspending agents. The particular pharmaceutically acceptable carrier and the ratio of active 20 compound to carrier are determined by the solubility and chemical properties of the composition, the particular mode of administration, and standard pharmaceutical practice. <br><br> . A "lipase" is a protein which can enzymatically cleave a lipid substrate. <br><br> 25 A "phospholipase" is a protein which can enzymatically cleave a phospholipid substrate. <br><br> A "triacylglycerol lipase" is a protein which can enzymatically cleave a triacylglyceride substrate. <br><br> "Phosphatidylcholine" is a glycerol phospholipid. 30 Phosphatidylcholine is also known as lecithin. <br><br> "Lipid profile" means the set of concentrations of cholesterol, triglyceride, lipoprotein cholesterol and other <br><br> f) <br><br> - 29 - <br><br> lipids in the body of a human or other animal. <br><br> An "undesirable lipid profile" is the condition in which the concentrations of cholesterol, triglyceride, or lipoprotein cholesterol are outside of the age- and gender-5 adjusted reference ranges. Generally, a concentration of total cholesterol &gt; 200 mg/dl, of plasma triglycerides &gt;200 mg/dl, of LDL cholesterol &gt; 130 mg/dl, of HDL cholesterol &lt; 39 mg/dl, or a ratio of total cholesterol to HDL cholesterol &gt; 4.0 is considered to.be an undesirable lipid profile. An 10 undesirable lipid profile is associated with a variety of pathological conditions, including hyperlipidaemias, diabetes hypercholesterolaemia, atherosclerosis, and other forms of coronary artery disease. <br><br> A "ribozyme" is an RNA molecule which can function as an 15 enzyme. <br><br> A "neutralizing antibody" is an antibody which can bind to an LIPG polypeptide and lower or eliminate the enzymatic activity of the LIPG polypeptide. These antibodies may be monoclonal antibodies or polyclonal antibodies. The present 20 invention includes chimeric, single chain, and humanized antibodies, as well as Fab fragments and the products of an Fab expression library, and Fv fragments and the products of an Fv expression library. <br><br> An "inhibitory molecule" or "inhibitor" is a molecule 25 which lowers or eliminates the expression of the LIPG polypeptide or which lowers or eliminates the enzymatic activity of the LIPG polypeptide. <br><br> An "enhancer molecule" or "enhancer" is a molecule which increases the expression of the LIPG polypeptide or which 30 increases the enzymatic activity of the LIPG polypeptide. <br><br> A "liposome" is is an artificial or naturally-occurring phospholipid vesicle. <br><br> - 30 - <br><br> A "cationic liposome" is a liposome having a net positive electrical charge. <br><br> The sections which follow discuss the elements used in the claimed methods and compositions and the preferred 5 embodiments of these elements. <br><br> polypeptides <br><br> The present invention utilizes polypeptides encoded by LIPG which are members of the triacylglycerol lipase family, and which comprise a 39 kD catalytic domain of the 10 triacylglycerol lipase family, e.g., having the sequence SEQ ID NO: 10. In certain embodiments of the present invention, an isolated LIPG polypeptide comprising the sequence SEQ ID NO: 6 and having an apparent molecular weight of about 40 kD on a 10% SDS-PAGE gel is utilized. In another embodiment of 15 the present invention, an isolated LIPG polypeptide comprising the sequence SEQ ID NO: 8 and having an apparent molecular weight of about 55 kD or 68 kD on a 10% SDS-PAGE gel is utilized. In yet another embodiment, the polypeptides utilized in the present invention are subfragments of these 20 polypeptides. In still yet another embodiment, the polypeptides used in the present invention are antibodies capable of binding to an LIPG polypeptide. <br><br> The polypeptides and proteins utilized in the present ' invention may be recombinant polypeptides, natural 25 polypeptides, or synthetic polypeptides, and may be of human, rabbit, or other animal origin. The polypeptides are characterized by a reproducible single molecular weight and/or multiple set of molecular weights, chromatographic response and elution profiles, amino acid composition and 30 sequence, and biological activity. <br><br> The polypeptides utilized in the present invention may be isolated from natural sources, such as placental extracts, human, plasma, or conditioned media from cultured cells such as macrophages or endothelial cells, by using the 35 purification procedures known to one of skill in the art. <br><br> - 31 - <br><br> Alternatively, the polypeptides utilized in the present invention may be prepared utilizing recombinant DNA technology, which comprises combining a nucleic acid encoding the polypeptide thereof in a suitable vector, inserting the 5 resulting vector into a suitable host cell, recovering the polypeptide produced by the resulting host cell, and purifying the polypeptide recovered. <br><br> Nucleic Acids <br><br> The present invention utilizes isolated nucleic acids 10 which encode LIPG polypeptides. <br><br> The present invention also utilizes antisense nucleic acids which can be used to down regulate or block the expression of LIPG polypeptides in vitro, ex vivo or in vivo. <br><br> The techniques of recombinant DNA technology are known 15 to those of ordinary skill in the art. General methods for the cloning and expression of recombinant molecules are described in Maniatis (Molecular Cloning. Cold Spring Harbor Laboratories, 1982), and in Ausubel (Current Protocols in Molecular Biology. Wiley and Sons, 1987), which are 20 incorporated by reference. <br><br> The nucleic acids of the present invention may be linked to one or more regulatory regions. Selection of the appropriate regulatory region or regions is a routine matter, within the level of ordinary skill in the art. Regulatory 25 regions include promoters, and may include enhancers, suppressors, etc.. <br><br> Promoters that may be used in the present invention include both constituitive promoters and regulated (inducible) promoters. The promoters may be prokaryotic or 30 eukaryotic depending on the host. Among the prokaryotic (including bacteriophage) promoters useful for practice of this invention are lacl, lacZ, T3, T7, lambda Pr, Plf and trp promoters. Among the eukaryotic (including viral) promoters useful for practice of this invention are ubiquitous <br><br> /' X <br><br> ) <br><br> - 32 - <br><br> promoters (e.g. HPRT, vimentin, actin, tubulin), intermediate filament promoters (e.g. desmin, neurofilaments, keratin, GFAP), therapeutic gene promoters (e.g. MDR type, CFTR, <br><br> factor VIII), tissue-specific promoters (e.g. actin promoter 5 in smooth muscle cells, or Fit and Flk promoters active in endothelial cells), including animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells (Swift et 10 al., 1984, Cell 38:639-646; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol., 50:399-409; MacDonald, 1987, Hepatology 7:425-515); insulin gene control region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-122), immunoglobulin gene control region which is 15 active in lymphoid cells (Grosschedl et al., 1984, Cell 38:647-658; Adames et al., 1985, Nature 318:533-538; <br><br> Alexander et al., 1987, Mol. Cell. Biol., 7:1436-1444), mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 20 1986, Cell 45:485-495), albumin gene control region which is active in liver (Pinkert et al., 1987, Genes and Devel. 1:268-276), alpha-fetoprotein gene control region which is active in liver (Krumlauf et al., 1985, Mol. Cell. Biol., 5:1639-1648; Hammer et al., 1987, Science 235:53-58), alpha 25 1-antitrypsin gene control region which is active in the . liver (Kelsey et al., 1987, Genes and Devel., 1:161-171), beta-globin gene control region which is active in myeloid cells (Mogram et al., 1985, Nature 315:338-340; Kollias et al., 1986, Cell 46:89-94), myelin basic protein gene control 30 region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712), myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286), and gonadotropic releasing hormone gene control region which is active in the 35 hypothalamus (Mason et al., 1986, Science 234:1372-1378). <br><br> Other promoters which may be used in the practice of the invention include promoters which are preferentially activated in dividing cells, promoters which respond to a <br><br> o <br><br> - 33 - <br><br> stimulus (e.g. steroid hormone receptor, retinoic acid receptor), tetracycline-regulated transcriptional modulators, cytomegalovirus immediate-early, retroviral LTR, metallothionein, SV-40, Ela, and MLP promoters. <br><br> 5 Tetracycline-regulated transcriptional modulators and CMV promoters are described in WO 96/01313, US 5,168,062 and 5,385,839, the contents of which are incorporated herein by reference. <br><br> Viral Vector Systems <br><br> 10 Preferably, the viral vectors used in the gene therapy methods of the present invention are replication defective, that is, they are unable to replicate autonomously in the target cell. In general, the genome of the replication defective viral vectors which are used within the scope of 15 the present invention lack at least one region which is necessary for the replication of the virus in the infected cell. These regions can either be eliminated (in whole or in part), or be rendered non-functional by any technique known to a person skilled in the art. These techniques include the 20 total removal, substitution (by other sequences, in particular by the inserted nucleic acid), partial deletion or addition of one or more bases to an essential (for replication) region. Such techniques may be performed in vitro (on the isolated DNA) or in situ, using the techniques 25 of genetic manipulation or by treatment with mutagenic agents. <br><br> Preferably, the replication defective virus retains the sequences of its genome which are necessary for encapsidating the viral particles. <br><br> 30 The retroviruses are integrating viruses which infect dividing cells. The retrovirus genome includes two LTRs, an encapsidation sequence and three coding regions (gag, pol and env) . The construction of recombinant retroviral vectors has been described: see, in particular, EP 453242, EP178220, 35 Bernstein et al. Genet. Eng. 7 (1985) 235; McCormick, <br><br> BioTechnology 3 (1985) 689, etc. In recombinant retroviral <br><br> - 34 - <br><br> vectors, the gag, pol and env genes are generally deleted, in whole or in part, and replaced with a heterologous nucleic acid sequence of interest. These vectors can be constructed from different types of retrovirus, such as, MoMuLV ("murine 5 Moloney leukaemia virus" MSV ("murine Moloney sarcoma virus"), HaSV ("Harvey sarcoma virus"); SNV ("spleen necrosis virus"); RSV ("Rous sarcoma virus") and Friend virus. Lentivirus vector systems may also be used in the practice of the present invention. The lentiviral genome is a positive-10 strand polyadenylated RNA of 9,000 to 10,000 base pairs containing three structural genes organized 5' to 3' (gag, pol, env), typical of all retroviruses. For an extensive review of lentiviral systems, see Fields Virology, Second Edition, Volume 2, Chapter 55, "Lentiviruses," pp. 1571-1589, 15 Raven Press, New York, 1990. <br><br> In general, in order to construct recombinant retroviruses containing a sequence encoding LIPG, a plasmid is constructed which contains the LTRs, the encapsidation sequence and the coding sequence. This construct is used to 20 transfect a packaging cell line, which cell line is able to supply in trans the retroviral functions which are deficient in the plasmid. In general, the packaging cell lines are thus able to express the gag,.pol and env genes. Such packaging cell lines have been .described in the prior art, in 25 particular the cell line PA317 (US4,861,719); the PsiCRIP cell line (W090/02806) and the GP+envAm-12 cell line (W089/07150). In addition, the recombinant retroviral vectors can contain modifications within the LTRs for suppressing transcriptional activity as well as extensive 30 encapsidation sequences which may include a part of the gag gene (Bender et al., J. Virol. 61 (1987) 1639). Recombinant retroviral vectors are purified by standard techniques known to those having ordinary skill in the art. <br><br> The adeno-associated viruses (AAV) are DNA viruses of 35 relatively small size which can integrate, in a stable and site-specific manner, into the genome of the cells which they infect. They are able to infect a wide spectrum of cells <br><br> - 35 - <br><br> without inducing any effects on cellular growth, morphology or differentiation, and they do not appear to be involved in human pathologies. The AAV genome has been cloned, sequenced and characterized. It encompasses approximately 4700 bases 5 and contains an inverted terminal repeat (ITR) region of approximately 145 bases at each end, which serves as an origin of replication for the virus. The remainder of the genome is divided into two essential regions which carry the encapsidation functions: the left-hand part of the genome, 10 which contains the rep gene involved in viral replication and expression of the viral genes; and the right-hand part of the genome, which contains the cap gene encoding the capsid proteins of the virus. <br><br> The use of vectors derived from the AAVs for 15 transferring genes in vitro and in vivo has been described <br><br> (see WO 91/18088; WO 93/09239; US 4,797,368, US 5,139,941, EP 488 528). These publications describe various AAV-derived constructs in which the rep and/or cap genes are deleted and replaced by a gene of interest, and the use of these 20 constructs for transferring the said gene of interest in vitro (into cultured cells) or in vivo, (directly into an organism). The replication defective recombinant AAVs utilized in the present invention can be prepared by cotransfecting a plasmid containing the nucleic acid sequence 25 of interest flanked by two AAV inverted terminal repeat (ITR) regions, and a plasmid carrying the AAV encapsidation genes (rep and cap genes), into a cell line which is infected with a human helper virus (for example an adenovirus). The AAV recombinants which are produced are then purified by standard 30 techniques. The invention also relates, therefore, to an AAV-derived recombinant virus whose genome encompasses a sequence encoding an LIPG polypeptide flanked by the AAV ITRs. The invention also relates to a plasmid encompassing a sequence encoding an LIPG polypeptide flanked by two ITRs from an AAV. 35 Such a plasmid can be used as it is for transferring the LIPG sequence, with the plasmid, where appropriate, being incorporated into a liposomal vector (pseudo-virus). <br><br> - 36 - <br><br> In a preferred embodiment, the vector utilized in the present invention is an adenovirus vector. <br><br> Adenoviruses are eukaryotic DNA viruses that can be modified to efficiently deliver a nucleic acid to a variety 5 of cell types. <br><br> Various serotypes of adenovirus exist. Of these serotypes, preference is given, within the scope of the present invention, to using type 2 or type 5 human adenoviruses (Ad 2 or Ad 5) or adenoviruses of animal origin 10 (see W094/26914) . Those adenoviruses of animal origin which can be used within the scope of the present invention include adenoviruses of canine, bovine, murine (example: Mavl, Beard et al., Virology 75 (1990) 81), ovine, porcine, avian, and simian (example: SAV) origin. Preferably, the adenovirus of 15 animal origin is a canine adenovirus, more preferably a CAV2 adenovirus (e.g. Manhattan or A26/61 strain (ATCC VR-800), for example). <br><br> Preferably, the replication defective adenoviral vectors comprise the ITRs, an encapsidation sequence and the nucleic 20 acid of interest. Still more preferably, at least the El region of the adenoviral vector is non-functional. The deletion in the El region preferably extends from nucleotides 455 to 3329 in the sequence of the Ad5 adenovirus. Other ) regions may also be modified, in particular the E3 region 25 (W095/02697), the E2 region (W094/28938), the E4 region (W094/28152, W094/12649 and W095/02697), or in any of the late genes L1-L5. Defective retroviral vectors are disclosed in W095/02697. <br><br> In a preferred embodiment, the adenoviral vector has a 30 deletion in the El and E4 regions. In another preferred embodiment, the adenoviral vector has a deletion in the El region into which the E4 region and the sequence encoding LLG are inserted (see FR94 13355). <br><br> - 37 - <br><br> The replication defective recombinant adenoviruses can be prepared by any technique known to the person skilled in the art (Levrero et al., Gene 101 (1991) 195, EP 185 573; Graham, EMBO J. 3 (1984) 2917). In particular, they can be 5 prepared by homologous recombination between an adenovirus and a plasmid which carries, inter alia, the DNA sequence of interest. The homologous recombination is effected following cotransfection of the said adenovirus and plasmid into an appropriate cell line. The cell line which is employed 10 should preferably (i) be transformable by the said elements, and (ii) contain the sequences which are able to complement the part of the genome of the replication defective adenovirus, preferably in integrated form in order to avoid the risks of recombination. Examples of cell lines which may 15 be used are the human embryonic kidney cell line 293 (Graham et al., J. Gen. Virol. 36 (1977) 59) which contains the left-hand portion of the genome of an Ad5 adenovirus (12%) integrated into its genome, and cell lines which are able to complement the El and E4 functions, as described in 20 applications W094/26914 and W095/02697. Recombinant adenoviruses are recovered and purified using standard molecular biological techniques, which are well known to one of ordinary skill in the art. <br><br> Antisense Nucleic Acids <br><br> 25 The down regulation of gene expression using antisense nucleic acids can be achieved at the translational or transcriptional level. Antisense nucleic acids of the invention are preferably nucleic acid fragments capable of specifically hybridizing with all or part of a nucleic acid 30 encoding LIPG or the corresponding messenger RNA. In addition, antisense nucleic acids may be designed or identified which decrease expression of the LIPG gene by inhibiting splicing of its primary transcript. With knowledge of the structure and partial sequence of the LIPG 35 gene, such antisense nucleic acids can be designed and tested for efficacy. <br><br> - 38 - <br><br> The antisense nucleic acids are preferably oligonucleotides and may consist entirely of deoxyribo-nucleotides, modified deoxyribonucleotides, or some combination of both. The antisense nucleic acids can be 5 synthetic oligonucleotides. The oligonucleotides may be chemically modified, if desired, to improve stability and/or selectivity. Since oligonucleotides are susceptible to degradation by intracellular nucleases, the modifications can include, for example, the use of a sulfur group to replace 10 the free oxygen of the phosphodiester bond. This modification is called a phosphorothioate linkage. Phosphorothioate antisense oligonucleotides are water soluble, polyanionic, and resistant to endogenous nucleases. In addition, when a phosphorothioate antisense 15 oligonucleotide hybridizes to its target site, the RNA-DNA duplex activates the endogenous enzyme ribonuclease (Rnase) H, which cleaves the mRNA component of the hybrid molecule. <br><br> In addition, antisense oligonucleotides with phosphoramidite and polyamide (peptide) linkages can be 20 synthesized. These molecules should be very resistant to nuclease degradation. Furthermore, chemical groups can be added to the 2• carbon of the sugar moiety and the 5 carbon (C-5) of pyrimidines to enhance stability and facilitate the binding of the antisense oligonucleotide to its target site. 25 Modifications may include 2' deoxy, 0-pentoxy, O-propoxy, O-methoxy, fluoro, methoxyethoxy phosphoro-thioates, modified bases, as well as other modifications known to those of skill in the art. <br><br> The antisense nucleic acids can also be DNA sequences 30 whose expression in the cell produces RNA complementary to all or part of the LIPG mRNA. Antisense nucleic acids can be prepared by expression of all or part of a sequence selected from the group consisting of SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 7, or SEQ ID No. 11, in the opposite orientation, as 35 described in EP 140308. Any length of antisense sequence is suitable for practice of the invention so long as it is capable of down-regulating or blocking expression of LIPG. <br><br> - 39 - <br><br> Preferably, the antisense sequence is at least 20 nucleotides in length. The preparation and use of antisense nucleic acids, DNA encoding antisense RNAs and the use of oligo and genetic antisense is disclosed in W092/15680, the contents of 5 which are incorporated herein by reference. <br><br> One approach to determining the optimum fragment of LIPG to use in an antisense nucleic acid treatment method involves preparing random fragments of LIPG cDNA by mechanical shearing, enzymatic treatment, and cloning the fragment into 10 any of the vector systems described herein. Individual clones or pools of clones are used to infect LIPG-expressing cells, and effective antisense LIPG cDNA fragments are identified by monitoring LIPG expression at the RNA or protein level. <br><br> 15 The retroviral, adeno-associated viral, and adenoviral vector systems discussed hereinabove may all be used to introduce and express antisense nucleic acids in cells. Antisense synthetic oligonucleotides may be introduced in a variety of ways, including the methods discussed hereinbelow. <br><br> 20 Ribozymes <br><br> Reductions in the levels of LIPG polypeptide may be accomplished using ribozymes. Ribozymes are catalytic RNA molecules (RNA enzymes) that have separate catalytic and substrate binding domains. The substrate binding sequence 25 combines by nucleotide complementarity and, possibly, <br><br> nonhydrogen bond interactions with its target sequence. The catalytic portion cleaves the target RNA at a specific site. The substrate domain of a ribozyme can be engineered to direct it to a specified mRNA sequence. The ribozyme 30 recognizes and then binds a target mRNA through complementary base-pairing. Once it is bound to the correct target site, the ribozyme acts enzymatically to cut the target mRNA. Cleavage of the LIPG mRNA by a ribozyme destroys its ability to direct synthesis of LIPG polypeptide. Once the ribozyme 35 has cleaved its target sequence, it is released and can repeatedly bind and cleave at other LIPG mRNAs. <br><br> - 40 - <br><br> In preferred embodiments of this invention, the ribozyme is formed in a hammerhead motif. Other forms include a hairpin motif, a hepatitis delta virus, group I intron or RnaseP RNA (in association with an RNA guide sequence) motif 5 or Neurospora VS RNA motif. Hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses, 8, 183. Hairpin motifs are described in Hampel and Tritz, 1989, Biochemistry, 28, 4929, and Hampel et al., 1990, <br><br> Nucleic Acids Res., 18, 299. The hepatitis delta virus motif 10 is described by Perrotta and Been, 1992, Biochemistry, 31, <br><br> 16, the RnaseP motif is described by Guerrier-Takada et al., 1983, Cell, 35, 849, the Neurospora VS RNA ribozyme motif is described by Collins (Saville and Collins, 1990, Cell, 61, 685-696; Saville and Collins, 1991, Proc. Natl. Acad. Sci. 15 USA, 88, 8826-8830; Collins and Olive, 1993, Biochemistry, 32, 2795-2799) the Group I intron motif is described by Cech et al., U.S. Patent No. 4,987,071. <br><br> One approach in preparing a ribozyme is to chemically synthesize an oligodeoxyribonucleotide with a ribozyme 20 catalytic domain (~20 nucleotides) flanked by sequences that hybridize to the target LIPG mRNA after transcription. The oligodeoxyribonucleotide is amplified by using the substrate binding sequences as primers. The amplification product is cloned into a eukaryotic expression vector. <br><br> 25 Ribozymes possessing a hammerhead or hairpin structure are readily prepared since these catalytic RNA molecules can be expressed within cells from eukaryotic promoters (e.g., Scanlon et al., 1991, Proc. Natl. Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; 30 Dropulic et al., 1992, J. Virol., 66, 1432-41; Weerasinghe et al., 1991, J. Virol., 65, 5531-4; Ojwang et al., 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Sarver et al., 1990, Science, 247, 1222-1225)). A ribozyme of the present invention can be 35 expressed in eukaryotic cells from the appropriate DNA <br><br> vector. If desired, the activity of the ribozyme may be augmented by its release from the primary transcript by a <br><br> - 41 - <br><br> second ribozyme (Ohkawa et al., 1992, Nucleic Acids Symp. Ser., 21, 15-6; Taira et al., 1991, Nucleic Acids Res., 19, 5125-30; Ventura et al., 1993, Nucleic Acids Res., 21, 3249-55) . <br><br> 5 In one approach to preparing ribozymes, ribozymes are expressed from transcription units inserted into DNA, RNA, or viral vectors. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol (I) , RNA polymerase II (pol II), or RNA polymerase III (pol 10 III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on nearby gene regulatory sequences. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA 15 polymerase enzyme is expressed in the appropriate cells <br><br> (Elroy-Stein and Moss, 1990, Proc. Natl. Acad. Sci. USA, 87, 6743-7; Gao and Huang 1993, Nucleic Acids Res., 21, 2867-72; Lieber et al., 1993, Methods Enzymol., 217, 47-66; Zhou et al., 1990, Mol. Cell. Biol., 10, 4529-37). It has been 20 demonstrated that ribozymes expressed from these promoters can function in mammalian cells (Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992, Proc. Natil. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992 Nucleic Acids Res., 20, 4581-9; Yu et al., 1993, Proc. Natl. Acad. 25 Sci. USA, 90, 6340-4; L'Huillier et al., 1992, EMBO J., 11, 4411-8; Lisziewicz et al., 1993, Proc. Natl. Acad. Sci. USA, 90, 8000-4). <br><br> In one embodiment of the present invention, a transcription unit expressing a ribozyme that cleaves LIPG 30 RNA is inserted into a plasmid DNA vector, a retrovirus vector, an adenovirus DNA viral vector or an adeno-associated virus vector. The recombinant vectors are preferably DNA plasmids or adenovirus vectors. However, other mammalian cell vectors that direct the expression of RNA may be used 35 for this purpose. The vectors are delivered as recombinant viral particles. DNA may be delivered alone or complexed with various vehicles. The DNA, DNA/vehicle complexes, or <br><br> o i j <br><br> - 42 - <br><br> the recombinant virus particles are locally administered to the site of treatment, as discussed below. Preferably, recombinant vectors capable of expressing the ribozymes are locally delivered as described below, and persist in target 5 cells. Once expressed, the ribozymes cleave the target LIPG mRNA. <br><br> Ribozymes may be administered to a patient by a variety of methods. They may be added directly to target tissues, complexed with cationic lipids, packaged within liposomes, or 10 delivered to target cells by other methods known in the art. Localized administration to the desired tissues may be done by catheter, infusion pump or stent, with or without incorporation of the ribozyme in biopolymers as discussed hereinbelow. Alternative routes of delivery include, but 15 are not limited to, intravenous injection, intramuscular injection, subcutaneous injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are 20 provided in Sullivan et al., PCT W094/02595 and Draper et al., PCT W093/23569, which are incorporated by reference herein. <br><br> Nop-Viral Delivery Systems <br><br> Certain non-viral systems have been used in the art and 25 can facilitate introduction of DNA encoding the LIPG <br><br> polypeptides or antisense nucleic acids into a patient. <br><br> A DNA vector encoding a desired LIPG polypeptide or antisense sequence can be introduced in vivo by lipofection. For the past decade, there has been increasing use of 30 liposomes for encapsulation and transfection of nucleic acids in vitro. Synthetic cationic lipids designed to limit the difficulties and dangers encountered with liposome mediated transfection can be used to prepare liposomes for in vivo transfection of a gene encoding a marker [Feigner, et. al., 35 Proc. Natl. Acad. Sci. U.S.A. 84:7413-7417 (1987); see <br><br> Mackey, et al., Proc. Natl. Acad. Sci. U.S.A. 85:8027-8031 <br><br> o <br><br> - 43 - <br><br> (1988); Ulmer et al., Science 259:1745-1748 (1993)]. The use of cationic lipids may promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes [Feigner and Ringold, 5 Nature 337:387-388 (1989)]. Particularly useful lipid compounds and compositions for transfer of nucleic acids are described in International Patent Publications W095/18863 and W096/17823, and in U.S. Patent No. 5,459,127. The use of lipofection to introduce exogenous genes into the specific 10 organs in vivo has certain practical advantages. Molecular targeting of liposomes to specific cells represents one area of benefit. It is clear that directing transfection to particular cell types would be particularly advantageous in a tissue with cellular heterogeneity, for example, pancreas, 15 liver, kidney, and the brain. Lipids may be chemically coupled to other molecules for the purpose of targeting [see Mackey, et. al., supra]. Targeted peptides, e.g., hormones or neurotransmitters, and proteins for example, antibodies, or non-peptide molecules could be coupled to liposomes 20 chemically. <br><br> Other molecules are also useful for facilitating transfection of a nucleic acid in vivo, for example, a cationic oligopeptide (e.g., International Patent Publication W095/21931), peptides derived from DNA binding proteins 25 (e.g., International Patent Publication W096/25508), or a cationic polymer (e.g., International Patent Publication W095/21931). <br><br> It is also possible to introduce A DNA vector encoding a LIPG polypeptide or antisense sequence in vivo as a naked DNA 30 plasmid (see U.S. Patents 5,693,622, 5,589,466 and <br><br> 5,580,859). Naked DNA vectors for gene therapy can be introduced into the desired host cells by methods known in the art, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate 35 precipitation, use of a gene gun, or use of a DNA vector transporter [see, e.g., Wilson et al., J. Biol. Chem. 267:963-967 (1992); Wu and Wu, J. Biol. Chem. 263:14621-14624 <br><br> o <br><br> - 44 - <br><br> (1988); Hartmut et al., Canadian Patent Application No. 2,012,311, filed March 15, 1990; Williams et al., Proc. Natl. Acad. Sci. USA 88:2726-2730 (1991)]. Receptor-mediated DNA delivery approaches can also be used [Curiel et al., Hum. <br><br> 5 Gene Ther. 3:147-154 (1992); Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)]. <br><br> Antibodies <br><br> The present invention provides antibodies against the LIPG polypeptide. These antibodies may be monoclonal 10 antibodies or polyclonal antibodies. The present invention includes chimeric, single chain, and humanized antibodies, as well as Fab fragments and the products of an Fab expression library, and Fv fragments and the products of an Fv expression library. <br><br> 15 Polyclonal antibodies may be prepared against an antigenic fragment of an LIPG polypeptide, as described in the Examples section hereinbelow. Antibodies may also be generated against the intact LIPG protein or polypeptide, or against a fragment, derivative, or epitope of the protein or 20 polypeptide. Antibodies may be obtained following the administration of the protein, polypeptide, fragment, derivative, or epitope to an animal, using the techniques and procedures known in the art. <br><br> Monoclonal antibodies may be prepared using the method 25 of Mishell, B. B., et al., Selected Methods In Cellular Immunology, (W.H. Freeman, ed.) San Francisco (1980). <br><br> Briefly, a polypeptide of the present invention is used to immunize spleen cells of Baib/C mice. The immunized spleen cells are fused with myeloma cells. Fused cells containing 30 spleen and myeloma cell characteristics are isolated by growth in HAT medium, a medium which kills both parental cells, but allows the fused products to survive and grow. <br><br> The monoclonal antibodies of the present invention may be "humanized" to prevent the host from mounting an immune 35 response to the antibodies. A "humanized antibody" is one in <br><br> - 45 - <br><br> which the complementarity determining regions (CDRs) and/or other portions of the light and/or heavy variable domain framework are derived from a non-human immunoglobulin, but the remaining portions of the molecule are derived from one 5 or more human immunoglobulins. Humanized antibodies also include antibodies characterized by a humanized heavy chain associated with a donor or acceptor unmodified light chain or a chimeric light chain, or vice versa. The humanization of antibodies may be accomplished by methods known in the art 10 (see, e.g. G.E. Mark and E.A. Padlan, "Chapter 4. <br><br> Humanization of Monoclonal Antibodies", The Handbook of Experimental Pharmacology Vol. 113, Springer-Verlag, New York, 1994). Transgenic animals may be used to express humanized antibodies. <br><br> 15 Techniques known in the art for the production of single chain antibodies can be adapted to produce single chain antibodies to the immunogenic polypeptides and proteins of the present invention. <br><br> In a preferred embodiment, an anti-LIPG antibody is used 20 to bind to and inhibit the enzymatic activity of LIPG in a patient. <br><br> The anti-LIPG antibodies are also useful in assays for detecting or quantitating levels of LIPG. In one embodiment, these assays provide a clinical diagnosis and assessment of 25 LIPG in various disease states and a method for monitoring treatment efficacy. These anti-LIPG antibodies may additionally be used to quantitate LIPG in a tissue sample in order to predict further susceptibility to lowered levels of HDL cholesterol and apolipoprotein AI. <br><br> 30 Methods of Identifying and Utilizing <br><br> Inhibitory Molecules and Enhancer Molecules <br><br> The present invention provides methods of screening small molecule libraries or natural product sources for enhancers (agonists) or co-activators including proteinaceous 35 co-activators or inhibitors (antagonists) of LIPG activity. <br><br> o <br><br> - 46 - <br><br> A potential enhancer or inhibitor is contacted with LIPG protein and a substrate of LIPG, and the ability of the potential enhancer or inhibitor to enhance or inhibit LIPG activity is measured. <br><br> 5 These screening methods may also be used to determine if a compound can function as a substrate specific enhancer or inhibitor, that is, whether a compound can enhance the enzymatic activity of LIPG toward one substrate while lowering or maintaining a given level of enzymatic activity 10 for a different substrate, for example, the LIPG polypeptide of the present invention utilizes HDL cholesterol as a substrate and also utilizes LDL cholesterol and VLDL cholesterol as substrates. In certain embodiments, it is desirable to isolate and identify substrate specific 15 enhancers or inhibitors which enhance the enzymatic activity of the LIPG polypeptide towards LDL cholesterol or VLDL cholesterol while lowering or maintaining the normal level of enzymatic activity for HDL cholesterol. <br><br> The LIPG protein used in these methods can be produced 20 recombinantly in a variety of host cells, including mammalian cells, baculovirus-infected insect cells, yeast, and bacteria. LIPG expression in stably transfected CHO cells can be optimized by methotrexate amplification of the cells. LIPG protein can also be purified from natural sources such 25 as human plasma, placental extracts, or conditioned media from cultured endothelial cells, THP-1 cells, or macrophages. <br><br> The optimization of assay parameters including pH, ion concentrations, temperature, concentration of substrate, and emulsification conditions are determined empirically by one 30 having ordinary skill in the art. <br><br> The fatty acid substituents of the substrates may vary in chain length as well as in degree and position of unsaturation. The substrates may be radiolabelled in any of several positions. Phospholipid substrates such as 35 phosphatidylcholine can be radiolabelled, for example, in the <br><br> o o <br><br> - 47 - <br><br> Sn-1 or Sn-2 fatty acid position, or in the glycerol, phosphate, or polar head group (choline in the case of phosphatidylcholine). <br><br> As an alternative to radiolabeled substrates, other 5 classes of labeled substrates, such as fluorescent substrates or thio-containing substrates, can also be used in the screening methods. <br><br> Fluorescent substrates are particularly useful in screening assays because enzymatic catalysis can be measured 10 continuously by measuring fluorescence intensity, without the physical separation (extraction) of the products from the substrates. An example of a fluorescent phosphatidylcholine substrate is C6NBD-PC{l-acyl-2-[6-(nitro-2,l,3-benzoxadiazol-4-yl)amino] caproylphosphatidylcholine. <br><br> 15 The thio-containing substrates include 1,2- <br><br> bis(hexanoylthio)-1,2-dideoxy-sn-glycero-3-phosphorylcholine (L.J. Reynolds, W.N. Washburn, R.A. Deems, and E.A. Dennis, 1991. Methods in Enzymology 197: 3-23; L. Yu and E.A. <br><br> Dennis, 1991. Methods in Enzymology 197: 65-75; L.A. 20 Wittenauer, K, Shirai, R.L. Jackson, and J.D. Johnson, 1984. Biochem. Biophys. Res. Commun. 118: 894-901). <br><br> In addition to inhibitory and enhancer molecules which operate at the level of enzymatic activity, there are inhibitory and enhancer molecules which operate at the level 25 of expression of the LIPG gene. One method for identifying compounds which are able to enhance or inhibit the expression of LIPG is to use a reporter gene system. These systems utilize reporter gene expression vectors which include a cloning site into which a given promoter may be cloned 30 upstream of a "reporter gene" which can be easily detected and quantified. One of skill in the art could readily identify and subclone the promoter for the LIPG gene as well as other control sequences into a commercially available reporter gene expression vector. The expression vector is 35 transferred into host cells and the cells are exposed to a <br><br> o o <br><br> - 48 - <br><br> test compound (a putative inhibitor or enhancer molecule) to determine the effect of the test compound on expression of the reporter gene product. In particular, the cells are assayed for the presence of the reporter gene product by 5 directly measuring the amount of reporter mRNA, the reporter protein itself or the enzymatic activity of the reporter protein. Ideally, the reporter gene is not endogenously expressed in the cell type of interest and lends itself to sensitive, quantitative and rapid assays. A variety of 10 reporter assay constructs are commercially available and several reporter genes and assays have been developed and can be readily prepared by those of skill in the art. The most popular systems for monitoring genetic activity in eukaryotic cells include the chloramphenicol acetyltransferase (CAT) , 3-15 galactosidase, firefly luciferase, growth hormone (GH) , (5-glucurorudase (GUS), alkaline phosphatase (AP), green fluorescent protein (GFP) and Renilla luciferase. Reporter assay constructs can be purchased from a variety of sources including Promega and Invitrogen. <br><br> 20 As mentioned above, reporter gene activity can be detected by assaying for the reporter mRNA or the reporter protein. The reporter mRNA can be detected by northern blot analysis, ribonuclease protection assays or RT-PCR. While these assays are more direct than measuring protein 25 expression, many assays have been developed to measure the ' presence of the reporter protein rather than the mRNA present in a cell. Reporter proteins can be assayed by spectrophotometry or by detecting enzymatic activity. <br><br> Reporter protein levels may also be measured with antibody-30 based assays. In general, the enzymatic assays are very sensitive and are a preferred method of monitoring reporter gene expression. <br><br> Compositions <br><br> The present invention provides compositions in a 35 biologically compatible (biocompatible) solution, comprising the polypeptides, nucleic acids, vectors, and antibodies of the invention. A biologically compatible solution is a <br><br> - 49 - <br><br> solution in which the polypeptide, nucleic acid, vector, or antibody of the invention is maintained in an active form, e.g., in a form able to effect a biological activity. For example, a polypeptide of the invention would have 5 phospholipase activity; a nucleic acid would be able to replicate, translate a message, or hybridize to a complementary nucleic acid; a vector would be able to transfect a target cell; an antibody would bind a polypeptide of the invention. Generally, such a biologically compatible 10 solution will be an agueous buffer, e.g., Tris, phosphate, or HEPES buffer, containing salt ions. Usually the concentration of salt ions will be similar to physiological levels. In a specific embodiment, the biocompatible solution is a pharmaceutically acceptable composition. Biologically 15 compatible solutions may include stabilizing agents and preservatives. <br><br> Such compositions can be formulated for administration by topical, oral, parenteral, intranasal, subcutaneous, and intraocular, routes. Parenteral administration is meant to 20 include intravenous injection, intramuscular injection, intraarterial injection or infusion techniques. The composition may be administered parenterally in dosage unit formulations containing standard, well known nontoxic physiologically acceptable carriers, adjuvants and vehicles <br><br> 25 as desired. <br><br> &gt; <br><br> The preferred sterile injectable preparations can be a solution or suspension in a nontoxic parenterally acceptable solvent or diluent. Examples of pharmaceutically acceptable carriers are saline, buffered saline, isotonic saline (e.g. 30 monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride, or mixtures of such salts), Ringer's solution, dextrose, water, sterile water, glycerol, ethanol, and combinations thereof. 1,3-butanediol and sterile fixed oils are conveniently employed as solvents or suspending 35 media. Any bland fixed oil can be employed including synthetic mono- or di-glycerides. Fatty acids such as oleic acid also find use in the preparation of injectables. <br><br> o <br><br> - 50 - <br><br> o <br><br> The composition medium can also be a hydrogel which is prepared from any biocompatible or non-cytotoxic (homo or hetero) polymer, such as a hydrophilic polyacrylic acid polymer that can act as a drug absorbing sponge. Such 5 polymers have been described, for example, in application W093/08845, the entire contents of which are hereby incorporated by reference. Certain of them, such as, in particular, those obtained from ethylene and/or propylene oxide are commercially available. A hydrogel can be 10 deposited directly onto the surface of the tissue to be treated, for example during surgical intervention. <br><br> Another preferred embodiment of the present invention relates to a pharmaceutical composition comprising a replication defective recombinant virus and poloxamer. More 15 specifically, the invention relates to a composition comprising a replication defective recombinant virus comprising a nucleic acid encoding an LIPG polypeptide and poloxamer. A preferred poloxamer is Poloxamer 407, which is commercially available (BASF, Parsippany, NJ) and is a non-20 toxic, biocompatible polyol, and is most preferred. A poloxamer impregnated with recombinant viruses may be deposited directly on the surface of the tissue to be treated, for example during a surgical intervention. Poloxamer possesses essentially the same advantages as <br><br> 25 hydrogel while having a lower viscosity. <br><br> &gt; <br><br> Methods of Treatment <br><br> The present invention provides methods of treatment which comprise the administration to a human or other animal of an effective amount of a composition of the invention. <br><br> 30 Effective amounts may vary, depending on the age, type and severity of the condition to be treated, body weight, desired duration of treatment, method of administration, and other parameters. Effective amounts are determined by a physician or other qualified medical professional. In most 35 cases, the dosage levels may be adjusted so that the desired levels of HDL cholesterol and apolipoprotein AI are achieved <br><br> o <br><br> - 51 - <br><br> and maintained. Similarly, the dosage levels may be adjusted to lower the VLDL cholesterol and LDL cholesterol levels to acceptable levels and bring the ratio HDL cholesterol to LDL cholesterol and VLDL cholesterol to within desirable levels. <br><br> 5 Polypeptides according to the invention are generally administered in doses of about 0.01 mg/kg to about 100 mg/kg, preferably about 0.1 mg/kg to about 50 mg/kg, and most preferably about 1 mg/kg to about 10 mg/kg of body weight per day. <br><br> 10 Neutralizing antibodies according to the invention may be delivered as a bolus only, infused over time or both administered as a bolus and infused over time. Although the dosage amount will vary based on the parameters above, and on the binding ability of the antibody, a dose 0.2 to 0.6 mg/kg 15 may be given as a bolus followed by a 2 to 12 hour infusion period. Alternatively, multiple bolus injections are administered every other day or every third or fourth day as needed. Dosage levels may be adjusted as determined by HDL cholesterol levels and/or VLDL and LDL cholesterol levels. <br><br> 20 As discussed hereinabove, recombinant viruses may be used to introduce both DNA encoding LIPG and subfragments of LIPG as well as antisense nucleic acids. Recombinant viruses according to the invention are generally formulated and administered in the form of doses of between about 104 and 25 about 1014 pfu. In the case of AAVs and adenoviruses, doses of from about 10° to about 1011 pfu are preferably used. The term pfu ("plaque-forming unit") corresponds to the infective power of a suspension of virions and is determined by infecting an appropriate cell culture and measuring the 30 number of plaques formed. The techniques for determining the pfu titre of a viral solution are well documented in the prior art. <br><br> 35 <br><br> Ribozymes according to the present invention may be administered in amounts ranging from about 5 to about 50 mg/kg/day in a pharmaceutically acceptable carrier. Dosage <br><br> o <br><br> - 52 - <br><br> levels may be adjusted based on the measured therapeutic efficacy. <br><br> Appropriate levels of inhibitor or enhancer molecules may be determined by qualified medical personnel using the 5 parameters discussed above. <br><br> The present invention provides compositions and methods for increasing the level of HDL cholesterol and apolipoprotein AI and lowering the levels of VLDL and LDL cholesterol in a patient. The present invention further 10 provides methods of treating a human or other animal having an undesirable lipid profile, wherein said undesirable lipid profile is the result of abnormally high expression of LIPG polypeptide activity. <br><br> Methods and Compositions for 15 Lowering Levels of LIPG Polypeptide Activity <br><br> The methods for decreasing the expression of LIPG polypeptide in order to increase the levels of HDL cholesterol and apolipoprotein AI and correct those conditions in which LIPG polypeptide activity contributes to 20 a disease or disorder associated with an undesirable lipid profile include but are not limited to administration of a composition comprising an antisense nucleic acid, administration of a composition comprising an intracellular binding protein such as an antibody, administration of an 25 inhibitory molecule which inhibits the enzymatic activity of LIPG, for example, a composition comprising an expression vector encoding a subfragment of LIPG, for example, LLGN polypeptide or a small molecular weight molecule, including administration of a small molecular weight compound which 30 down regulates LIPG expression at the level of transcription, translation or post-translation, and administration of a ribozyme which cleaves mRNA encoding LIPG. <br><br> Methods Utilizing Antisense Nucleic Acids <br><br> In one embodiment, a composition comprising an antisense 35 nucleic acid is used to down-regulate or block the expression <br><br> - 53 - <br><br> of LIPG. In one preferred embodiment, the nucleic acid encodes antisense RNA molecules. In this embodiment, the nucleic acid is operably linked to signals enabling expression of the nucleic acid sequence and is introduced 5 into a cell utilizing, preferably, recombinant vector constructs, which will express the antisense nucleic acid once the vector is introduced into the cell. Examples of suitable vectors includes plasmids, adenoviruses, adeno-associated viruses, retroviruses, and herpes viruses. 10 Preferably, the vector is an adenovirus. Most preferably, <br><br> the vector is a replication defective adenovirus comprising a deletion in the El and/or E3 regions of the virus. <br><br> In another embodiment, the antisense nucleic acid is synthesized and may be chemically modified to resist 15 degradation by intracellular nucleases, as discussed above. Synthetic antisense oligonucleotides can be introduced to a cell using liposomes. Cellular uptake occurs when an antisense oligonucleotide is encapsulated within a liposome. With an effective delivery system, low, non-toxic 20 concentrations of the antisense molecule can be used to inhibit translation of the target mRNA. Moreover, liposomes that are conjugated with cell-specific binding sites direct an antisense oligonucleotide to a particular tissue. <br><br> Methods Utilizing Neutralizing 25 Antibodies and Other Binding Proteins <br><br> In another embodiment, the expression of LIPG is down-regulated or blocked by the expression of a nucleic acid sequence encoding an intracellular binding protein which is capable of selectively interacting with LIPGv WO 94/29446 30 and WO 94/02610, the contents of which are incorporated herein by reference, disclose cellular transfection with genes encoding an intracellular binding protein. An intracellular binding protein includes any protein capable of selectively interacting, or binding, with LIPG in the cell in 35 which it is expressed and of neutralizing the function of bound LLG. Preferably, the intracellular binding protein is a neutralizing antibody or a fragment of a neutralizing <br><br> O o <br><br> - 54 - <br><br> antibody. More preferably, the intracellular binding protein is a single chain antibody. <br><br> WO 94/02610 discloses preparation of antibodies and identification of the nucleic acid encoding a particular 5 antibody. Using LIPG or a fragment thereof, a specific monoclonal antibody is prepared by techniques known to those skilled in the art. A vector comprising the nucleic acid encoding an intracellular binding protein, or a portion thereof, and capable of expression in a host cell is 10 subsequently prepared for use in the method of this invention. <br><br> Alternatively, LIPG activity can be blocked by administration of a neutralizing antibody into the circulation. Such a neutralizing antibody can be 15 administered directly as a protein, or it can be expressed from a vector (with a secretory signal). <br><br> Methods Utilizing an Inhibitory Molecule which Inhibits the Bnzvmatic Activity of LIPG <br><br> In another embodiment, LIPG activity is inhibited by the 20 administration of a composition comprising a subfragment of LIPG polypeptide, for example, LLGN. This composition may be administered in a convenient manner, such as by the oral, topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, or intradermal routes. The 25 composition may be administered directly or it may be encapsulated (e.g. in a lipid system, in amino acid microspheres, or in globular dendrimers). The polypeptide may, in some cases, be attached to another polymer such as serum albumin or polyvinyl pyrrolidone. <br><br> 30 In another embodiment, LIPG activity is inhibited through the use of gene therapy, that is, through the administration of a composition comprising a nucleic acid which encodes and directs the expression of a subfragment of LIPG, for example, LLGN. <br><br> o <br><br> - 55 - <br><br> In another embodiment, LIPG activity is inhibited through the use of inhibitory molecules. These low molecular weight compounds interfere with LIPG's enzymatic properties or prevent its appropriate recognition by cellular binding 5 sites. <br><br> In a specific embodiment, the LIPG polypeptide of the present invention also has an affinity for heparin. LIPG polypeptide binding to extracellular heparin in the lumen of blood vessels would permit LIPG to bind to and accelerate LDL 10 uptake by acting as a bridge between LDL and the extracellular heparin. In the localized area of an atherosclerotic lesion, an increased level of lipase activity is hypothesized to accelerate the atherogenic process (Zilversmit, D.B. (1995) Clin. Chem. 41,153-158; Zambon, 15 A., Torres, A., Bijvoet, S., Gagne, C., Moojani, S., Lupien, P.J., Hayden M.R., and Brunzell, J.D. (1993) Lancet 341, 1119-1121) . This may be due to an increase in the binding and uptake of lipoproteins by vascular tissue mediated by lipases (Eisenberg, S., Sehayek, E., Olivecrona, T. 20 Vlodavsky, I. (1992) J. Clin. Invest. 90,2013-2021; Tabas, I., Li, I., Brocia R.W., Xu, S.W., Swenson T.L. Williams, K.J. (1993) J.Biol. Chem. 268,20419-20432; Nordestgaard, B.G., and Nielsen, A.G. (1994) Curr. Opin. Lipid. 5,252-257; Williams, K.J., and Tabas, I. (1995) Art. Thromb. and Vase. 25 Biol. 15,551-561). Additionally, a high local level of lipase activity may result in cytotoxic levels of fatty acids and lysophosphatidylcholine being produced in precursors of atherosclerotic lesions. This particular activity of LLG may contribute to the development or progression of 30 atherosclerosis, particularly in the context of excessive lipid levels in a subject due to dietary or genetic factors. Thus, the present invention permits inhibition of lipoprotein accumulation by inhibiting LIPG polypeptide expression or binding to lipoprotein (e.g., LDL). <br><br> o <br><br> (.) <br><br> - 56 - <br><br> Methods Utilizing an Inhibitory <br><br> Molecule which Prevents LIPG Gene Expression <br><br> In another embodiment, inhibitory molecules, including small molecular weight compounds, are able to down regulate 5 LIPG expression at the level of transcription, translation or post-translation. In order to identify such inhibitory molecules, the reporter gene systems described above may be used. These inhibitory molecules may be combined with a pharmaceutically acceptable carrier and administered using 10 conventional methods known in the art. <br><br> Methods Utilizing Ribozymes <br><br> Ribozymes may be administered to cells by encapsulation in liposomes, by iontophoresis, by incorporation into hydrogels, cyclodextrins, biodegradable nanocapsules, and 15 bioadhesive microspheres or by any of a variety of other methods dicussed above. The ribozyme may be delivered to a target tissue by direct injection or by use of a catheter, infusion pump or stent. Alternative routes of delivery include intravenous injection, intramuscular injection, 20 subcutaneous injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. <br><br> In preferred embodiments, a ribozyme-encoding sequence is cloned into a DNA expression vector. Transcription of the 25 ribozyme sequence is driven from an eukaryotic RNA polymerase II (pol II), or RNA polymerase III (pol III) promoter. The expression vector can be incorporated into a variety of vectors including the viral DNA vectors such as adenovirus or adeno-associated virus vectors discussed above. <br><br> 30 In a preferred embodiment of the invention, a transcription unit expressing a ribozyme that cleaves LIPG RNA is inserted into an adenovirus DNA viral vector. The vector is delivered as recombinant viral particles and is locally administered to the site of treatment, through the 35 use of a catheter, stent or infusion pump. <br><br> - 57 - <br><br> Administration of Apolipoprotein AI <br><br> In another embodiment, any of the methods discussed above for lowering the levels of LIPG polypeptide activity are utilized in combination with administration of 5 apolipoprotein AI or an expression system capable of expressing apolipoprotein AI in a patient (see, for example, U.S. Patent No. 5,866,551, which is incorporated herein by reference), <br><br> Methods and compositions for 10 Increasing Levels of LIPG Polypeptide Activity <br><br> The methods for increasing the expression or activity of LIPG polypeptide to lower the levels of VLDL and LDL cholesterol include, but are not limited to, administration of a composition comprising the LIPG polypeptide, 15 administration of a composition comprising an expression vector which encodes the LIPG polypeptide, administration of a composition comprising an enhancer molecule which enhances the enzymatic activity of the LIPG polypeptide and administration of an enhancer molecule which increases 20 expression of the LIPG gene. <br><br> Methods Utilizing LIPG Polypeptides <br><br> In one embodiment, the level of LIPG activity is increased through the administration of a composition comprising the LIPG polypeptide. This composition may be 25 administered in a convenient manner, such as by the oral, topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, or intradermal routes. The composition may be administered directly or it may be encapsulated (e.g. in a lipid system, in amino acid 30 microspheres, or in globular dendrimers) . The polypeptide may, in some cases, be attached to another polymer such as serum albumin or polyvinyl pyrrolidone. <br><br> Methods Utilizing Vectors that Express LIPG <br><br> In another embodiment, the level of LIPG is increased 35 through the use of gene therapy, that is, through the • <br><br> administration of composition comprising a nucleic acid which <br><br> o <br><br> - 58 - <br><br> encodes and directs the expression of the LIPG polypeptide. In this embodiment, the LIPG polypeptide is cloned into an appropriate expression vector. Possible vector systems and promoters are extensively discussed above. The expression 5 vector is transferred into the target tissue using one of the vector delivery systems discussed above. This transfer is carried out either ex vivo in a procedure in which the nucleic acid is transferred to cells in the laboratory and the modified cells are then administered to the human or 10 other animal, or in vivo in a procedure in which the nucleic acid is transferred directly to cells within the human or other animal. In preferred embodiments, an adenoviral vector system is used to deliver the expression vector. If desired, a tissue specific promoter is utilized in the expression 15 vector as described above. <br><br> Non-viral vectors may be transferred into cells using any of the methods known in the art, including calcium phosphate coprecipitation, lipofection (synthetic anionic and cationic liposomes), receptor-mediated gene delivery, naked 20 DNA injection, electroporation and bioballistic or particle acceleration. <br><br> Methods Utilizing an Enhancer Molecule which Enhances the Enzymatic Activity of LIPG <br><br> In another embodiment, the activity of LIPG is enhanced 25 by enhancer molecules that increase the enzymatic activity of LIPG or increase its appropriate recognition by cellular binding sites. These enhancer molecules may be introduced by the same methods discussed above for the administration of polypeptides. <br><br> 30 Methods Utilizing an Enhancer <br><br> Molecule which Increases LIPG Gene Expression <br><br> In another embodiment, the level of LIPG is increased through the use of small molecular weight compounds, which can upregulate LIPG expression at the level of transcription, 35 translation, or post-translation. These compounds may be <br><br> 59 <br><br> administered by the same methods discussed above for the administration of polypeptides. <br><br> 5 <br><br> Treatment Methods Relating to Impaired Biliary Excretion <br><br> Intrahepatic cholestasis can be characterized by increased serum cholesterol and phospholipid levels. A recently described, phalloidin drug-induced intrahepatic cholestasis model in rats demonstrated significant increases in the serum levels of cholesterol and phospholipid 10 (Ishizaki, K., Kinbara, S., Miyazawa, N. , Takeuchi, Y., Hirabayashi, N., Kasai, H., and Araki, T. (1997) Toxicol. Letters 90, 29-34). The products of this invention may be used to treat intrahepatic cholestasis in patients that have increased serum cholesterol and/or phospholipid. In 15 addition, this rat model also exhibited a severe decrease in biliary cholesterol excretion rates. The LIPG polypeptide and nucleic acid products of this invention may be used to treat patients with an impaired,biliary excretion system. <br><br> 20 impaired bile flow from the liver. Recently, the loci for progressive familial intrahepatic cholestasis (PFIC or Byler disease) and benign recurrent intrahepatic cholestasis (BRIC) were mapped to 18q21-q22 (Carlton, V.E.H., Knisely, A.S., and Freimer, N.B. (1995) Hum. Mol. Genet. 4, 1049-1053 and 25 Houwen, R.H., Baharloo, S., Blankenship, K., Raeymaekers, P., Juyn, J., Sandkuijl, L.A., and Freimer, N.B. (1994) Nature Genet. 8, 380-386, respectively). As LLG gene maps within this chromosomal region at 18q21, the LLG gene or products of this invention may be used to treat patients with 30 intrahepatic cholestasis that is caused by a mutation or defective expression of the PFIC/BRIC disease gene(s). <br><br> In another embodiment, the LLG gene or polypeptide products of this invention may be used to treat patients with intrahepatic cholestasis that is not due to a defect in the 35 PFIC/BRIC disease gene(s) at 18q21-q22. A recent study suggested that another locus, located outside of the 18q21- <br><br> Intrahepatic cholestasis is also characterized by <br><br> o <br><br> - 60 - <br><br> q22 region may also produce the PFIC phenotype (Strautnieks, S.S., Kagalwalla, A.F., Tanner, M.S., Gardiner, R.M., and Thompson, R.J. (1996) J. Med. Genet. 33, 833-836). Nevertheless, administration of LLG polypeptide, either 5 directly or via gene therapy, may alleviate this form of the condition. <br><br> Methods and Compositions for <br><br> Diagnosing a Predisposition to Low HDL Levels <br><br> Given the ability of LIPG polypeptide to lower the 10 levels of HDL cholesterol and apolipoprotein AI, the level of LIPG polypeptide in the body may be used to determine whether an individual is predisposed to low levels of HDL cholesterol and apolipoprotein AI. In this method, a tissue sample is taken from the patient. The tissue may be blood or one of 15 the tissues which has been demonstrated to express LIPG as discussed in the Examples section. Measurement of the level of LIPG may be performed by a variety of methods known to those of skill in the art. In preferred embodiments, an antibody directed against LIPG polypeptide may be used to 20 measure the level of LIPG in a tissue sample. <br><br> EXAMPLES <br><br> The following examples illustrate the invention. These examples are illustrative only, and do not limit the scope of the invention. <br><br> 25 EXAMPLE 1 - Identification of a Differentially Expressed cDNA <br><br> RNA Preparation <br><br> Human monocytic THP-1 cells (Smith, P.K., Krohn, R.I., Hermanson, G.T., Mallia, A.K., Gartner, F.H. Provenzano, M.D., Fujimoto, E.K., Goeke, H.M., Olson, B.J., and Klenk, 30 D.C. (1985) Anal. Biochem. 150,76-85) were cultured in RPMI-1640 medium (GIBCO) with 25 mM HEPES, 10% fetal bovine serum, 100 units/ml penicillin G sodium and 100 units/ml streptomycin sulfate. Cells were plated onto 15 cm tissue culture dishes at 1.5xl07 cells/plate, and treated with 40 35 ng/ml phorbol 12-myristate 13-acetate (Sigma) for 48 hours to induce differentiation of the cells. Human low density <br><br> o o <br><br> t <br><br> - 61 - <br><br> lipoproteins (LDL) were purchased from Calbiochem, and were dialyzed exhaustively versus PBS at 4°C. The LDL was then diluted to 500/ig/ml and dialyzed versus 5/zM CuS04 in PBS at 37°C for 16 hours. To stop oxidation, the LDL was dialyzed 5 exhaustively versus 150 mM NaCl, 0.3 mM EDTA, then filter sterilized. Protein concentration was determined by the BCA method (Schuh, J. Fairclough, G.F., and Haschemeyer, R.H. (1978) Proc. Natl. Acad. Sci. USA 75, 3173-3177) (Pierce). The degree of oxidation was determined by TBARS (Chomczynski, 10 P. (1993) Biotechniques 15,532-53.7), and was between 25-30 nmol MDA eguivalents/mg protein. The differentiated THP-1 cells were exposed for 24 hours to either 50 (ig/ml oxidized LDL or NaCl—EDTA buffer in RPMI medium with 10% lipoprotein-deficient fetal bovine serum (Sigma). To harvest the RNA, 15 the plates were rinsed with 10 ml of PBS, then 14 ml of <br><br> TRIZOL (Liang, P. and Pardee, A.B. (1992) Science 257,967-971) (GIBCO) were added to each plate. The solution was pipetted several times to mix, then like samples were pooled into centrifuge tubes and 3 ml chloroform per plate were 20 added and mixed. The tubes were centrifuged for 15 minutes at 12000 x g. After centrifugation the upper layer was transferred to a new tube and 7.5 ml isopropanol per plate was added and mixed. The tubes were centrifuged at 12000 x g for 20 minutes. The pellet was rinsed with ice-cold 70% 25 ethanol and dried at room temperature. The pellets were suspended in 500 til TE (Tris-EDTA) and treated with 200 units RNase-free DNAse I and 200 units RNasin placental RNase inhibitor (Promega) for 30 minutes at 37°C. The RNA was purified by sequential extractions with phenol, 30 phenol/chloroform/isoamyl alcohol (25:24:1), and chloroform/isoamyl alcohol (24:1) followed by ethanol precipitation. <br><br> CDMA Synthesis cDNA synthesis and PCR amplification were accomplished 35 using protocols from the Differential Display Kit, version 1.0 (Display Systems Biotechnology, Inc.) This system is based on the technique originally described by Liang and Pardee (Mead, D.A., Pey, N.K., Herrnstadt, C., Marcil, R.A., <br><br> o <br><br> - 62 - <br><br> and Smith, L.M., (1991) Bio/Technology 9,657-663). The primer pairs which yielded the cDNA fragment containing the first information of the lipase like gene were downstream primer 7 and upstream primer 15. The cDNA for the 5 amplification was synthesized as follows, using RNA derived from PMA treated THP-1 cells exposed to either buffer or oxidized LDL: 3 (i 1 of 25 n$l downstream primer 7 and 7.5 nl of diethylpyrocarbonate (DEPC)-treated water were added to 300 ng (3.0 p.1) RNA from either sample of THP-1 RNA. This 10 was heated to 70°C for 10 minutes then chilled on ice. To this tube were added 3 ^tl of 5x PCR buffer (250 mM Tris-HCl pH 8.3, 375 mM KCl) (GIBCO) , 3 [il 25 mM MgCl2, 3 ^1 0.1M DTT, <br><br> 1.2 |il 500 MM dNTPs, 0.7 j.il RNasin, and 5.6 DEPC-treated water. The tubes were incubated for 2 minutes at room 15 temperature, after which 1.5 |il (300 units) Superscript II RNase H- reverse transcriptase (GIBCO) were added. The tubes were incubated sequentially at room temperature for 2 minutes, 60 minutes at 37°C, and 5 minutes at 95°C, followed by chilling on ice. PCR amplification was performed using a <br><br> 20 master mix containing 117 ^1 lOx PCR buffer (500 mM KCl, 100 mM Tris-HCl pH 8.3, 15 mM MgCl2, and 0.01% (w/v) gelatin), 70.2 Hi 25 mM MgCl2, 5.9 |_il alpha-33P dATP (10m Ci/ml, DuPont NEN), 4.7 [Ll 500 |iM dNTP mix, 11 }.il AmpliTaq DNA polymerase (5 units/nl, Perkin-Elmer) , and 493.3 |il DEPC-treated water. 25 For each reaction, 12 {il of the master mix was added to 2 nl downstream primer #7, 1 nl of cDNA, and 5 nl of upstream primer #15. The reaction mixes were heated to 94°C for 1 minute, then thermocycled 40 times with a denaturing step of 94°C for 15 seconds, annealing step of 40°C for 1 minute, and 30 an extension step of 72°c for 30 seconds. Following the 40 cycles, the reactions were incubated at 72°C for 5 minutes and stored at 10°C. The PCR reactions were performed in a Perkin-Elmer GeneAmp System 9600 thermocycler. <br><br> Four microliters of the amplification reaction were 35 mixed with an equal volume of loading buffer (0.2% <br><br> o <br><br> - 63 - <br><br> bromphenol blue, 0.2% Xylene cyanol, 10 mM EDTA pH 8.0, and 20% glycerol). Four microliters of this 'mix was run on a 6% nondenaturing acrylamide sequencing format gel for 3 hours at 1200 volts (constant voltage). The gel was dried at 80°C 5 for 1.5 hours and exposed to Kodak XAR film. An amplification product found only in the reaction containing cDNA from THP-1 cells exposed to oxidized LDL was identified and excised from the gel. 100 pi of DEPC-treated water was added to a microcentrifuge tube containing the excised gel 10 fragment and was incubated for 30 minutes at room temperature followed by 15 minutes at 95°C. <br><br> To reamplify the PCR product, 26.5 microliters of the eluted DNA were used in a amplification reaction that also included 5 |il lOx PCR buffer, 3 |il 25 mM MgCl2, 5 pi 500 {XM 15 dNTPs, 5 |il 2(iM downstream primer 7, 7.5 p.! upstream primer 15, and 0.5 J.Q Amplitag polymerase. The PCR cycling parameters and instrument were as described above. <br><br> Following amplification, 20 pi of the reamplification was analyzed on an agarose gel and 4 pi was used to subclone the 20 PCR products into the vector pCRII using the TA cloning system (Frohman, M.A., Dush, M.K., and Martin, G.R. (1988) Proc. Natl. Acad. Sci. USA 85,8998-9002) (Invitrogen) . Following an overnight ligation at 14°C, the ligation products were used to transform E. coli. Resulting 25 transformants were picked and 3 ml overnight cultures were used in plasmid minipreparations. Insert sizes were determined using EcoRI digestions of the plasmids and clones containing inserts of the approximate size of the original PCR product were sequenced using fluorecent dye-terminator 30 reagents (Prism, Applied Biosystems) and an Applied <br><br> Biosystems 373 DNA sequencer. The sequence of the PCR product is shown in Figure 2. The sequence of the amplification primers is underlined. <br><br> 5'RACE Reaction <br><br> 35 Extension of the cDNA identified through RT-PCR was accomplished using the 5'RACE system (Loh, E.Y., Eliot, <br><br> o o <br><br> - 64 - <br><br> J.F., Cwirla, S., Lanier, L.L., and Davis, M.M. (1989) Science 243, 217-219; Simms, D., Guan, N., and Sitaraman, K., (1991) Focus 13, 99-100) (GIBCO). One microgram of the THP-1 RNA (oxidized LDL treated) used initially in the 5 differential display reactions was utilized in the 5'RACE procedure: <br><br> lpl (1 pg) of RNA was combined with 3 pi (3 pmol) primer 2 a and 11 |il DEPC-treated water and heated to 70°C for 10 minutes followed by 1 minute on ice. 2.5 |il l°x reaction 10 buffer (200 mM Tris-HCl pH 8.4, 500 mM KCl), 3 pi 25 mM <br><br> MgCl2, 1 pi 10 mM dNTP mix, and 2.5 |il 0.1 M DTT were added. The mix was incubated at 42°C for 2 minutes, then 1 p.1 Superscript II reverse transcriptase was added. The reaction was incubated for an additional 30 minutes at 42°C, 15 15 minutes at 70°C, and on ice for 1 minute. One microliter of RNase H (2 units) was added and the mixture was incubated at 55°C for 10 minutes. The cDNA was purified using the GlassMax columns (Sambrook, J. Fritsch, E.F., and Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, second 20 edition, Cold Spring Harbor Laboratory Press, Plainview, NY) included in the kit. The cDNA was eluted from the column in 50 jxl dH20, lyophilized, and resuspended in 21 jil dH20. Tailing of the cDNA was accomplished in the following reaction: 7.5 pi dH20, 2.5 pi reaction buffer (200 mM Tris-25 HC1 pH 8.4, 500 mM KCl), 1.5 pi 25 mM MgCl2, 2.5 pi 2 mM dCTP, and 10 pi of the cDNA were incubated at 94°C for 3 <br><br> minutes, then 1 minute on ice. 1 pi (10 units) of terminal deoxynucleotidyl transferase was added and the mixture was incubated for 10 minutes at 37°C. The enzyme was heat 30 inactivated by incubation at 70°C for 10 minutes and the mixture was placed on ice. PCR amplification of the cDNA <br><br> was performed in the following steps: 5 pi of the tailed cDNA was included in a reaction which also contained 5 pi lOx PCR buffer (500 mM KCl, 100 mM Tris-HCl pH 8.3, 15 mM 35 MgCl2, and 0.01% (w/v) gelatin), 1 Hi 10 mM dNTP mix, 2 p.1 <br><br> o o <br><br> ? <br><br> - 65 - <br><br> (10 pmol) anchor primer, 1 pi (20 pmol) primer 3a, and 35 pi dH20. The reaction was heated to 95°C for 1 minute', then 0.9 Hi (4.5 units) Amplitaq polymerase was added. The reaction was cycled 40 times under the following conditions: 94°C for 5 5 seconds, 50°C for 20 seconds, and 72°C for 30 seconds. One microliter of this reaction was used in a nested reamplification to increase levels of specific product for subsequent isolation. The reamplification included: 1 Hi primary amplification, 5 pi lOx PCR buffer, 1 |il 10 mM dNTP <br><br> 10 mix, 2 pi (20 pmol) universal amplification primer, 2 pi (20 <br><br> pmol) primer 4a, and 38 pi dH20. The reaction was heated to <br><br> 95°C for 1 minute, then 0.7 pi (3.5 units) Amplitaq polymerase was added. The reaction was cycled 40 times under these conditions; 94°C for 5 seconds, 50°C for 20 15 seconds, and 72°C for 30 seconds. The amplification products were analyzed via 0.8% agarose gel electrophoresis. A predominant product of approximately 1.2 kilobase pairs was detected. Two microliters of the reaction products were cloned into the pCRII vector from the TA cloning kit 20 (Invitrogen) and incubated at 14°C overnight. The ligation products were used to transform coli. The insert sizes of the resulting transformants were determined following EcoRI digestion. Clones containing inserts of the approximate size of the PCR product were sequenced using 25 fluorescent dye-terminator reagents (Prism, Applied <br><br> Biosystems) and an Applied Biosystems 373 DNA sequencer. The sequence of the RACE product including the EcoRI sites from the TA vector are shown in Figure 3. The sequences of the amplimers (universal amplification primer and the 30 complement to 5'RACE primer 4a) are underlined. <br><br> EXAMPLE 2 - Cloning and Chromosomal <br><br> Localization of the LIPG Gene cDNA library screening <br><br> A human placental cDNA library (Oligo dT and random 35 primed, Cat #5014b, Lot #52033) was obtained from Clontech (Palo Alto, CA). A radiolabeled probe was created by <br><br> o o <br><br> - 66 - <br><br> excising the insert of a plasmid containing the 5'RACE reaction PCR product described above. The probe was radiolabeled using the random priming technique: the DNA fragment (50-100 ng) was incubated with 1 jig of random 5 hexamers (Gibco) at 95°C for 10 minutes followed by 1 minute on ice. At room temperature the following were added: 3 p.1 lOx Klenow buffer (100 mM Tris-HCl pH 7.5, 50 mM MgCL2, 57 mM dithiothreitol; New England Biolabs), 3 pi 0.5 mM dATP, <br><br> dGTP, dTTP), 100 HCi a -32PdCTP (3000 Ci/mmol, New England 10 Nuclear), and 1 jil Klenow fragment of DNA polymerase I (5 <br><br> units, Gibco). The reaction was incubated for 2-3 hours at room temperature and the reaction was then stopped by increasing the volume to 100 |xl with TE pH 8.0 and adding EDTA to a final concentration of l mM. The unincorporated 15 nucleotides were removed by raising the reaction volume to 100 pi and passing over a G-50 spin column (Boehringer Mannheim). The resulting probes had a specific activity greater than 5xl08 cpm/pg DNA. <br><br> The library was probed using established methods 20 (Walter, P., Gilmore, R., and Blobel, G. (1984) Cell 38,5-8). Briefly, the filters were hybridized for 24 hours at 65°C in 4.8X SSPE (20X SSPE = 3.6 M NaCl, 0.2 M NaH2P04, 0.02 M EDTA, pH 7.7), 20 mM Tris-HCl pH 7.6, IX Denhardt's solution (100X= 2% Ficoll 400, 2% polyvinylpyrrolidone, 2% <br><br> 25 BSA), 10% dextran sulfate, 0.1% SDS, 100 Hg/ml salmon sperm DNA, and 1x10s cpm/ml radiolabelled probe. Filters were then washed three times for 15 minutes at room temperature in 2X SSC (IX SSC = 150 mM NaCl, 15 mM sodium citrate pH 7.0), 0.1% sodium dodecyl sulfate (SDS) followed by three washes 30 for 15 minutes each at 65°C in 0.5X SSC, 0.1% SDS. Phage which hybridized to the probe were isolated and amplified. DNA was purified from the amplified phage using LambdaSorb reagent (Promega) according to the manufacturer's instructions. The inserts were excised from the phage DNA 35 by digestion with EcoRI. The inserts were subcloned into the EcoRI site of a plasmid vector (Bluescript II SK, <br><br> o <br><br> - 67 - <br><br> Stratagene). The sequence of the open reading frame contained within the 2.6 kb EcoRI fragment of the cDNA was determined by automated sequencing as described above. The sequence is shown in Figure 4. The amino acid sequence of 5 the predicted protein encoded by the open reading frame is shown in Figure 5 and has been termed LLGXL. The first methionine is predicted to be encoded by nucleotide pairs 252-254. The predicted protein is 500 amino acids in length. The first 18 amino acids form a sequence 10 characteristic of a secretory signal peptide (Higgins, D.G., and Sharp, P.M. (1988) Gene 73, 237-244). The propeptide is predicted to have a molecular weight of 56,800 Daltons. Assuming cleavage of the signal peptide at position 18, the unmodified mature protein has a molecular weight of 54,724 15 Daltons. <br><br> The overall similarities between this protein and the other known members of the triacylglycerol lipase family is illustrated in Figure 6 and Table 1. In the alignment shown in Figure 6, LIPG is the polypeptide (SEQ ID NO: 6) encoded 20 by the cDNA (SEQ ID NO: 5) described in Example 1, and hereafter referred to as LLGN. This protein is identical with the LLGXL protein in the amino terminal 345 residues. Nine unique residues are followed by a termination codon, producing a propolypeptj.de of 39.3 kD and a mature protein 25 of 37.3 kD. The sequences which are common to LLGN and <br><br> LLGXL are nucleic acid sequence SEQ ID NO: 9 and amino acid sequence SEQ ID NO: 10. <br><br> Interestingly, the position at which the LLGN and LLGXL proteins diverge is at a region known from the structure of 30 the other lipase to be between the amino and carboxy domains of the proteins. Therefore, the LLGN protein appears to consist of only one of the two domains of triaclyglycerol lipases. This sequence contains the characteristic "GXSXG" lipase motif at positions 167-171 as well as conservation of 35 the catalytic triad residues at Ser 169, Asp 193, and His 274. Conservation of cysteine residues (positions 64, 77, 252, 272, 297, 308, 311, 316, 463, and 483) which have been <br><br> o o <br><br> - 68 - <br><br> implicated in disulfide linkage in the other lipases suggests that the LLGXL protein has structural similarities to the other enzymes. There are five predicted sites for N-linked glycosylation; at amino acid positions 80, 136, 393, 5 469, and 491. The protein sequences used in the comparisons are human lipoprotein lipase (LPL; Genbank accession #M15856, SEQ ID NO: 13), Human hepatic lipase (HL; Genbank accession #J03540, SEQ ID NO: 14), human pancreatic lipase (PL; Genbank accession # M93285, SEQ ID NO: 15), human 10 pancreatic lipase related protein-1 (PLRP-1; Genbank accession # M93283), and human pancreatic lipase related protein-2 (PLRP-2; Genbank accession # M93284). <br><br> TABLE 1. Similarity of triacylglycerol lipase gene family <br><br> LLGXL <br><br> LPL <br><br> HL <br><br> PL <br><br> PLRP1 <br><br> PLRP2 <br><br> LLGXL <br><br> - <br><br> 42.7 <br><br> 36.5 <br><br> 24.5 <br><br> 22.5 <br><br> 22.6 <br><br> LPL <br><br> 42.7 <br><br> - <br><br> 40.0 <br><br> 22.8 <br><br> 22.7 <br><br> 20.9 <br><br> HL <br><br> 36.5 <br><br> 40.0 <br><br> - <br><br> 22.8 <br><br> 24.0 <br><br> 22.0 <br><br> PL <br><br> 24.5 <br><br> 22.8 <br><br> 22.8 <br><br> — <br><br> 65.2 <br><br> 62.2 <br><br> PLRP1 <br><br> 22.5 <br><br> 22.7 <br><br> r24.0 <br><br> 65.2 <br><br> - <br><br> 61.7 <br><br> PRLP2 <br><br> 22.6 <br><br> 20.9 <br><br> 22.0 <br><br> 62.2 <br><br> 61.7 <br><br> - <br><br> 20 Percent similarity was based on pairwise alignment using the Clustal algorithm (Camps, L., Reina, M., Llobera, M., <br><br> Vilaro, S., and Olivecrona, T. (1990) Am. J. Physiol. 258,C673-C681) in the Megalign program of the Lasergene Biocomputing Software Suite (Dnastar). <br><br> 25 Chromosomal Localization <br><br> DNA from a PI clone (Sternberg, N., Ruether, J. and DeRiel, K. The New Biologist 2:151-62, 1990) containing genomic LLG DNA was labelled with digoxigenin UTP by nick translation. Labelled probe was combined with sheared human 30 DNA and hybridized to PHA stimulated peripheral blood lymphocytes from a male donor in a solution containing 50% formamide, 10% dextran sulfate, and 2X SSC. Specific hybridization signals were detected by incubating the hybridized cells in fluoresceinated antidigoxigenin <br><br> o <br><br> - 69 - <br><br> antibodies followed by counterstaining with DAPI. This initial experiment resulted in specific labeling of a group E chromosome, which was believed to be chromosome 18 on the basis of DAPI staining. <br><br> 5 A second experiment was conducted in which a biotin labelled probe specific for the centromere of chromosome 18 was cohybridized with the LLG probe. This experiment resulted in the specific labeling of the chromosome 18 centromere in red and the long arm of chromosome 18 in 10 green. Measurements of 11 specifically labelled hybridized chromosomes 18 demonstrated that LLG has a Flter of 0.67 (Franke measurement of 0.38), which corresponds to band 18q21. Several genetic diseases, including intrahepatic cholestasis, cone rod dystrophy, and familial expansile 15 osteolysis, are believed to involve defects in this chromosomal region. <br><br> EXAMPLE 3 - LIPG RNA Analysis <br><br> Expression of LIPG RNA in THP-1 cells <br><br> Analysis of the mRNA from which the cDNA was derived was 20 performed by northern analysis of THP-1 RNA. RNA from these cells was prepared as described above. The mRNA was purified from the total RNA through the use of a poly-dT-magnetic bead system (Polyattract system, Promega). Three micrograms of poly (A)-containing mRNA was electrophoresed 25 on a 1% agarose-formaldehyde gel. The gel was washed for 30 minutes in dH20. RNAs were vacuum transferred to a nylon membrane using alkaline transfer buffer (3M NaCl, 8 mM NaOH, 2 mM sarkosyl). After transfer, the blot was neutralized by incubation for 5 minutes in 200 mM phosphate buffer pH 6.8. 30 The RNA was crosslinked to the membrane using an ultraviolet crosslinker apparatus (Stratagene). <br><br> A probe was made by excising the insert of a plasmid containing the 5'RACE reaction PCR product described above. The probe was radiolabeled using the random priming 35 technique described in Example 2. <br><br> o o <br><br> - 70 - <br><br> The filters were prehybridized in QuikHyb rapid hybridization solution (Stratagene) for 30 minutes at 65°C. The radiolabeled probe (1-2 x 106 cpm/ml) and sonicated salmon sperm DNA (final concentration 100 pg/ml) were 5 denatured by heating to 95°C for 10 minutes and quick-chilled on ice before adding to the filter in QuikHyb. <br><br> Hybridization was for 3 hours at 65°C. The unhybridized probe was removed by washing the filters two times for 15 minutes with 2X SSC, 0.1 % sodium dodecyl sulfate at room 10 temperature followed by two times for 15 minutes in 0.1X SSC, 0.1% SDS at 62°C. Following the washes, the filters were allowed to dry briefly and then exposed to Kodak XAR-2 film with intensifying screens at -80°C. The results are shown in Figure 7, which shows a major mRNA species of 15 approximately 4.5 kilobases. Minor species of 4.3 and 1.6 kilobases are also present. The expected size of the LLGN cDNA is 1.6 kb. The LLGXL sequence is likely to be encoded by the major species of mRNA detected. <br><br> Expression of LIPG RNA in various human tissues <br><br> 20 A commercially prepared filter containing 3 j.ig each of mRNAs from human tissues (heart, brain, placenta, lung, <br><br> liver, skeletal muscle, kidney, and pancreas) was obtained from Clontech (Catalog #7760-1). This filter was probed and processed as described above. After probing with the 25 radiolabeled LLG fragment and autoradiography, the probe was stripped by washing in boiling 0.1X SSC, 0.1% SDS for 2 xl5 min. in a 65°C incubator. The membranes were then probed with a 1.4 kilobase pair DNA fragment encoding human lipoprotein lipase. This fragment was obtained by RT-PCR of 30 the THP-1 RNA (PMA and oxLDL treated) using the 5*LPL and 3*LPL primers described in Figure 1. and the RT-PCR conditions described above. After autoradiography, the membranes were stripped again and reprobed with a radiolabeled fragment of the human beta actin cDNA to 35 normalize for RNA content. The results of these analyses are shown in Figure 8. The highest levels of LIPG message were detected in placental RNA, with lower levels found in RNAs derived from lung, liver, and kidney tissue. In <br><br> - 71 - <br><br> agreement with previous studies by others (Verhoeven, A.J.M., Jansen, H. (1994) Biochem. Biophys. Acta 1211,121-124), lipoprotein lipase message was found in many tissues, with highest levels found in heart and skeletal muscle 5 tissue. Results of this analysis indicates that the tissue distribution of LIPG expression is very different from that of LPL. The pattern of LIPG expression is also different from that of either hepatic lipase or pancreatic lipase, as reported by others (Wang, C.-S., and Hartsuck, J.A. (1993) 10 Biochem. Biophys. Acta 1166,1-19; Semenkovich, C.F., Chen, S.-W., Wims, M., Luo C.-C., Li, W.-H., and Chan, L. (1989) J. Lipid Res. 30,423-431; Adams., M.D., Kerlavage, A.R., Fields, C., and Ventex*, C. (1993) Nature Genet. 4,256-265). <br><br> 15 To determine the expression pattern in additional human tissues, another commercially prepared membrane was probed with LLGXL cDNA. This dot blot (Human RNA Master Blot, Clontech Cat. # 7770-1) contains 100-500 ng mRNA from 50 different tissues and is normalized for equivalent 20 housekeeping gene expression (Chen, L., and Morin, R. (1971) Biochim. Biophys. Acta 231,194-197). A 1.6 kb Dral-Srfl fragment of the LLGXL cDNA was labeled with 32PdCTP using a random oligonucleotide priming system (Prime It II, Stratagene) according to the manufacturer's instructions. 25 After 30 minutes prehybridization at 65°C, the probe was added to QuikHyb hybridization solution at l.3xl06 cpm/ml. Hybridization was for 2 hours at 65°C . The unhybridized probe was removed by washing the filters two times for 15 minutes with 2X SSC, 0.1 % sodium dodecyl sulfate at room 30 temperature followed by two times for 15 minutes in 0.1X SSC, 0.1% SDS at 62°C. Following the washes, the filters were allowed to dry briefly and then exposed to Kodak XAR-2 film with intensifying screens at -80°C. for varying amounts of time. The resulting images were quantitated by 35 densitometry. The results are shown in Table 2. The relative expression levels of tissues represented in both the multiple tissue northern and the multiple tissue dot blot are similar, with highest levels in placenta, and lower <br><br> o o <br><br> - 72 - <br><br> levels in lung, liver and kidney. Fetal liver, kidney, and lung also express roughly the same levels as the adult tissues. Surprisingly, thyroid tissue expression levels were the highest of all tissues represented, with expression 5 of 122% of that in placental tissue. While there is precedence for lipase expression by the placenta (Rothwell, J.E., Elphick, M.C. (1982) J. Dev. Physiol. 4,153-159; Verhoeven, Carling D., and Jansen H. (1994) J. Lipid <br><br> Res. 35, 966-975; Burton, B.K., Mueller, H.W. (1980) 10 Biochim. Biophys. Acta 618,449-460), the thyroid was not previously known to express any lipase. These results suggest that LIPG expression may be involved in maintenance of the placenta, where LIPG may serve to liberate free fatty acids from substrates such as phospholipids as a source of 15 energy. The LIPG expressed in the thyroid may provide precursors for the synthesis of bioactive molecules by that gland. <br><br> Table 2. Expression of LIPG mRNA in various human tissues <br><br> 20 <br><br> 25 <br><br> 30 <br><br> whole fanin <br><br> N.D. <br><br> substantial nigra <br><br> N.D. <br><br> uterus <br><br> N.D. <br><br> mammary gland <br><br> N.D. <br><br> lung <br><br> 29 <br><br> amygdala <br><br> N.D. <br><br> temporal lobe <br><br> N.D. <br><br> prostate <br><br> 5 <br><br> kidney <br><br> 44 <br><br> trachea <br><br> 12 <br><br> nucleus <br><br> NJX <br><br> thalamus <br><br> N.D. <br><br> stomach <br><br> N.D. <br><br> fcver til placenta <br><br> . 100 <br><br> cerebellum <br><br> 4 <br><br> sub-thalamic nuclcu* <br><br> N.D. <br><br> testes <br><br> 9 <br><br> small intestine <br><br> ' 6 <br><br> fetal brain <br><br> 5 <br><br> cerebral cortex <br><br> N.D. <br><br> spina) cord <br><br> N.D <br><br> ovary <br><br> N.P. <br><br> spleen <br><br> N.D. <br><br> fetal heart <br><br> N.D. <br><br> frontal k&amp;e <br><br> N.D. <br><br> heart <br><br> N.D. <br><br> pancreas <br><br> N.D. <br><br> thymus <br><br> N.D. <br><br> fetal kidney <br><br> 56 <br><br> hippocampus <br><br> N.D. <br><br> aorta <br><br> ND. <br><br> pituitary piand <br><br> N.D. <br><br> peripheral leukocyte <br><br> N.D. <br><br> fetal bv« <br><br> 14 <br><br> medulla oblongata. <br><br> N.D. <br><br> skeletal musdc <br><br> NX). <br><br> adrenal gbnd <br><br> N.D. <br><br> lymph node <br><br> N.D. <br><br> foal spleen <br><br> N.D. <br><br> occipital lobe <br><br> N.D. <br><br> colon <br><br> 8 <br><br> thyroid gland <br><br> 122 <br><br> bone marrow <br><br> N.D. <br><br> fetal thymus <br><br> N.D. <br><br> putamen <br><br> N.D. <br><br> tdaddet <br><br> N.D. <br><br> salivary gland <br><br> N.D. <br><br> appendix <br><br> 7 <br><br> fetal lung <br><br> 8 <br><br> Values given are percentage of expression with levels in placental tissue arbitrarily set at 100%. Values are average of densitometric measurements from two 35 autoradiographic exposures. N.D. = not detectable. <br><br> Expression of LIPG RNA in cultured endothelial cells <br><br> Human umbilical vein endothelial cells (HUVEC) and human coronary arterial endothelial cells (HCAEC) were obtained from Clonetics. HUVECs were propagated in a commercially <br><br> o <br><br> - 73 - <br><br> prepared endothelial cell growth medium (EGM, Clonetics) supplemented with 3 mg/ml bovine brain extract (Maciag, T., Cerundolo, J., Ilsley, s., Kelley, P.R., and Forand, R. (1979) Proc. Natl. Acad. Sci. USA 76, 5674-5678), 5 Clonetics), while HCAECs were propagated in EGM supplemented with 3 mg/ml bovine brain extract and 3% fetal bovine serum (5% final concentration). Cells were grown to confluence, then the medium was changed to EGM without bovine brain extract. Cultures were stimulated by adding 100 ng/ml of 10 phorbol myristate (Sigma). After 24 hours incubation, the RNAs were extracted from the cells via the Trizol method described above. Twenty micrograms of total RNA was electrophoresed and transferred to the membrane for analysis. The membranes were probed with LIPG and LPL 15 probes as described above. The results are shown in Figure 9. Twenty micrograms of total RNA from THP-1 cells stimulated with PMA was run on the blot for comparison. RNA hybridizing to the LIPG probe was detected in unstimulated and PMA stimulated HUVEC cells. In contrast, detectable 20 levels of LIPG mRNA were only found in HCAEC cultures after stimulation with PMA. In agreement with previous studies of others, no detectable lipoprotein lipase mRNA was detected in any of the endothelial RNAs (Verhoeven, A. J.M., Jansen, H. (1994) Biochem. Biophys. Acta 1211,121-124). <br><br> 25 EXAMPLE 4 - LIPG Protein Analysis Antibody preparation <br><br> Antisera were generated to peptides with sequences corresponding to a region of the predicted protein encoded by the LIPG cDNA open reading frame. This peptide was 30 chosen because of its high predicted antigenicity index <br><br> (Jameson B.A., and Wolf, H. (1988) Comput. Applic. in the Biosciences 4,181-186). The sequence of the immunizing peptide was not found in any protein or translated DNA sequence in the Genbank database. Its corresponding 35 position in the LIPG protein is shown in Figure 10. The carboxy terminal cysteine of the peptide does not correspond to the residue in the LIPG putative protein, but was introduced to facilitate coupling to the carrier protein. <br><br> {) <br><br> - 74 - <br><br> The peptide was synthesized on a Applied Biosystems Model 433A peptide synthesizer. Two milligrams of peptide was coupled to two milligrams of maleimide-activated keyhole limpet hemocyanin following the protocols included in the 5 Imject Activated Immunogen Conjugation Kit (Pierce <br><br> Chemical) . After desalting, one-half of the conjugate was emulsified with an equal volume of Freund's complete adjuvant (Pierce). This emulsification was injected into a New Zealand White rabbit. Four weeks after the initial 10 inoculation, a booster inoculation was made with an emulsification made exactly as described above except Freund's incomplete adjuvant (Pierce) was used. Two weeks after the boost, a test bleed was made and titers of specific antibodies were determined via ELISA using 15 immobilized peptide. A subsequent boost was made one month after the first boost. <br><br> Western analysis of medium from endothelial cell cultures <br><br> HUVEC and HCEAC cells were cultured and stimulated with 20 PMA as described in Example 3C, except that the cells were stimulated with PMA for 48 hours. Samples of conditioned medium (9 ml) were incubated with 500 pi of a 50% slurry of heparin-Sepharose CL-6B in phosphate buffered saline (PBS, 150 mM sodium chloride, 100 mM sodium phosphate, pH 7.2). 25 Heparin-Sepharose was chosen to partially purify and concentrate • the LIPG proteins because of the conservation of residues in the LLGXL sequence which have been identified as critical for the heparin-binding activity of LPL (Ma, Y., Henderson, H.E., Liu, M.-S., Zhang, H., Forsythe, I.J., 30 Clarke-Lewis, I., Hayden, M.R., and Brunzell, J.D. J. Lipid Res. 35, 2049-2059; and Fig. 6.). After rotation at 4°C for 1 hour, the samples were centrifuged for 5 minutes at 150 x g. The medium was aspirated and the Sepharose was washed with 14 ml PBS. After centrifugation and aspiration, the 35 pelleted heparin-Sepharose was suspended in 200 pi 2x SDS <br><br> loading buffer (4% SDS, 20% glycerol, 2% p-mercaptoethanol, 0.002% bromphenol blue, and 120 mM Tris pH 6.8). The <br><br> o <br><br> - 75 - <br><br> samples were heated to 95°C for 5 minutes and 40 p.1 was loaded onto a 10% Tris-Glycine SDS gel. After electrophoresis at 140 V for approximately 90 minutes, the proteins were transferred to nitrocellulose membranes via a 5 Novex electroblotting apparatus (210 V, 1 hour). The membranes were blocked for 30 minutes in blocking buffer (5% nonfat dried milk, 0.1% Tween 20, 150 mM sodium chloride, 25 mM Tris pH 7.2). Antipeptide antisera and normal rabbit serum was diluted 1:5000 in blocking buffer and was 10 incubated with the membranes overnight at 4°C with gentle agitation. The membranes were then washed 4x 15 minutes with TBST (0.1% Tween 20, 150 mM sodium chloride, 25 mM Tris pH 7.2). Goat anti-rabbit peroxidase conjugated antisera (Boehringer Mannheim) was diluted 1:5000 in blocking buffer 15 and incubated with the membrane for 1 hour with agitation. <br><br> The membranes were washed as above, reacted with Renaissance chemiluminescent reagent (DuPont NEN),and exposed to Kodak XAR-2 film. The results are shown in Figure 11. Two species of immunoreactive proteins are present in the 20 samples from unstimulated HUVEC and HCAEC cells. Levels of immunoreactive protein in the unstimulated HCAEC samples are much lower than the corresponding HUVEC sample. Upon stimulation with PMA, three immunoreactive proteins are secreted by the endothelial cell cultures. PMA exposure 25 greatly increased the level of LIPG proteins produced by the HCAEC cultures. PMA induction of LLG proteins was not as dramatic in the HUVEC cultures. <br><br> EXAMPLE 5 - Recombinant LIPG Protein Production LIPG expression constructs <br><br> 30 The cDNAs encoding the LLGN and LLGXL proteins were cloned into the mammalian expression vector pCDNA3 (Invitrogen). This vector allows expression of foreign genes in many mammalian cells through the use of the cytomegalovirus major late promoter. The LLGN 5*RACE product 35 was cloned into the EcoRI site of pCDNA3. The LLGXL cDNA <br><br> was digested with Dral and Srfl to yield a 1.55 kb cDNA (see Figure 4.). The vector was digested with the restriction enzyme EcoRV and the vector and insert were ligated using T4 <br><br> o <br><br> - 76 - <br><br> DNA ligase and reagents from the Rapid Ligation Kit (Boehringer Mannheim) according to the manufacturers . instructions. The ligation products were used to transform competent EL. coli . Resultant colonies were screened by 5 restriction analysis and sequencing for the presence and orientation of the insert in the expression vector. <br><br> Transient transfection of LIPG in COS-7 cells <br><br> The LIPG expression vectors were introduced into COS-7 cells through the use of Lipofectamine cationic lipid 10 reagent (GIBCO). Twenty-four hours before the transfection, COS-7 cells were plated onto 60 mm tissue culture dishes at a density of 2x10s cells/plate. The cells were propagated in Dulbecco's modified Eagle's medium (DMEM; GIBCO) <br><br> supplemented with 10% fetal calf serum, 100 U/ml penicillin, 15 100 ng/ml streptomycin. One microgram of plasmid DNA was added to 300 pi of Optimem I serum-free medium (Gibco). Ten microliters of Lipofectamine reagent were diluted into 300 jil of Optimem I medium and this was combined with the DNA solution and allowed to sit at room temperature for 30 20 minutes. The medium was removed from the plates and the cells were rinsed with 2 ml of Optimem medium. The DNA-Lipofectamine solution was added to the plates along with 2.7 ml Optimem medium and the plates were incubated for 5 hours at 37°C. After the incubation, the serum free medium 25 was removed and replaced with DMEM supplemented with 2% FBS and antibiotics. Twelve hours post-tranisfection, some of the cultures were treated with either 0.25 mM Pefabloc SC (Boehringer Mannheim), a protease inhibitor, or 10 U/ml heparin. Thirty minutes before harvest, the heparin treated 30 samples were treated with an additional 40 U/ml heparin. The medium was removed from the cells 60 hours after transfection. Heparin-Sepharose CL-4B (200 p.1 of a 50% slurry in PBS pH 7.2) was added to l ml of medium and was mixed at 4°C for 1 hour. The Sepharose was pelleted by low 35 speed centrifugation and was washed three times with 1 ml cold PBS. The Sepharose was pelleted and suspended in 100 Hi 2x loading buffer. The samples were heated to 95°C for 5 <br><br> o o <br><br> - 77 - <br><br> minutes. 40 (j.1 of each sample,was loaded onto a 10% SDS-PAGE gel. Electrophoresis and western analysis was performed using the anti-LIPG antiserum as described above. The results are shown in Figure 12. Proteins from HCAEC 5 conditioned medium were included for size references. LLGN migrates at approximately 40 kD, corresponding to the lowest band in HCAEC. The medium from COS cells transfected with LLGXL cDNA contains both 68 kD and 40 kD species. When these cells were treated with heparin, the amount of both 68 10 kD and 40 kD proteins recovered from the medium increased dramatically, indicating either the release of proteoglycan-bound protein from the cell surface or stabilization of the proteins by heparin. When the cells were treated with the protease inhibitor Pefabloc, the amount of 68 kD protein 15 increased relative to that of the 40 kD species. This suggests that the lower molecular weight protein produced by these cells is a proteolysis product of the larger 68 kD form. The role of the mRNA identified through differential display which encodes a shorter, 40 kD species is not known. 20 There has, however, been a report of an alternately-spliced form of hepatic lipase which apparently is expressed in a tissue-specific manner and would create a truncated protein. <br><br> EXAMPLE 6 - LIPG in Animal Species <br><br> Cloning the Rabbit Homo log of LIPG <br><br> 25 A commercially available lambda cDNA library derived from rabbit lung tissue (Clontech, Cat. #TL1010b) was used to isolate a fragment of the rabbit homolog of the LIPG gene. Five microliters of the stock library were added to 45 p.1 water and heated to 95°C for 10 minutes. The following were <br><br> 30 added in a final volume of 100 |il: 200 pM dNTPs, 20 mM Tris- <br><br> HCl pH 8.4, 50 mM KCl, 1.5 mM MgCl2, 100 HM each primer DLIP774 and LLGgen2a, and 2.5 U Taq polymerase (GIBCO). The reaction was thermocycled 35 times with the parameters of: 15 seconds at 94°C, 20 seconds at 50°C and 30 seconds at 72°C. <br><br> 35 Ten microliters of the reaction was analyzed via agarose gel electrophoresis. A product of approximately 300 basepairs was detected. A portion (4 pi) of the reaction mix was used <br><br> - 78 - <br><br> to clone the product via the TA cloning system. The insert of a resulting clone was sequenced (SEQ ID NO: 11). An alignment between the deduced rabbit amino acid sequence (SEQ ID NO: 12) and the corresponding sequence of the human cDNA 5 is also shown in Figure 14. Of the nucleotides not part of either amplification primer, there is an 85.8% identity between the rabbit and human LLG sequences. The predicted protein encoded by this rabbit cDNA shares 94.6 % identity with that of the human protein, with most of the nucleotide 10 substitutions in the third or "wobble" positions of the codons. Notably, this region spans the "lid" sequence of the predicted LLG proteins and is a variable domain in the lipase gene family. This is evidence that there is a high degree of conservation of this gene between species. <br><br> 15 LIPG in Other Species <br><br> To demonstrate the presence of LLG genes in other species, genomic DNAs from various species were restriction digested with EcoRI, separated by electrophoresis in agarose gels, and blotted onto nitrocellulose membranes. <br><br> 20 The membranes were hybridized overnight at 65°C with 2.5 <br><br> x 106 cpm/ml of random primed 32 P-LLG or 32 P-LPL (lipoprotein lipase) probe in a hybridization solution of 6X SSC, 10% <br><br> dextran sulfate, 5 X Dendardt's solution, 1% SDS, and 5 pg/ml salmon sperm DNA. The membranes were washed with 0.1X SSC, 25 0.5% SDS for ten minutes at room temperature, then sequentially for ten minutes at 40°C, 50°C, and 55°C. Autoradiograms of the blots are shown in Figure 16. <br><br> Figure 16 shows the presence of LLG and LPL genes in all species examined, with the exception that no hybridization 30 was observed with the LLG probe against rat DNA. The exceptional data from rat may represent an artifact caused by generation of abnormally sized restriction fragments containing LLG sequences. Such fragments may be outside of the fractionation range of the agarose gel or may blot 35 inefficiently. The different bands detected by the two <br><br> o <br><br> - 79 - <br><br> probes indicate that LPL and LIPG are separate, <br><br> evolutionarily conserved genes. <br><br> EXAMPLE 7 - Enzvmatic Activity of LLGXL <br><br> Phospholipase Activity <br><br> 5 Conditioned media from COS-7 cells transiently expressing human lipoprotein lipase (LPL), LLGN, or LLGXL were assayed for phospholipase activity. MEM containing 10% FBS (MEM) was used as the blank, and conditioned media from COS-7 cells transfected with an antisense LLGXL plasmid (AS) 10 was used as a negative control. <br><br> A phosphatidylcholine (PC) emulsion was made up using 10 pi phosphatidylcholine (10 mM), 40 pi 14C- <br><br> phosphatidylcholine, dipalmitoyl (2 HCi) , labeled at the sn 1 and 2 positions, and 100 pi Tris-TCNB [100 mM Tris, 1% 15 Triton, 5 mM CaCl2, 200 mM NaCl, 0.1% BSA) . The emulsion was evaporated for 10 minutes, then brought to a final volume of l ml in Tris-TCNB. <br><br> Reactions were performed in duplicate and contained 50 Hi PC emulsion and 950 pi medium. Samples were incubated in a 20 shaking water bath for 2-4 hours at 37°C. The reactions were terminated by adding 1 ml IN HC1, then extracted with 4 ml of 2-propanol:hexane (1:1) . The upper 1.8 ml hexane layer was passed through a silica gel column, and the liberated 14C-free fatty acids contained in the flow-thru fraction were 25 quantitated in a scintillation counter. The results of these assays are shown in Figure 14. <br><br> Triacylglycerol Lipase Activity <br><br> Conditioned media from COS-7 cells transiently expressing human lipoprotein lipase (LPL), LLGN, or LLGXL 30 were assayed for triglycerol lipase activity. MEM containing 10% FBS was used as the blank, and conditioned media from COS-7 cells transfected with an antisense LLGXL plasmid (AS) was used as a negative control. <br><br> - 80 - <br><br> A concentrated substrate was prepared as an anhydrous emulsion of labeled triolein, [9,10- 3H(N)) and unlabeled triolein (final total triolein = 150 mg with 6.25 x 108cpm), which was stabilized by adding 9 mg of lecithin in 100% 5 glycerol. 0.56 ml of 3H-triolein, (0.28 mCi) was mixed with <br><br> 0.17 ml of unlabeled triolein and 90 pi of lecithin (9 mg) . The mixture was evaporated under a stream of nitrogen. The dried lipid mixture was emulsified in 2.5 ml 100% glycerol by sonication (30 second pulse level 2 followed by 2 second 10 chill cycles over 5 minutes]. <br><br> The assay substrate was prepared by dilution of l volume of concentrated substrate with 4 volumes of 0.2M Tris-HCl buffer (pH 8.0) containing 3% w/v fatty acid free bovine serum albumin. The diluted substrate was vortexed vigorously 15 for 5 seconds. <br><br> Reactions were performed in duplicate in a total volume of 0.2 ml containing 0.1 ml of assay substrate and 0.1 ml of the indicated conditioned media. The reactions were incubated for 90 minutes at 37°C. The reactions were 20 terminated by adding 3.25 ml of methanol-chloroform-heptane 1.41:1.25:1 (v/v/v) followed by 1.05 ml of 0.1M potassium carbonate-borate buffer (pH 10.5). After vigorous mixing for 15 seconds, the samples were centrifuged for 5 minutes at 1000 rpm. A 1.0 ml aliquot of the upper aqueous phase was 25 counted in a scintillation counter. The results of these assays are shown in Figure 15. <br><br> EXAMPLE 8 - Use of LIPG Polypeptide to Screen for Enhancers or Inhibitors <br><br> Recombinant LIPG is produced in baculovirus-infected 30 insect cells or stably transfected CHO cells or other acceptable mammalian host cells. Recombinant LIPG is purified from the serum-containing or serum-free conditioned medium by chromatography on heparin-Sepharose, followed by chromatography on a cation exchange resin. A third 35 chromatographic or further chromatographic steps, such as molecular sieving, is used in the purification of LIPG if <br><br> o <br><br> - 81 - <br><br> needed. During purification, anti-peptide antibodies are used to monitor LIPG protein and the phospholipase assay is used to follow LIPG activity. <br><br> In the fluorescent assay, the final assay conditions are 5 approximately 10 mM Tris-HCl (pH 7.4), 100 mM KCl, 2 mM CaCl2, <br><br> 5 pM C6NBD-PC{l-acyl-2-[6-(nitro-2,l,3-benzoxadiazol-4-yl) amino] caproylphosphatidylcholine, and LIPG protein (approx. 1-100 ng). The reaction is subjected to fluorescence excitation at 470 nm, and enzyme activity, as 10 measured by the fluorescence emission at 540 nm is continuously monitored. Compounds and/or substances to be tested for stimulation and/or inhibition of LIPG activity are added as 10-200 mM solutions in dimethylsulfoxide. Compounds which stimulate or inhibit LIPG activity are identified as 15 causing an increased or decreased fluorescence emission at 540 nm. <br><br> In the thio assay, the final assay conditions are approximately 25 mM Tris-HCl (pH 8.5), 100 mM KCl, 10 mM CaCl2, 4.24 mM Triton X-100, 0.5 mM l,2-bis(hexanoylthio)-20 l,2-dideoxy-sn-glycero-3-phosphorylcholine, 5 mM 4,4*- <br><br> dithiobispyridine (from a 50 mM stock solution in ethanol), and 1-100 ng recombinant LIPG. Phospholipase activity is determined by measuring the increase in absorption at 342 nm. Compounds and/or substances to be tested for stimulation 25 and/or inhibition of LIPG activity are added as 10-200 mM <br><br> solutions in dimethylsulfoxide. Compounds which stimulate or inhibit LIPG activity are identified as causing an increased or decreased absorption at 342 nm. <br><br> EXAMPLE 9 - Transgenic Mice Expressing Human LIPG <br><br> 30 To further study the physiological role of LIPG, <br><br> transgenic mice expressing human LIPG are generated. <br><br> The 1.53 kb Dral/Srfl restriction fragment encoding LLGXL (see Figure 4) was cloned into a plasmid vector (pHMG) downstream of the promoter for the ubiquitously expressed 3-35 hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase gene. <br><br> f) <br><br> o <br><br> - 82 - <br><br> Transgenic mice expressing different levels of human LLGXL are generated using standard methods (see, e.g., G.L. Tremp et al. Gene 156:199-205, 1995). The transgenic mice are used to determine the impact of LLGXL over express ion on lipid 5 profile, vascular pathology, rate of development and severity of atherosclerosis, and other physiological parameters. <br><br> EXAMPLE 10 - Expression of LIPG in Atherosclerotic Tissues <br><br> LLGXL expression in atherosclerosis was examined by performing a reverse transcription-polymerase chain reaction 10 (RT-PCR) using mRNA isolated from vascular biopsies from four patients with atherosclerosis. The tissue samples were from the aortic wall (one sample), the iliac artery (two samples), and the carotid artery (one sample). <br><br> Atherosclerosis biopsies were received from 15 Gloucestershire Royal Hospital, England, and polyA+ mRNA was prepared and resuspended in diethylpyrocarbonate (DEPC) <br><br> treated water at a concentration of 0.5 ixg/p.1 mRNA. Reverse transcriptase reactions were performed according to the GibcoBRL protocol for Superscript Preampl if ication System for 20 First Strand cDNA Synthesis. Briefly, the cDNA was synthesized as follows: 2 pi of each mRNA was added to 1 pi oligo (dT)12_18 primer and 9 ji.1 of DEPC water. The tubes were incubated at 70 °C for 10 minutes and put on ice for 1 minute. To each tube, the following components were added: 2 nl 10X 25 PCR buffer, 2 pi 25 mM MgCl2, 1 pi 10 mM dNTP mix and 2 pi <br><br> 0.1M DTT. After 5 minutes at 42 °C, 1 pi (200 units) of Super Script II reverse transcriptase was added. The reactions were mixed gently, then incubated at 42 °C for 50 minutes. The reactions were terminated by incubation at 70 °C for 15 30 minutes then put on ice. The remaining mRNA was destroyed by the addition of 1 [il of RNase H to each tube and incubated for 20 minutes at 37 °C. <br><br> PCR amplifications were performed using 2 |il of the cDNA reactions. To each tube the following were added: 5 p.1 10X <br><br> - 83 - <br><br> PCR buffer, 5 pi 2 mM dNTPs, 1 nl hllg-gspl primer (20 pmol/ml, see Figure 1), 1 |il hllg-gsp2a primer (20 pmol/ml, see Figure 1), 1.5 |Al 50 mM MgCl2, 0.5 p,l Taq polymerase (5U/mI) and 34 Ml water. After holding the reactions at 95°C 5 for 2 minutes, thirty cycles of PCR were performed as follows: 15 seconds at 94°C, 20 seconds at 52°C, and 30 seconds at 72°C. The finished reactions were held for 10 minutes at 72°C before analysis by agarose gel electrophoresis. The hllg-gsp primers are specific for LIPG 10 and yield an expected product of 300 bp. In a parallel PCR to show that the cDNA synthesis reactions had been successful, primers specific for the housekeeping gene, G3PDH (human glyceraldehyde 3-phosphate dehydrogenase) were used (1 Ml each at 20 pmol/ml) . <br><br> 15 The G3PDH primers (see Figure 1) yielded the expected product of 983 bp in all four vascular biopsy samples. LIPG expression was detected in three of the four samples, with no expression being detected in the carotid artery sample. <br><br> Example 11 - Differential display, <br><br> 20 RT-PCR and cDNA library screening <br><br> To perform the experiments discussed in Examples 12 to 16, the following procedure (based on the procedure outlined in Example 1) was used to obtain the cDNA for LIPG. THP-1 cells were plated in the presence of phorbol 12-myristate 13-25 acetate (PMA, 40 ng/ml; Sigma) for 48 hours. The differentiated THP-1 cells were exposed for 24 hours to either oxLDL (50 ng/ml) or control medium. Total RNAs were collected and purified using standard procedures. Poly(A)+ RNA was purified from total RNA using a poly-dT magnetic bead 30 system (Promega) . cDNA synthesis and PCR amplification were accomplished using protocols from the Differential Display kit, version 1.0 (Display Systems Biotechnology). The primer pairs that yielded the initial cDNA fragment of EL were downstream primer 7 (5'-TTTTTTTTTTTGA-3') and upstream primer 35 15 (5 • -GATCCAATCGC-3 ') . The amplification reaction was fractionated on a 6% nondenaturing acrylamide sequencing <br><br> o o <br><br> - 84 - <br><br> format gel and an amplification product found only in the reaction containing cDNA from THP-1 cells exposed to oxLDL was identified and excised from the gel. A reamplification using the same primers was performed and the product was 5 excised and subcloned into the pCRII vector using the TA cloning system (Invitrogen). Insert sizes were determined using EcoRI digestions of the plasmids, and clones containing inserts of the approximate size of the original PCR product were sequenced using fluorescent dye-terminator reagents 10 (Prism, Applied Biosystems) and an Applied Biosystems 373 DNA sequencer. We extended the cDNA sequence of the original, gel-excised cDNA using the 5*-RACE system (GIBCO). RNA (1 <br><br> ixg) from the THP-1 cells used initially in the differential display reactions was used in the 51-RACE procedure using a 15 gene-specific primer (5•-TAGGACATGCACAGTGTAATCTG-3•) for first strand cDNA synthesis. We performed PCR amplification of the cDNA using an anchor primer and gene-specific primer 2 <br><br> (5' -GATTGTGCTGGCCACTTCTC-3 1) . This reaction (1 /nl) was used in a nested re-amplification using the universal 20 amplification primer (5 '-CUACUACUACUAGGCCACGCGTCGACTAGTAC-3 •) and the gene-specific primer 3 (5 • —GACACTCCAGGGACTGAAG-3 ') to increase levels of specific product for subsequent isolation. The reaction products were cloned into the pCRII vector from the TA cloning kit and determined the sequence. A human 25 placental cDNA library (oligo dT and random primed) was obtained from Clontech and probed with the 5'-RACE reaction PCR product. The DNA from hybridizing clones was purified using LambdaSorb reagent (Promega). Inserts were excised from the phage DNA by digestion with EcoRI, subcloned into 30 the JScoJ?I site of the Bluescript II SK plasmid vector (Stratagene) and sequenced. <br><br> Example 12 - Antibody Preparation <br><br> A 17-residue peptide (GPEGRLEDKLHKPKATC) was synthesized corresponding to residues 8-23 of the secreted LIPG gene 35 product pn a Model 433A peptide synthesizer (Applied Biosystems). Peptide (2 mg) was coupled to maleimide-activated keyhole limpet haemocyanin (2 mg) following the protocols included in the Imject Activated Immunogen <br><br> - 85 - <br><br> Conjugation kit (Pierce Chemical). After desalting, one-half of the conjugate was emulsified with an equal volume of Freund's complete adjuvant (Pierce) and injected into a New Zealand White rabbit. Four weeks after the initial 5 inoculation, a booster inoculation was administered with an emulsification made exactly as described above except for the use of Freund's incomplete adjuvant (Pierce). Two weeks after the boost, the titres of specific antibodies were determined in a test bleed via ELISA using immobilized 10 peptide. <br><br> Example 13 - Gene expression studies <br><br> HUVECs were propagated in a commercially prepared endothelial cell growth medium (EGM, Clonetics) supplemented with bovine brain extract (3 mg/ml; Clonetics), whereas 15 HCAECs were propagated in EGM with bovine grain extract (3 mg/ml) and 5% fetal bovine serum. Cultures were stimulated by addition of PMA (100 ng/ml). After 24 hours incubation, RNA was extracted from the cells via the Trizol method, electrophoresed on a 1% agarose-formaldehyde gel, transferred 20 to Nytran membrane on a Turboblotter apparatus (Schleicher and Schuell) and crosslinked to the membrane using a Stratalinker ultraviolet crosslinker (Stratagene). The 5'-RACE reaction PCR product was radiolabelled using the random priming technique. The radiolabelled probe (1-2X106 cpm/ml) 25 was denatured by heating to 95 °C for 10 minutes and quick-chilled on ice before adding to the filter in QuikHyb. Hybridization was allowed to proceed for 3 hours at 65 °C. Filters were exposed to Kodak XAR-2 film with intensifying screens at -80 °C. We incubated HUVEC- and HCEAC-conditioned 30 medium with heparin-Sepharose CL-6B at 4 °C for 1 hour. <br><br> After centrifugation, the pelleted heparin-Sepharose was suspended in SDS loading buffer, heated to 95 °C for 5 minutes and loaded onto a 10% Tris-Glycine SDS gel (NOVEX). After electrophoresis at 140 V for 90 minutes, the proteins 35 were transferred to nitrocellulose membranes and detected with rabbit anti-LIPG peptide antisera (1:5,000), with goat anti-rabbit peroxidase conjugated antisera (1:5,000; Boehringer) as the secondary antibody. The membranes were <br><br> o <br><br> I <br><br> o <br><br> - 86 - <br><br> reacted with Renaissance chemiluminescent reagent (DuPont NEN) and exposed to Kodak XAR-2 film. A commercially prepared filter containing poly (A)+ RNAs (3 ng each) from human heart, brain, placenta, lung, liver, skeletal muscle, 5 kidney and pancreas (Clontech)was hybridized with a radiolabelled fragment and processed as described above. Following autoradiography, the blot was stripped by washing in boiling O.lxSSC, 0.1% SDS for 2x15 minutes at 65 °C and then probed as described above with a 1.4-kb cDNA fragment 10 encoding human LPL. This fragment was obtained by RT-PCR of the THP-1 RNA (PMA and oxLDL treated) using the 5' LPL and 3' LPL primers 5 1-ACCACCATGGAGAGCAAAGCCCTG-3 » and 5'-CCAGTTTCAGCCTGACTTCTTATTC-3 1 , respectively. After exposure to film, the membranes were stripped again and reprobed with 15 a radiolabelled fragment of human p actin cDNA to normalize to RNA content. <br><br> Human umbilical vein endothelial cells (HUVEC) were negative for LPL mRNA expression as expected, but were found to constitutively express a high level of mRNA for the LIPG 20 gene (Figure 9). <br><br> Human coronary artery endothelial cells (HCAEC) were also found to express the mRNA which was further upregulated on treatment of these cells with phorbol ester (Figure 9) . <br><br> Conditional medium from stimulated HUVEC and HCAEC 25 contained immunoreactive proteins of approximately 68 kD and 40 kD, as well as a less prominent band of 55 kD (Figure 11) . <br><br> To determine the tissue sites of LIPG production in vivo, a multiple human tissue northern blot analysis with probes for both LIPG and LPL was performed. Abundant levels 30 of LIPG mRNA were found in lung, liver and kidney (Figure 8) tissues, which showed low levels of LPL expression. LIPG was also expressed at high levels in the placenta (Figure 8), suggesting the potential for a role in development. <br><br> o o <br><br> - 87 - <br><br> In tissues such as heart and skeletal muscle, which expressed the highest amount of LPL (confirming previous reports, Goldberg, J.I., «J. Lipid Res., 37, 693-707 (1996)), we did not detect LIPG expression. This analysis indicated 5 that the tissue distribution of LIPG expression is very different from that of LPL, as well as that reported for HL and PL. We found no LIPG mRNA in adrenals or ovaries, but did find a very low level of LIPG mRNA in the testes (data not shown) . We also found that HepG2 cells express LIPG mRNA 10 and protein in vitro (data not shown), but at levels less than 10% of that expressed by HUVECs. <br><br> Example 14 - Lipase Assays <br><br> The cDNA and the 1.4-kb LPL cDNA were cloned into the EcoRV site of the mammalian expression vector pCDNA3 15 (Invitrogen) . An antisense pCDNA3 vector was used as negative control. The recombinant expression vectors (3 ng) were mixed with lipofectamine (Life Technologies) and transfected in quadruplicate into semiconfluent COS7 cells in 60-mm dishes. Established methods were used to assay samples 20 of conditioned media from transfected COS7 cells for TG <br><br> lipase and phospholipase activities (Goldberg, J.I., J. Lipid Res., 37, 693-707 (1996)). for the TG lipase assay, 9,10-3H(N)-triolein (250 /xCi; NEN) was mixed with unlabeled triolein (150 mg) and type IV-S-a lecithin (9 mg; Sigma) in 25 glycerol. The mixture was evaporated under nitrogen and emulsified in glycerol (2.5 ml) by sonication with a Branson Sonifier 450. The assay substrate was prepared by combining one volume of the emulsified substrate, four volumes of Tris-HCl (0.2 M, pH 8.0) containing 3% (w/v) fatty acid-free 30 bovine serum albumin (BSA) and one volume of heat-inactivated bovine serum. Reactions were performed in triplicate in a total volume (0.2 ml) containing assay substrate (0.1 ml) and conditioned media (0.1 ml). The reactions were incubated for 2 hours at 37°C and terminated by adding methanol-chloroform-35 heptane (1.41:1.25:1; 3.25 ml) followed by potassium carbonate-borate buffer (1.05 ml; 0.1 M, pH 10.5). After vigorous mixing for 15 seconds, the samples were centrifuged for 5 minutes at 1,000 rpm and the upper aqueous phase (1.0 <br><br> o <br><br> - 88 - <br><br> ml) was counted in a scintillation counter. For the phospholipase assay, a phosphatidylcholine (PC) emulsion was made by combining 14C-dipalmitoyl PC (2 fxCi; NEN) and lecithin <br><br> (10 /xl) with Tris-TCNB (100 /xl; 100 mM Tris-HCl pH 7.4, 1% 5 Triton X-100, 5 mM CaCl2, 200 mM NaCl, 0.1% BSA) . The mixture was vortexed for 2 minutes and then evaporated under nitrogen. The dried lipid was reconstituted with TCNB (1 ml) and vortexed for 10 seconds. Reactions were performed in triplicate and contained PC emulsion (50 /xl) , conditioned 10 media (600 /x 1) and MEM (350 /x 1) . Samples were incubated at 37°C for 2 hours, terminated by addition of HC1 (1 ml) and extracted with 2-propanol:hexane (1:1; 4ml). A sample (1.8 £ ml) of the upper hexane layer was passed through a silica gel column, and the liberated 14C-free fatty acids contained in 15 the flow-through fraction were quantitated in a scintillation counter. For both assays, MEM containing 10% FBS was used as a blank and conditioned media from COS7 cells transfected with an antisense plasmid (AS) was used as a negative control. <br><br> 20 Example 15 - Recombinant Adenovirus <br><br> Construction and Animal Studies <br><br> A recombinant adenovirus encoding human LIPG was constructed as described (Tsukamoto et al., J. Clin. Invest., 100, 107-114 (1997); Tsukamoto et al., J. Lipid Res., 38, 25 1869-1876 (1997)). In brief, the full-length human cDNA was subcloned into the shuttle plasmid vector pAdCMVLinkl. After screening for the appropriate orientation by restriction analysis, the plasmid was linearized with NheI and cotransfected into 293 cells along with adenoviral DNA 30 digested with Clal. Cells were overlaid with agar and incubated at 37°C for 15 days. Six plaques were picked and screened by PCR; two plaques positive for cDNA were subjected to a second round of plaque purification. After confirmation of the presence of cDNA, the recombinant adenovirus was 35 expanded in 293 cells at 37 °C. Cell lysates were used to infect HeLa cells for confirmation of the expression of human LIPG by western blot of conditioned media. The recombinant adenovirus (AdhEL) was further expanded in 293 cells and <br><br> o o <br><br> - 89 - <br><br> purified by cesium chloride ultracentrifugation. Control adenovirus containing no cDNA insert (Adnull) was also subjected to plaque purification and purified as described above. The purified viruses were stored in 10% glycerol/PBX 5 at -80 °C. Wild-type C57BL/6, human apoA-I transgenic and LDL receptor mutant mice were obtained from Jackson Laboratory. All mice were fed chow diets, wild-type and human apoA-I transgenic mice were injected intravenously via the tail vein with AdhEL or Adnull lxlO11 particles 10 (approximately 2xl09 pfu) and LDLR-deficient mice were injected with lxlO10 particles. In all experiments, blood was obtained from the retro-orbital plexus 1 day before injection and at multiple time points following injection. <br><br> Intravenous injection of AdhEL into wild-type C57BL/6 15 mice resulted in expression in the liver (Figure 17) and reduction of plasma levels of HDL cholesterol that remained significantly lower than control virus-injected mice through at least 41 days post-injection (Figure 18) . Lipoproteins were separated by FPLC gel filtration, demonstrating that HDL 20 was undetectable 14 days after adenovirus injection (Figure <br><br> 19). Injection of recombinant LIPG adenovirus into human apoA-I transgenic mice (which have much higher levels of HDL cholesterol and apoA-I) reduced both HDL cholesterol (Figure <br><br> 20) and apoA-I (Figure 21) levels. To determine the relative 25 effects of LIPG expression on HDL compared with the apoB- <br><br> containing lipoproteins VLDL and LDL, we injected a lower dose of the LIPG adenovirus into chow-fed LDL receptor-deficient mice, which have approximately 70% of cholesterol in VLDL/LDL and approximately 30% in HDL. As before, 30 expression of LIPG reduced HDL cholesterol levels (Figure 23). Although LIPG expression reduced VLDL/LDL cholesterol levels in the same mice (Figure 24), the effect was proportionately less. Overexpression of LIPG reduced VLDL/LDL cholesterol, therefore a role of LIPG in the 35 modulation of apoB-containing lipoproteins cannot be excluded. <br><br> o <br><br> - 90 - <br><br> Example 16 - Lipid/Lipoprotein Analyses <br><br> The plasma total cholesterol and HDL cholesterol levels were measured enzymatically on a Cobas Fara (Roche Diagnostic Systems) using Sigma reagents. ApoA-I was quantitated using 5 a turbidometric assay (Sigma) on a Cobas Fara. Pooled plasma samples were subjected to fast protein liquid chromatography (FPLC) gel filtration (Pharmacia LKB Biotechnology) using two Superose 6 columns in series as described (Tsukamoto et al., J. Clin. Investsupra) . Fractions (0.5 ml) were collected, 10 and cholesterol concentrations were determined using an enzymatic assay (Wako Pure Chemical Industries) . <br><br> O <br><br> Example 17 - Identification of Inhibitors of LIPG <br><br> Modulators of EL activity may be found using the following method: <br><br> 15 Recombinant LIPG would be purified from the conditioned medium of stably transfected Chinese hamster ovary cells, <br><br> from baculovirus infected insect cells, yeast (Pichia pastoris, Kluveromyces Lactis) or other sources. Non-recombinant sources of LIPG (such as human plasma, 20 endothelial cell conditioned media, etc.) could also be employed. An example of a primary screen to look for modulators of LIPG activity would utilize the soluble fluorescent substrate 4-methy lumber iferyl hepatanoate. This assay is continuous and homogeneous. Hydrolysis of this 25 substrate by LIPG results in the production of highly fluorescent 4-methy1umbelliferone that can be measured in a microplate fluorimeter. Other primary screening assay formats that could be used are a scintillation proximity assay (Amersham) that measures phospholipase activity, the 30 lower-throughput radiometric phospholipase assay described in Example 7 (and proposed below as a secondary assay), or the alternative phospholipase assays described in Example 8. <br><br> The catalytic center of LIPG, like other TG lipases, consists of the same catalytic triad (ser, his, asp) found in 35 serine proteases. Indeed, other TG lipases, such as lipoprotein lipase, are inhibited by serine protease <br><br> i 7 <br><br> - 91 - <br><br> inhibitors such as PMSF and DFP. Either one of these compounds may serve as a positive control for inhibitors of LIPG activity. <br><br> Secondary assay; Compounds active in the esterase assay or 5 alternative screening assays described above will be assayed in a standard, radiometric phospholipase A assay. This assay measures the release of radiolabelled palmitic acid from mixed micelles containing tl4C]-dipalmitoyl- <br><br> phosphatidylcholine. Other assay formats could be envisioned 10 which utilize fluorescent substrates and which would be amenable to a greater throughput. <br><br> Selectivity assays: Compounds would be assayed for inhibition of the related enzymes lipoprotein lipase (LPL) and pancreatic lipase (PL). Human PL and bovine LPL are 15 commercially available and assays could be readily implemented. The phospholipase activity of PL is measured in exactly the same way as described above for the secondary assay of LIPG. Since LPL is primarily a TG lipase, the secondary assay would measure radiolabelled fatty acid (oleic 20 acid) release from a radiolabelled TG (triolein) substrate (described in Example 7) . This assay has a similar capacity and may be adapted to other assay formats which utilize fluorescent substrates and which would be amenable to a greater throughput. <br><br> 25 Phospholipase activity of LIPG would be tested on its in vivo substrate, HDL, in an in vitro assay. Radiolabelled HDL could be generated by exchange with a radiolabelled phospholipid, and then used to measure LIPG phosphospholipase activity and the activity of compounds emerging from the 30 screens. <br><br> An additional assay could measure the impact of preincubation of LIPG, HDL, +/- compounds on radiolabelled cholesterol efflux from cultured cells such as the rat Fu5AH hepatoma line. <br><br> o o <br><br> ; f&lt; <br><br> - 92 - <br><br> In vivo assays for assessment of compounds can be run in wild-type, LIPG-overexpressing, and as control, LIPG null mice. If, as in the case of adenoEL expression, the transgenic mice exhibit decreased HDL relative to control 5 mice, then treatment of transgenic mice with LIPG inhibitory compounds would be expected to raise HDL to the levels of control mice. It is also possible that compounds could be tested for their LIPG inhibitory activity (elevation of HDL) in other animals such as the LDLR-/- mouse, apoAl transgenic 10 mice hamsters, or rabbits. Compounds which elevated LIPG or LIPG activity would be expected to raise HDL in these or other animal models. <br><br> Example 18 - Inhibitory Small Molecule Treatment Method <br><br> A small molecule (hereafter an "inhibitory small 15 molecule") identified in the screening outlined in Example 17 as able to inhibit the LIPG polypeptide in vitro is tested for its ability to inhibit the LIPG polypeptide in vivo. Wild-type and LIPG transgenic mice will be studied by administering the small molecule orally (if orally 20 bioavailable) or by intravenous injection. Activity of the LIPG polypeptide will be measured in plasma before and after heparin injection (to release the enzyme from bound sites). In addition, cholesterol, VLDL, LDL and HDL cholesterol and apoA-I levels will be monitored in animals receiving the 25 inhibitory small molecule. Finally, LDL receptor deficient mice will be fed an atherogenic diet and administered the inhibitory small molecule or placebo for a period of 8 weeks. Atherosclerosis will be quantitated in the aortas of the mice in order to determine whether administration of the 30 inhibitory small molecule recudes the progression or induces regression of atherosclerosis. Based on these preclinical data, additional animal models such as hamsters, rabbits, or pigs will be studied for the ability of the inhibitory small molecule to raise HDL cholesterol levels, reduce VLDL and LDL 35 cholesterol levels, and/or inhibit the progression of atherosclerosis. <br><br> o o <br><br> - 93 - <br><br> Those inhibitory small molecules found to have the desired properties will be administered to patients in combination with pharmaceutically acceptable carriers. The inhibitory small molecules may be administered in a variety 5 of ways, including oral administration and intravenous injection. The patients1 HDL, VLDL and LDL cholesterol levels will be monitored to determine efficacy of the inhibitory small molecule and to optimize dosage and administration protocols. <br><br> 10 Example 19 - Inhibitory Peptide Treatment Method <br><br> Therapeutic peptides are identified by testing fragments of the LIPG polypeptide to determine which of these fragments inhibit LIPG polypeptide activity in vitro. Once identified, an "inhibitory peptide" is then tested for its ability to 15 inhibit the LIPG polypeptide in vivo. Inhibitory peptides will be produced recombinantly in E.coli and purified by methods known in the art. The effect of the inhibitory peptides will be studied in wild-type and LIPG transgenic mice by administering the inhibitory peptide by intravenous 20 injection. Activity of the LIPG polypeptide will be measured in plasma before and after heparin injection (to release the enzyme from bound sites) . In addition, cholesterol, VLDL, LDL and HDL cholesterol and apoA-I levels will be monitored in animals receiving the inhibitory peptide. Finally, LDL 25 receptor deficient mice will be fed an atherogenic diet and administered the inhibitory peptide or placebo for a period of 8 weeks. Atherosclerosis will be quantitated in the aortas of the mice in order to determine whether administration of the inhibitory peptide reduces the 30 progression or induces regression of atherosclerosis. Based on these preclinical data, additional animal models such as hamsters, rabbits or pigs will be studied for the ability of the inhibitory small molecule to raise HDL cholesterol levels, reduce VLDL and LDL cholesterol levels, and/or 35 inhibit the progression of atherosclerosis. <br><br> Those inhibitory peptides found to have the desired properties will be administered to patients in combination <br><br> o o <br><br> - 94 - <br><br> with pharmaceutically acceptable carriers. The inhibitory peptides may be administered in a variety of ways, including oral administration and intravenous injection. The patients' HDL, VLDL and LDL cholesterol levels will be monitored to 5 determine efficacy of the inhibitory peptides and to optimize dosage and administration protocols. <br><br> Example 20 - Antisense Treatment Method <br><br> A series of antisense oligonucleotides, each complementary to about 20 bases of the LIPG cDNA sequence are 10 chemically synthesized by standard techniques. To determine the most efficient oligonucleotide to use therapeutically, each oligonucleotide is individually transfected into cells expressing the LIPG gene, using standard transfection protocols. <br><br> 15 At about 24-48 hours following transfection of the oligonucleotides, the LIPG mRNA level in cells is determined by quantitative PCR, northern blot, RNAse protection, or other appropriate methods. Alternatively, LIPG expression may be monitored with specific antibodies, which can be used 20 to screen for effective antisense oligonucleotides. <br><br> Oligonucleotides which effectively reduce LIPG mRNA levels are then formulated for in vivo delivery as therapeutics. <br><br> Antisense LIPG sequences may be delivered in a gene therapy vector, such as adenovirus, adeno-associated virus, 25 retrovirus, naked DNA, or other systems discussed in the detailed description. Such fragments can be used therapeutically when delivered in gene therapy, vectors. Hepatic expression of such recombinant vectors is a preferred approach. Alternatively, synthetic antisense 30 oligonucleotides may be formulated for in vivo delivery as therapeutics as described above. <br><br> Antisense oligonucleotides may be administered by the following routes: intravenous, subcutaneous, introdermal, pulmonary, oral, intraventricular, intrathecal, and topical. 35 The route of administration may include direct administration <br><br> o <br><br> - 95 - <br><br> to vessel walls (i.e., endothelium and/or vascular smooth muscle). As an example, patients with low HDL-C could receive a dose of 0.5-2 mg/kg of an effective antisense oligonucleotide, infused intravenously, every other day for 5 up to 2-3 weeks. As LIPG is expressed in the liver, it may be desireable to deliver antisense reagents to the portal circulation. This may be accomplished by conjugating or complexing the oligonucleotide with a liver-targeting moiety, such as asialoglycoprotein. Dose and timing of therapy would 10 depend on efficiency of antisense delivery, as well as parameters such as half life, specificity and toxicology of the antisense oligonucleotide. <br><br> Increase in HDL-C can be monitored using standard clinical laboratory procedures. The original dosing schedule 15 (such as that described above) is repeated as often as required to maintain HDL-C above 35 mg/dL. <br><br> Example 21 - Ribozvme Treatment Method <br><br> Based on the LIPG cDNA sequence, hammerhead ribozymes which effectively reduce LIPG mRNA levels are prepared. 20 These consist of two "arms" of 6-7 bases each of nucleotide sequence complementary to LIPG mRNA, separated by the catalytic moiety of the ribozyme. Examples of such hammerhead motifs are described by Rossi et al., 1992, Aids Research and Human Retroviruses, 8, 183. The ribozymes are 25 expressed in eukaryotic cells from an appropriate DNA vector. <br><br> The ribozymes may be administered encapsulated in liposomes, as discussed above. <br><br> The ribozyme/liposome composition is delivered to the liver by direct injection or by use of a catheter, infusion 30 pump or stent. The route of administration may include direct administration to vessel walls (i.e., endothelium and/or vascular smooth muscle). Patients are treated for up to 2 weeks with 5-50 mg/kg/day ribozyme in a pharmaceutically effective carrier. Increase in HDL-C and dosing regimen are 35 monitored and determined as for antisense oligonucleotides. <br><br> o O <br><br> % ; <br><br> - 96 - <br><br> Example 22 - Neutralizing Antibody Treatment Method <br><br> Anti-LIPG antibodies, antibody fragments, or chimeric antibodies consisting of at least one LIPG-binding moiety, prepared as described in Example 12, are used to inhibit LIPG 5 activity in vivo. The antibodies may be delivered as a bolus only, infused over time, or both. Typically a dose of 0.2-0.6 mg/kg is given as bolus, followed by a 2 to 12-hour infusion. Alternatively, multiple bolus injections are administered every other day, or every third of fourth day, 10 as required to reduce LIPG and raise HDL-C. Repeat dosing is performed as determined by measurement of HDL-C levels. Antibodies to LIPG may also be delivered in a gene therapy vehicle to facilitate expression in vivo. The level of expression of the antibody is determined indirectly by 15 measuring HDL-C levels and additional vectors may be introduced as needed. <br><br> Example 23 - Use of Inhibitory <br><br> Molecules or Enhancer Molecules <br><br> Fragments of LIPG protein, which can inhibit LIPG 20 activity by competing for binding to intact LIPG, required coactivator molecules, cell surface receptors or binding proteins, may be delivered as therapeutic recombinant proteins or from gene therapy vectors. <br><br> As an example, the LLGN polypeptide based on LIPG is 25 cloned into a recombinant adenovirus as described (Tsukamoto et al., J. Clin. Invest., 100, 107-114 (1997); Tsukamoto et al., J. Lipid Res., 38, 1869-1876 (1997)). The LLGN cDNA is cloned into the shuttle plasmid vector pAdCMVLinkl. After screening for the appropriate orientation by restriction 30 analysis, the plasmid is linearized with NheI and cotransfected into 293 cells along with adenoviral DNA digested with Clal. Cells are then overlaid with agar and incubated at 37°C for 15 days. Plaques are picked and screened by PCR; plaques positive for cDNA are subjected to a 35 second round of plaque purification. After confirmation of the presence of cDNA, the recombinant adenovirus is expanded in 293 cells at 37°C. Cell lysates are used to infect HeLa <br><br> o o <br><br> - 97 - <br><br> cells for confirmation of the expression of human EL by western blot of conditioned media. The recombinant adenovirus is further expanded in 293 cells and purified by cesium chloride ultracentrifugation. The purified viruses are 5 stored in 10% glycerol/PBX at -80°C. The patient is injected intravenously with the recombinant adenovirus lxlO11 particles (approximately 2xl09 pfu) . <br><br> Example 24 - Methods of Increasing the <br><br> Level of LIPG in a Patient by 10 Expression of LIPG from an Expression Vector <br><br> The full length LIPG cDNA is cloned into a recombinant adenovirus (Tsukamoto et al., J. Clin. Invest., 100, 107-114 (1997); Tsukamoto et al., J. Lipid Res., 38, 1869-1876 (1997)) encoding human LIPG. The full-length human LIPG cDNA 15 is cloned into the shuttle plasmid vector pAdCMVLinkl. After screening for the appropriate orientation by restriction analysis, the plasmid is linearized with Nhel and cotransfected into 293 cells along with adenoviral DNA digested with Clal. Cells are overlaid with agar and 20 incubated at 37°C for 15 days. Plaques are picked and screened by PCR; plaques positive for cDNA are subjected to a second round of plaque purification. After confirmation of the presence of cDNA, the recombinant adenovirus is expanded in 293 cells at 37°C. Cell lysates are used to infect HeLa 25 cells for confirmation of the expression of human LIPG polypeptide by western blot of conditioned media. The recombinant adenovirus (AdhEL) is further expanded in 293 cells and purified by cesium chloride ultracentrifugation. l*he purified viruses are stored in 10% glycerol/PBX at -80°C. 30 Patients are injected intravenously with AdhEL or Adnull lxlO11 particles (approximately 2xl09 pfu) . <br><br> Example 25 - Methods of Increasing the Level of LIPG Activity by Administration of a Full-Length Wild-Type or 35 Engineered Recombinant LIPG Protein <br><br> The wild-type LIPG protein reduces VLDL and LDL cholesterol levels and LIPG could be engineered to act specifically on VLDL and LDL cholesterol without having <br><br> I <br><br> - 98 - <br><br> effects on HDL cholesterol. Administration of wild-type or engineered recombinant LIPG could in certain circumstances be used as a therapy for reducing VLDL and/or LDL cholesterol levels. Wild-type and/or engineered LIPG protein 5 ("recombinant LIPG protein") will be produced recombinantly in E.coli and purified using methods known in the art. Wild-type mice will be studied by administering the recombinant LIPG protein by intravenous injection. Activity of LIPG will be measured in plasma. In addition, cholesterol, VLDL, LDL 10 and HDL cholesterol and apoA-I levels will be monitored in animals receiving the recombinant LIPG protein. Finally, LDL receptor deficient mice will be fed an atherogenic diet and administered the recombinant LIPG protein or placebo for a period of 8 weeks. Atherosclerosis will be quantitated in 15 the aortas of the mice in order to determine whether admins itr at ion of the recombinant LIPG protein reduces the progression or induces regression of atherosclerosis. Based on these preclinical data, additional animal models such as hamsters, rabbits, or pigs will be studied for the ability of 20 the recombinant LIPG protein to reduce VLDL and LDL cholesterol levels and/or inhibit the progression of atherosclerosis. Those recombinant LIPG proteins found to have the desired ability to reduce VLDL and LDL cholesterol levels and/or inhibit the progression of atherosclerosis will 25 be combined with a pharmaceutically acceptable carrier and administered to patients. The recombinant LIPG polypeptides may be administered in a variety of ways, including oral administration and intravenous injection. <br><br> All the references discussed herein are incorporated by 30 reference. <br><br> One skilled in the art will readily appreciate the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The peptides, polynucleotides, 35 methods, procedures and techniques described herein are presented as representative of the preferred embodiments, and intended to be exemplary and not intended as limitations on <br><br> o o <br><br> - 99 - <br><br> the scope of the present invention. Changes therein and other uses will occur to those of skill in the art which are encompassed within the spirit of the invention or defined by the scope of the appended claims. <br><br> 5 Example 26 - Demonstration of Glycosylation and Biological Activity of LIPG <br><br> A number of experiments were conducted to further characterize LIPG. The experiments which follow further define the biological activity of LIPG (referred to in these 10 experiments and in the figures as "endothelial lipase," or "EL"). <br><br> Figure 24 demonstrates that endothelial lipase is a glycoprotein. EL was treated v/ith the glycosidases EndoF, EndoH, and neuraminidase. This resulted in reductions in the 15 size of the EL band on western blotting, consistent with the removal of carbohydrate from the polypeptide chain. Glycosylation was also inhibited with tunicamycin in EL-expressing cells. This resulted in a substantial reduction in the size of the EL band. The glycosidase and tunicamycin 20 experiments indicate that EL is a glycoprotein. <br><br> Figure 25 demonstrates that endothelial lipase is a heparin binding protein. An adenoviral vector expressing EL AdhEL (see Example 15) was injected into mice. Blood was drawn before (pre) and after (post) heparin injection. The 25 intensity of the full-length 68 kD EL band greatly increased after heparin injection consistent with EL bound to heparin sulfate proteoglycans and released by the administration of heparin. <br><br> Figures 26A and 26B and 27A and 27B illustrate the 30 lipolytic activity of endothelial lipase relative to lipoprotein lipase (LPL) and hepatic lipase (HL). This experiment was conducted in order to determine whether endothelial lipase has triglyceride (TG) lipase activity (Fig. 27A). In multiple studies on different conditioned 35 media, unequivocal evidence of TG lipase activity was <br><br> o o <br><br> - 100 - <br><br> repeatedly found. The phospholipase activity of endothelial lipase was assayed in the same conditioned media samples (Fig. 27B). The ratio of phospholipase to TG lipase activity was highly consistent as assayed in different batches of 5 conditioned media. In absolute terms, the moles of fatty acid generated per volume conditioned media was significantly greater for phospholipase than for TG lipase activity. The ability of serum to effect endothelial lipase activity was then tested (Figures 27A and 27B) . As expected, the addition 10 of serum significantly increased lipoprotein lipase TG lipase activity in the absence of any other source of apoC-II. Interestingly, serum reproduci.bly and substantially reduced the activity of endothelial lipase. This confirms that neither apoC-II nor any other serum factor is a required co-15 factor for endothelial lipase. It also indicates that there is an endogenous inhibitor of endothelial lipase in serum. <br><br> Figures 28A and 28B illustrate that expression of EL increased post-heparin plasma phospholipase activity in a dose dependent fashion. Injection of three different doses 20 (lxlO11, 3xl010 or lxlO50 particles) of AdhEL into mice resulted in the presence of EL protein in the post-heparin plasma. Major bands of 68 kD and 40 kD and a minor band of 55 kD were observed (Fig. 28A). The 68 kD band is believed to be the full-length glycosylated form of EL and the 40 kD 25 band a proteolytic fragment. The amount of EL protein in plasma assessed by western blot is proportional to the dose of vector injected (Fig. 28B). The expression of different levels of human EL resulted in a significant and dose-dependent increase in post-heparin plasma phospholipase 30 activity, compared to Adnull injected mice (9779 +/- 733, 6501 +/- 1299, 3963 +/- 796 maol.ml*1 .hr"1 vs 952 +/- 258 nmol.mo-1.hr-1, respectively). The post-heparin phospholipase activity correlated with levels of human EL protein for the 68 kD (r=0.98, P&lt;0.01), 55 kD (r=0.83, P&lt;0.05), and 40 kD 35 (r=0.94, P&lt;0.05) bands. <br><br> o <br><br> - 101 -SEQUENCE LISTING <br><br> (1) GENERAL INFORMATION: <br><br> (i) APPLICANTS Jaye, Michael C. <br><br> Doan, Kim-Anh T. <br><br> Krawiec, John A. <br><br> Lynch, Kevin J. <br><br> Amin, Dilip V. <br><br> South, Victoria J. <br><br> Marchadier, Dawn Haugeais, Cyrille Rader, Daniel J. <br><br> (ii) TITLE OF INVENTION: COMPOSITIONS AND METHODS FOR EFFECTING THE <br><br> LEVELS OF HIGH DENSITY LIPOPROTEIN (HDL) CHOLESTEROL AND APOLIPOPROTEIN AI, VERY LOW DENSITY LIPOPROTEIN (VLDL) CHOLESTEROL AND LOW DENSITY LIPOPROTEIN (LDL) CHOLESTEROL <br><br> (iii) NUMBER OF SEQUENCES: 31 <br><br> (iv) CORRESPONDENCE ADDRESS: <br><br> (A) ADDRESSEE: Synnestvedt &amp; Lechner LLP <br><br> (B) STREET: Suite 2600 Aramark Tower, 1101 Market Street <br><br> (C) CITY: Philadelphia <br><br> (D) STATE: PA <br><br> (E) COUNTRY: USA <br><br> (F) ZIP: 19107 <br><br> (V) COMPUTER READABLE FORM: <br><br> (A) MEDIUM TYPE: Floppy disk <br><br> (B) COMPUTER: IBM PC compatible <br><br> (C) OPERATING SYSTEM: PC-Windows 95 <br><br> (D) SOFTWARE: Corel WordPerfect 8.0 converted to ASCII (vi) CURRENT APPLICATION DATA: <br><br> (A) APPLICATION NUMBER: <br><br> (B) FILING DATE: <br><br> (C) CLASSIFICATION: <br><br> (viii) ATTORNEY/AGENT INFORMATION: <br><br> (A) NAME: Kelly, Ph.D., Patrick J. <br><br> (B) REGISTRATION NUMBER: 34,638 <br><br> (C) REFERENCE/DOCKET NUMBER: 22,944-D PCT (ix) TELECOMMUNICATION INFORMATION: <br><br> (A) TELEPHONE: (215)923-4466 <br><br> (B) TELEFAX: (215)923-2189 <br><br> o <br><br> - 102 - <br><br> (2) INFORMATION FOR SEQ ID NO: Is <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 367 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 22-.180 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l: <br><br> GAATTCGGCT TGATCAATCG C TTC AAA AAG GGG ATC TGT CTG AGC TGC CGC 51 <br><br> Phe Lys Lys Gly lie Cys Leu Ser Cys Arg 15 10 <br><br> AAG AAC CGT TGT AAT AGC ATT GGC TAC AAT GCC AAG AAA ATG AGG AAC 99 <br><br> Lys Asn Arg Cys Asn Ser lie Gly Tyr Asn Ala Lys Lys Met Arg Asn <br><br> 15 20 25 <br><br> AAG AGG AAC AGC AAA ATG TAC CTA AAA ACC CGG GCA GGC ATG CCT TTC 147 <br><br> Lys Arg Asn Ser Lys Met Tyr Leu Lys Thr Arg Ala Gly Met Pro Phe <br><br> 30 35 40 <br><br> AGA GGT AAC CTT CAG TCC CTG GAG TGT CCC TGA GGAAGGCCCT TAATACCTCC 200 <br><br> Arg Gly Asn Leu Gin Ser Leu Glu Cys Pro * <br><br> 45 50 <br><br> TTCTTAATAC CATGCTGCAG AGCAGGGCAC ATCCTAGCCC AGGAGAAGTG GCCAGCACAA 260 TCCAATCAAA TCGTTGCAAA TCAGATTACA CTGTGCATGT CCTAGGAAAG GGAATCTTTA 320 CAAAATAAAC AGTGTGGACC CCTCAAAAAA AAAAAAAAGC CGAATTC 367 <br><br> (2) INFORMATION FOR SEQ ID NO:2: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 53 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: <br><br> Phe Lys Lys Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Ser <br><br> 1 5 10 15 <br><br> lie Gly Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met <br><br> 20 25 30 <br><br> Tyr Leu Lys Thr Arg Ala Gly Met Pro Phe Arg Gly Asn Leu Gin Ser <br><br> (.) <br><br> ( ) <br><br> - 103 - <br><br> 35 40 45 <br><br> Leu Glu Cys Pro * <br><br> 50 <br><br> (2) INFORMATION FOR SEQ ID NO: 3: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 1382 base pairs <br><br> (B) TYPE: nucleic acid ' <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 312..1370 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: <br><br> GAATTCGGCT TCTACTACTA CTAGGCCACG CGTCGCCTAG TACGGGGGGG GGGGGGGGGG 60 <br><br> TCAGCGAGTC CTTGCCTCCC GGCGGCTCAG GACGAGGGCA GATCTCGTTC TGGGGCAAGC 120 <br><br> CGTTGACACT CGCTCCCTGC CACCGCCCGG GCTCCGTGCC GCCAAGTTTT CATTTTCCAC 180 <br><br> CTTCTCTGCC TCCAGTCCCC CAGCCCCTGG CCGAGAGAAG GGTCTTACCG GCCGGGATTG 240 CTGGAAACAC CAAGAGGTGG TTTTTGTTTT TTAAAACTTC TGTTTCTTGG GAGGGGGTGT 300 <br><br> GGCGGGGCAG G ATG AGC AAC TCC GTT CCT CTG CTC TGT TTC TGG AGC CTC 350 <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu 55 60 65 <br><br> TGC TAT TGC TTT GCT GCG GGG AGC CCC GTA CCT TTT GGT CCA GAG GGA 398 <br><br> Cys Tyr Cys Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly <br><br> 70 75 80 <br><br> CGG CTG GAA GAT AAG CTC CAC AAA CCC AAA GCT ACA CAG ACT GAG GTC 446 <br><br> Arg Leu Glu Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val <br><br> 85 90 95 <br><br> AAA CCA TCT GTG AGG TTT AAC CTC CGC ACC TCC AAG GAC CCA GAG CAT 494 <br><br> Lys Pro Ser Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His <br><br> 100 105 110 <br><br> GAA GGA TGC TAC CTC TCC GTC GGC CAC AGC CAG CCC TTA GAA GAC TGC 542 <br><br> Glu Gly Cys Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys 115 120 125 130 <br><br> AGT TTC AAC ATG ACA GCT AAA ACC TTT TTC ATC ATT CAC GGA TGG ACG 590 <br><br> Ser Phe Asn Met Thr Ala Lys Thr Phe Phe He lie His Gly Trp Thr 135 140 145 <br><br> o o <br><br> - 104 - <br><br> ATG AGC GGT ATC TTT GAA AAC TGG CTG CAC AAA CTC GTG TCA GCC CTG 638 <br><br> Met Ser Gly lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu <br><br> 150 155 160 <br><br> CAC ACA AGA GAG AAA GAC GCC AAT GTA GTT GTG GTT GAC TGG CTC CCC 686 <br><br> His Thr Arg Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro <br><br> 165 170 175 <br><br> CTG GCC CAC CAG CTT TAC ACG GAT GCG GTC AAT AAT ACC AGG GTG GTG 734 <br><br> Leu Ala His Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val <br><br> 180 185 190 <br><br> GGA CAC AGC ATT GCC AGG ATG CTC GAC TGG CTG CAG GAG AAG GAC GAT 782 <br><br> Gly His Ser lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp 195 200 205 210 <br><br> TTT TCT CTC GGG AAT GTC CAC TTG ATC GGC TAC AGC CTC GGA GCG CAC 830 <br><br> Phe Ser Leu Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His <br><br> 215 220 225 <br><br> GTG GCC GGG TAT GCA GGC AAC TTC GTG AAA GGA ACG GTG GGC CGA ATC 878 <br><br> Val Ala Gly Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie <br><br> 230 235 240 <br><br> ACA GGT TTG GAT CCT GCC GGG CCC ATG TTT GAA GGG GCC GAC ATC CAC 926 <br><br> Thr Gly Leu Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His <br><br> 245 250 255 <br><br> AAG AGG CTC TCT CCG GAC GAT GCA GAT TTT GTG GAT GTC CTC CAC ACC 974 <br><br> Lys Arg Leu Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr <br><br> 260 265 270 <br><br> TAC ACG CGT TCC TTC GGC TTG AGC ATT GGT ATT CAG ATG CCT GTG GGC 1022 <br><br> Tyr Thr Arg Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly 275 280 285 290 <br><br> CAC ATT GAC ATC TAC CCC AAT GGG GGT GAC TTC CAG CCA GGC TGT GGA 1070 <br><br> His lie Asp lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly <br><br> 295 300 305 <br><br> CTC AAC GAT GTC TTG GGA TCA ATT GCA TAT GGA ACA ATC ACA GAG GTG 1118 <br><br> Leu Asn Asp Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val <br><br> 310 315 320 <br><br> GTA AAA TGT GAG CAT GAG CGA GCC GTC CAC CTC TTT GTT GAC TCT CTG 1166 <br><br> Val Lys Cys Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu <br><br> 325 330 335 <br><br> GTG AAT CAG GAC AAG CCG AGT TTT GCC TTC CAG TGC ACT GAC TCC AAT 1214 <br><br> Val Asn Gin Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn <br><br> 340 345 350 <br><br> CGC TTC AAA AAG GGG ATC TGT CTG AGC TGC CGC AAG AAC CGT TGT AAT 1262 <br><br> o o <br><br> Arg Phe Lys Lys 355 <br><br> AGC ATT GGC TAC Ser lie Gly Tyr <br><br> ATG TAC CTA AAA Met Tyr Leu Lys 390 <br><br> TCC CTG GAG TGT Ser Leu Glu Cys 405 <br><br> Gly lie Cys Leu 360 <br><br> AAT GCC AAG AAA Asn Ala Lys Lys 375 <br><br> ACC CGG GCA GGC Thr Arg Ala Gly <br><br> CAAGCCGAAT TC <br><br> - 105 - <br><br> Ser Cys Arg Lys 365 <br><br> ATG AGG AAC AAG Met Arg Asn Lys 380 <br><br> ATG CCT TTC AGA Met Pro Phe Arg 395 <br><br> Asn Arg Cys Asn 370 <br><br> AGG AAC AGC AAA Arg Asn Ser Lys 385 <br><br> GGT AAC CTT CAG Gly Asn Leu Gin 400 <br><br> (2) INFORMATION FOR SEQ ID NO:4: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 353 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein <br><br> o o <br><br> - 106 - <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 15 10 15 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Leu Glu <br><br> 20 25 30 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser <br><br> 35 40 45 <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys <br><br> 50 55 60 <br><br> Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys Ser Phe Asn 65 70 75 80 <br><br> Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr Met Ser Gly <br><br> 85 90 95 <br><br> lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu His Thr Arg <br><br> 100 105 110 <br><br> Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro Leu Ala His <br><br> 115 120 125 <br><br> Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val Gly His Ser <br><br> 130 135 140 <br><br> lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp Phe Ser Leu 145 150 155 160 <br><br> Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His Val Ala Gly <br><br> 165 170 175 <br><br> Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie Thr Gly Leu <br><br> 180 185 190 <br><br> Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His Lys Arg Leu <br><br> 195 200 205 <br><br> Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr Tyr Thr Arg 210 215 220 <br><br> o o <br><br> - 107 - <br><br> Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly His lie Asp 225 230 235 240 <br><br> lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly Leu Asn Asp <br><br> 245 250 255 <br><br> Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val Val Lys Cys <br><br> 260 265 270 <br><br> Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin <br><br> 275 280 285 <br><br> Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys <br><br> 290 295 300 <br><br> Lys Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Ser lie Gly 305 310 315 320 <br><br> Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met Tyr Leu <br><br> 325 330 335 <br><br> Lys Thr Arg Ala Gly Met Pro Phe Arg Gly Asn Leu Gin Ser Leu Glu 340 345 350 <br><br> Cys <br><br> (2) INFORMATION FOR SEQ ID NO:5: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 1065 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 1..1065 <br><br> o <br><br> - 108 - <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5: <br><br> ATG AGC AAC TCC GTT CCT CTG CTC TGT TTC TGG AGC CTC TGC TAT TGC 48 <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 355 360 365 <br><br> TTT GCT GCG GGG AGC CCC GTA CCT TTT GGT CCA GAG GGA CGG CTG GAA 96 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Leu Glu 370 375 380 385 <br><br> GAT AAG CTC CAC AAA CCC AAA GCT ACA CAG ACT GAG GTC AAA CCA TCT 144 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser <br><br> 390 395 400 <br><br> GTG AGG TTT AAC CTC CGC ACC TCC AAG GAC CCA GAG CAT GAA GGA TGC 192 <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys <br><br> 405 410 415 <br><br> TAC CTC TCC GTC GGC CAC AGC CAG CCC TTA GAA GAC TGC AGT TTC AAC 240 <br><br> Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys Ser Phe Asn <br><br> 420 425 430 <br><br> ATG ACA GCT AAA ACC TTT TTC ATC ATT CAC GGA TGG ACG ATG AGC GGT 288 <br><br> Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr Met Ser Gly <br><br> 435 440 445 <br><br> ATC TTT GAA AAC TGG CTG CAC AAA CTC GTG TCA GCC CTG CAC ACA AGA 336 <br><br> lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu His Thr Arg 450 455 460 465 <br><br> GAG AAA GAC GCC AAT GTA GTT GTG GTT GAC TGG CTC CCC CTG GCC CAC 384 <br><br> Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro Leu Ala His <br><br> 470 475 480 <br><br> CAG CTT TAC ACG GAT GCG GTC AAT AAT ACC AGG GTG GTG GGA CAC AGC 432 <br><br> Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val Gly His Ser 485 490 495 <br><br> o o <br><br> ATT GCC AGG ATG lie Ala Arg Met 500 <br><br> GGG AAT GTC CAC Gly Asn Val His 515 <br><br> TAT GCA GGC AAC Tyr Ala Gly Asn 530 <br><br> GAT CCT GCC GGG Asp Pro Ala Gly <br><br> TCT CCG GAC GAT Ser Pro Asp Asp 565 <br><br> TCC TTC GGC TTG Ser Phe Gly Leu 580 <br><br> ATC TAC CCC AAT lie Tyr Pro Asn 595 <br><br> GTC TTG GGA TCA Val Leu Gly Ser 610 <br><br> GAG CAT GAG CGA Glu His Glu Arg <br><br> GAC AAG CCG AGT Asp Lys Pro Ser 645 <br><br> AAG GGG ATC TGT Lys Gly lie Cys 660 <br><br> TAC AAT GCC AAG Tyr Asn Ala Lys 675 <br><br> AAA ACC CGG GCA Lys Thr Arg Ala 690 <br><br> TGT CCC TGA <br><br> CTC GAC TGG CTG Leu Asp Trp Leu 505 <br><br> TTG ATC GGC TAC Leu lie Gly Tyr 520 <br><br> TTC GTG AAA CGA Phe Val Lys Gly 535 <br><br> CCC ATG TTT GAA Pro Met Phe Glu 550 <br><br> GCA GAT TTT GTG Ala Asp Phe Val <br><br> AGC ATT GGT ATT Ser lie Gly lie 585 <br><br> GGG GGT GAC TTC Gly Gly Asp Phe 600 <br><br> ATT GCA TAT GGA lie Ala Tyr Gly 615 <br><br> GCC GTC CAC CTC Ala Val His Leu 630 <br><br> TTT GCC TTC CAG Phe Ala Phe Gin <br><br> CTG AGC TGC CGC Leu Ser Cys Arg 665 <br><br> AAA ATG AGG AAC Lys Met Arg Asn 680 <br><br> GGC ATG CCT TTC Gly Met Pro Phe 695 <br><br> - 109 - <br><br> CAG GAG AAG GAC Gin Glu Lys Asp <br><br> AGC CTC GGA GCG Ser Leu Gly Ala 525 <br><br> ACG GTG GGC CGA Thr Val Gly Arg 540 <br><br> GGG GCC GAC ATC Gly Ala Asp lie <br><br> 555 <br><br> GAT GTC CTC CAC Asp Val Leu His 570 <br><br> CAG ATG CCT GTG Gin Met Pro Val <br><br> CAG CCA GGC TGT Gin Pro Gly Cys 605 <br><br> ACA ATC ACA GAG Thr lie Thr Glu 620 <br><br> TTT GTT GAC TCT Phe Val Asp Ser 635 <br><br> TGC ACT GAC TCC Cys Thr Asp Ser 650 <br><br> AAG AAC CGT TGT Lys Asn Arg Cys <br><br> AAG AGG AAC AGC Lys Arg Asn Ser 685 <br><br> AGA GGT AAC CTT Arg Gly Asn Leu 700 <br><br> GAT TTT TCT CTC Asp Phe Ser Leu 510 <br><br> CAC GTG GCC GGG His Val Ala Gly <br><br> ATC ACA GGT TTG lie Thr Gly Leu 545 <br><br> CAC AAG AGG CTC His Lys Arg Leu 560 <br><br> ACC TAC ACG CGT Thr Tyr Thr Arg 575 <br><br> GGC CAC ATT GAC Gly His lie Asp 590 <br><br> GGA CTC AAC GAT Gly Leu Asn Asp <br><br> GTG GTA AAA TGT Val Val Lys Cys 625 <br><br> CTG GTG AAT CAG Leu Val Asn Gin 640 <br><br> AAT CGC TTC AAA Asn Arg Phe Lys 655 <br><br> AAT AGC ATT GGC Asn Ser lie Gly 670 <br><br> AAA ATG TAC CTA Lys Met Tyr Leu <br><br> CAG TCC CTG GAG Gin Ser Leu Glu 705 <br><br> o <br><br> - no - <br><br> Cys Pro * <br><br> (2) INFORMATION FOR SEQ ID NO:6: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 355 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 15 10 15 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Leu Glu <br><br> 20 25 30 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser <br><br> 35 40 45 <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys <br><br> 50 55 60 <br><br> Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys Ser Phe Asn 65 70 75 80 <br><br> i <br><br> o <br><br> 111 - <br><br> Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr Met Ser Gly <br><br> 85 90 95 <br><br> lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu His Thr Arg <br><br> 100 105 110 <br><br> Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro Leu Ala His <br><br> 115 120 125 <br><br> Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val Gly His Ser <br><br> 130 135 140 <br><br> lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp Phe Ser Leu 145 150 155 160 <br><br> Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His Val Ala Gly <br><br> 165 170 175 <br><br> Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie Thr Gly Leu <br><br> 180 185 190 <br><br> Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His Lys Arg Leu <br><br> 195 200 205 <br><br> Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr Tyr Thr Arg <br><br> 210 215 220 <br><br> Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly His lie Asp 225 230 235 240 <br><br> lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly Leu Asn Asp <br><br> 245 250 255 <br><br> Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val Val Lys Cys <br><br> 260 265 270 <br><br> Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin <br><br> 275 280 285 <br><br> Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys <br><br> 290 295 300 <br><br> Lys Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Ser lie Gly 305 310 315 320 <br><br> Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met Tyr Leu <br><br> 325 330 335 <br><br> Lys Thr Arg Ala Gly Met Pro Phe Arg Gly Asn Leu Gin Ser Leu Glu 340 345 350 <br><br> Cys Pro * <br><br> 355 <br><br> (2) INFORMATION FOR SEQ ID NO:7: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 2565 base pairs <br><br> o <br><br> 112 <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 252..1754 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: <br><br> GAATTCGCGG CCGCGTCGAC GGCGGCTCAG GACGAGGGCA GATCTCGTTC TGGGGCAAGC 60 <br><br> CGTTGACACT CGCTCCCTGC CACCGCCCGG GCTCCGTGCC GCCAAGTTTT CATTTTCCAC 120 <br><br> CTTCTCTGCC TCCAGTCCCC CAGCCCCTGG CCGAGAGAAG GGTCTTACCG GCCGGGATTG 180 <br><br> CTGGAAACAC CAAGAGGTGG TTTTTGTTTT TTAAAACTTC TGTTTCTTGG GAGGGGGTGT 240 <br><br> GGCGGGGCAG G ATG AGC AAC TCC GTT CCT CTG CTC TGT TTC TGG AGC CTC 290 <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu <br><br> 360 <br><br> 365 <br><br> TGC TAT TGC TTT GCT GCG GGG AGC CCC GTA CCT TTT GGT CCA GAG GGA <br><br> 338 <br><br> Cys Tyr Cys Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly <br><br> 370 <br><br> 375 <br><br> 380 <br><br> o o <br><br> - 113 - <br><br> CGG CTG GAA GAT AAG CTC CAC AAA CCC AAA GCT ACA CAG ACT GAG GTC 386 <br><br> Arg Leu Glu Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val <br><br> 385 390 395 400 <br><br> AAA CCA TCT GTG AGG TTT AAC CTC CGC ACC TCC AAG GAC CCA GAG CAT 434 <br><br> Lys Pro Ser Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His <br><br> 405 410 415 <br><br> GAA GGA TGC TAC CTC TCC GTC GGC CAC AGC CAG CCC TTA GAA GAC TGC 482 <br><br> Glu Gly Cys Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys <br><br> 420 425 430 <br><br> AGT TTC AAC ATG ACA GCT AAA ACC TTT TTC ATC ATT CAC GGA TGG ACG 530 <br><br> Ser Phe Asn Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr <br><br> 435 440 445 <br><br> ATG AGC GGT ATC TTT GAA AAC TGG CTG CAC AAA CTC GTG TCA GCC CTG 578 <br><br> Met Ser Gly lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leiu <br><br> 450 455 460 <br><br> CAC ACA AGA GAG AAA GAC GCC AAT GTA GTT GTG GTT GAC TGG CTC CCC 626 <br><br> His Thr Arg Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro 465 470 475 480 <br><br> CTG GCC CAC CAG CTT TAC ACG GAT GCG GTC AAT AAT ACC AGG GTG GTG 674 <br><br> Leu Ala His Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val <br><br> 485 490 495 <br><br> GGA CAC AGC ATT GCC AGG ATG CTC GAC TGG CTG CAG GAG AAG GAC GAT 722 <br><br> Gly His Ser lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp <br><br> 500 505 510 <br><br> TTT TCT CTC GGG AAT GTC CAC TTG ATC GGC TAC AGC CTC GGA GCG CAC 770 <br><br> Phe Ser Leu Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His <br><br> 515 520 525 <br><br> GTG GCC GGG TAT GCA GGC AAC TTC GTG AAA GGA ACG GTG GGC CGA ATC 818 <br><br> Val Ala Gly Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie <br><br> 530 535 540 <br><br> ACA GGT TTG GAT CCT GCC GGG CCC ATG TTT GAA GGG GCC GAC ATC CAC 866 <br><br> Thr Gly Leu Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His 545 550 555 560 <br><br> AAG AGG CTC TCT CCG GAC GAT GCA GAT TTT GTG GAT GTC CTC CAC ACC 914 <br><br> Lys Arg Leu Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr <br><br> 565 570 575 <br><br> TAC ACG CGT TCC TTC GGC TTG AGC ATT GGT ATT CAG ATG CCT GTG GGC 962 <br><br> Tyr Thr Arg Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly <br><br> 580 585 590 <br><br> CAC ATT GAC ATC TAC CCC AAT GGG GGT GAC TTC CAG CCA GGC TGT GGA 1010 <br><br> - 114 - <br><br> His lie Asp lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly <br><br> 595 600 605 <br><br> CTC AAC GAT GTC TTG GIA TCA ATT GCA TAT GGA ACA ATC ACA GAG GTG 1058 <br><br> Leu Asn Asp Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val <br><br> 610 615 620 <br><br> GTA AAA TGT GAG CAT GAG CGA GCC GTC CAC CTC TTT GTT GAC TCT CTG 1106 <br><br> Val Lys Cys Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu 625 630 635 640 <br><br> GTG AAT CAG GAC AAG CCG AGT TTT GCC TTC CAG TGC ACT GAC TCC AAT 1154 <br><br> Val Asn Gin Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn <br><br> 645 650 655 <br><br> CGC TTC AAA AAG GGG ATC TGT CTG AGC TGC CGC AAG AAC CGT TGT AAT 1202 <br><br> Arg Phe Lys Lys Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn <br><br> 660 * 665 670 <br><br> AGC ATT GGC TAC AAT GCC AAG AAA ATG AGG AAC AAG AGG AAC AGC AAA 1250 <br><br> Ser lie Gly Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys <br><br> 675 680 685 <br><br> ATG TAC CTA AAA ACC CGG GCA GGC ATG CCT TTC AGA GTT TAC CAT TAT 1298 <br><br> Met Tyr Leu Lys Thr Arg Ala Gly Met Pro Phe Arg Val Tyr His Tyr <br><br> 690 695 700 <br><br> CAG ATG AAA ATC CAT GTC TTC AGT TAC AAG AAC ATG GGA GAA ATT GAG 1346 <br><br> Gin Met Lys lie His Val Phe Ser Tyr Lys Asn Met Gly Glu lie Glu 705 710 715 720 <br><br> CCC ACC TTT TAC GTC ACC CTT TAT GGC ACT AAT GCA GAT TCC CAG ACT 1394 <br><br> Pro Thr Phe Tyr Val Thr Leu Tyr Gly Thr Asn Ala Asp Ser Gin Thr <br><br> 725 730 735 <br><br> CTG CCA CTG GAA ATA GTG GAG CGG ATC GAG CAG AAT GCC ACC AAC ACC 1442 <br><br> Leu Pro Leu Glu lie Val Glu Arg He Glu Gin Asn Ala Thr Asn Thr <br><br> 740 745 750 <br><br> TTC CTG GTC TAC ACC GAG GAG GAC TTG GGA GAC CTC TTG AAG ATC CAG 1490 <br><br> Phe Leu Val Tyr Thr Glu Glu Asp Leu Gly Asp Leu Leu Lys lie Gin <br><br> 755 760 765 <br><br> CTC ACC TGG GAG GGG GCC TCT CAG TCT TGG TAC AAC CTG TGG AAG GAG 1538 <br><br> Leu Thr Trp Glu Gly Ala Ser Gin Ser Trp Tyr Asn Leu Trp Lys Glu <br><br> 770 775 780 <br><br> TTT CGC AGC TAC CTG TCT CAA CCC CGC AAC CCC GGA CGG GAG CTG AAT 1586 <br><br> Phe Arg Ser Tyr Leu Ser Gin Pro Arg Asn Pro Gly Arg Glu Leu Asn 785 790 795 800 <br><br> ATC AGG CGC ATC CGG GTG AAG TCT GGG GAA ACC CAG CGG AAA CTG ACA 1634 <br><br> lie Arg Arg lie Arg Val Lys Ser Gly Glu Thr Gin Arg Lys Leu Thr <br><br> - 115 - <br><br> 805 810 815 <br><br> TTT TGT ACA GAA GAC CCT GAG AAC ACC AGC ATA TCC CCA GGC CGG GAG 1682 <br><br> Phe Cys Thr Glu Asp Pro Glu Asn Thr Ser lie Ser Pro Gly Arg Glu <br><br> 820 825 830 <br><br> CTC TGG TTT CGC AAG TGT CGG GAT GGC TGG AGG ATG AAA AAC GAA ACC 1730 <br><br> Leu Trp Phe Arg Lys Cys Arg Asp Gly Trp Arg Met Lys Asn Glu Thr <br><br> 835 840 845 <br><br> AGT CCC ACT GTG GAG CTT CCC TGA GGGTGCCCGG GCAAGTCTTG CCAGCAAGGC 1784 <br><br> Ser Pro Thr Val Glu Leu Pro * <br><br> 850 855 <br><br> AGCAAGACTT <br><br> CCTGCTATCC <br><br> AAGCCCATGG <br><br> AGGAAAGTTA <br><br> CTGCTGAGGA <br><br> CCCACCCAAT <br><br> 1844 <br><br> GGAAGGATTC <br><br> TTCTCAGCCT <br><br> TGACCCTGGA <br><br> GCACTGGGAA <br><br> CAACTGGTCT <br><br> CCTGTGATGG <br><br> 1904 <br><br> CTGGGACTCC <br><br> TCGCGGGAGG <br><br> GGACTGCGCT <br><br> GCTATAGCTC <br><br> TTGCTGCCTC <br><br> TCTTGAATAG <br><br> 1964 <br><br> CTCTAACTCC <br><br> AAACCTCTGT <br><br> CCACACCTCC <br><br> AGAGCACCAA <br><br> GTCCAGATTT <br><br> GTGTGTAAGC <br><br> 2024 <br><br> AGCTGGGTGC <br><br> CTGGGGCCTC <br><br> TCGTGCACAC <br><br> TGGATTGGTT <br><br> TCTCAGTTGC <br><br> TGGGCGAGCC <br><br> 2084 <br><br> TGTACTCTGC <br><br> CTGACGAGGA <br><br> ACGCTGGCTC <br><br> CGAAGAGGCC <br><br> CTGTGTAGAA <br><br> GGCTGTCAGC <br><br> 2144 <br><br> TGCTCAGCCT <br><br> GCTTTGAGCC <br><br> TCAGTGAGAA <br><br> GTCCTTCCGA <br><br> CAGGAGCTGA <br><br> CTCATGTCAG <br><br> 2204 <br><br> GATGGCAGGC <br><br> CTGGTATCTT <br><br> GCTCGGGCCC <br><br> TGGCTGTTGG <br><br> GGTTCTCATG <br><br> GGTTGCACTG <br><br> 2264 <br><br> ACCATACTGC <br><br> TTACGTCTTA <br><br> GCCATTCCGT <br><br> CCTGCTCCCC <br><br> AGCTCACTCT <br><br> CTGAAGCACA <br><br> 2324 <br><br> CATCATTGGC <br><br> TTTCCTATTT <br><br> TTCTGTTCAT <br><br> TTTTTAATTG <br><br> AGCAAATGTC <br><br> TATTGAACAC <br><br> 2384 <br><br> TTAAAATTAA <br><br> TTAGAATGTG <br><br> GTAATGGACA <br><br> TATTACTGAG <br><br> CCTCTCCATT <br><br> TGGAACCCAG <br><br> 2444 <br><br> TGGAGTTGGG <br><br> ATTTCTAGAC <br><br> CCTCTTTCTG <br><br> TTTGGATGGT <br><br> GTATGTGTAT <br><br> ATGCATGGGG <br><br> 2504 <br><br> AAAGGCACCT <br><br> GGGGCCTGGG <br><br> GGAGGCTATA <br><br> GGATATAAGC <br><br> AGTCGACGCG <br><br> GCCGCGAATT <br><br> 2564 <br><br> C <br><br> 2565 <br><br> (2) INFORMATION FOR SEQ ID N0:8: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 501 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein (Xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 15 10 15 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Leu Glu <br><br> 20 25 30 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser <br><br> 35 40 45 <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys 50 55 60 <br><br> o o <br><br> - 116 - <br><br> Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys Ser Phe Asn .65 70 75 80 <br><br> Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr Met Ser Gly <br><br> 85 90 95 <br><br> lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu His Thr Arg <br><br> 100 105 110 <br><br> Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro Leu Ala His <br><br> 115 120 125 <br><br> Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val Gly His Ser <br><br> 130 135 140 <br><br> lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp Phe Ser Leu 145 150 155 160 <br><br> Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His Val Ala Gly <br><br> 165 170 175 <br><br> Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie Thr Gly Leu <br><br> 180 185 190 <br><br> Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His Lys Arg Leu <br><br> 195 200 205 <br><br> Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr Tyr Thr Arg <br><br> 210 215 220 <br><br> Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly His lie Asp 225 230 235 240 <br><br> lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly Leu Asn Asp <br><br> 245 250 255 <br><br> Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val Val Lys Cys <br><br> 260 265 270 <br><br> Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin <br><br> 275 280 285 <br><br> Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys <br><br> 290 295 300 <br><br> Lys Gly He Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Ser lie Gly 305 310 315 320 <br><br> Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met Tyr Leu <br><br> 325 330 335 <br><br> Lys Thr Arg Ala Gly Met Pro Phe Arg Val Tyr His Tyr Gin Met Lys <br><br> 340 345 350 <br><br> He His Val Phe Ser Tyr Lys Asn Met Gly Glu lie Glu Pro Thr Phe <br><br> 355 360 365 <br><br> Tyr Val Thr Leu Tyr Gly Thr Asn Ala Asp Ser Gin Thr Leu Pro Leu 370 375 380 <br><br> o o <br><br> - 117 - <br><br> Glu lie Val Glu Arg lie Glu Gin Asn Ala Thr Asn Thr Phe Leu Val 385 390 395 400 <br><br> Tyr Thr Glu Glu Asp Leu Gly Asp Leu Leu Lys lie Gin Leu Thr Trp <br><br> 405 410 415 <br><br> Glu Gly Ala Ser Gin Ser Trp Tyr Asn Leu Trp Lys Glu Phe Arg Ser <br><br> 420 425 430 <br><br> Tyr Leu Ser Gin Pro Arg Asn Pro Gly Arg Glu Leu Asn lie Arg Arg <br><br> 435 440 445 <br><br> lie Arg Val Lys Ser Gly Glu Thr Gin Arg Lys Leu Thr Phe Cys Thr <br><br> 450 455 460 <br><br> Glu Asp Pro Glu Asn Thr Ser lie Ser Pro Gly Arg Glu Leu Trp Phe 465 470 475 480 <br><br> Arg Lys Cys Arg Asp Gly Trp Arg Met Lys Asn Glu Thr Ser Pro Thr <br><br> 485 490 495 <br><br> Val Glu Leu Pro * <br><br> 500 <br><br> (2) INFORMATION FOR SEQ ID NO:9: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 1035 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 1..1035 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: <br><br> ATG AGC AAC TCC GTT CCT CTG CTC TGT TTC TGG AGC CTC TGC TAT TGC 48 <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 505 510 515 <br><br> TTT GCT GCG GGG AGC CCC GTA CCT TTT GGT CCA GAG GGA CGG CTG GAA 96 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Leu Glu <br><br> 520 525 530 <br><br> GAT AAG CTC CAC AAA CCC AAA GCT ACA CAG ACT GAG GTC AAA CCA TCT 144 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser <br><br> 535 540 545 <br><br> GTG AGG TTT AAC CTC CGC ACC TCC AAG GAC CCA GAG CAT GAA GGA TGC 192 <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys 550 555 560 565 <br><br> o o <br><br> TAC CTC TCC GTC GGC CAC Tyr Leu Ser Val Gly His 570 <br><br> ATG ACA GCT AAA ACC TTT Met Thr Ala Lys Thr Phe 585 <br><br> ATC TTT GAA AAC TGG CTG lie Phe Glu Asn Trp Leu 600 <br><br> - 118 - <br><br> AGC CAG CCC TTA GAA GAC Ser Gin Pro Leu Glu Asp 575 <br><br> TTC ATC ATT CAC GGA TGG Phe lie lie His Gly Trp 590 <br><br> CAC AAA CTC GTG TCA GCC His Lys Leu Val Ser Ala 605 <br><br> TGC AGT TTC AAC 240 <br><br> Cys Ser Phe Asn 580 <br><br> ACG ATG AGC GGT 288 <br><br> Thr Met Ser Gly 595 <br><br> CTG CAC ACA AGA 336 <br><br> Leu His Thr Arg <br><br> 610 <br><br> o o <br><br> GAG AAA GAC GCC Glu Lys Asp Ala 615 <br><br> CAG CTT TAC ACG Gin Leu Tyr Thr 630 <br><br> ATT GCC AGG ATG lie Ala Arg Met <br><br> GGG AAT GTC CAC Gly Asn Val His 665 <br><br> TAT GCA GGC AAC Tyr Ala Gly Asn 680 <br><br> GAT CCT GCC GGG Asp Pro Ala Gly 695 <br><br> TCT CCG GAC GAT Ser Pro Asp Asp 710 <br><br> TCC TTC GGC TTG Ser Phe Gly Leu <br><br> ATC TAC CCC AAT lie Tyr Pro Asn 745 <br><br> GTC TTG GGA TCA Val Leu Gly Ser 760 <br><br> GAG CAT GAG CGA Glu His Glu Arg 775 <br><br> GAC AAG CCG AGT Asp Lys Pro Ser 790 <br><br> AAG GGG ATC TGT Lys Gly lie Cys <br><br> TAC AAT GCC AAG <br><br> AAT GTA GTT GTG Asn Val Val Val 620 <br><br> GAT GCG GTC AAT Asp Ala Val Asn 635 <br><br> CTC GAC TGG CTG Leu Asp Trp Leu 650 <br><br> TTG ATC GGC TAC Leu lie Gly Tyr <br><br> TTC GTG AAA GGA Phe Val Lys Gly 685 <br><br> CCC ATG TTT GAA Pro Met Phe Glu 700 <br><br> GCA GAT TTT GTG Ala Asp Phe Val 715 <br><br> AGC ATT GGT ATT Ser lie Gly lie 730 <br><br> GGG GGT GAC TTC Gly Gly Asp Phe <br><br> ATT GCA TAT GGA lie Ala Tyr Gly 765 <br><br> GCC GTC CAC CTC Ala Val His Leu 780 <br><br> TTT GCC TTC CAG Phe Ala Phe Gin 795 <br><br> CTG AGC TGC CGC Leu Ser Cys Arg 810 <br><br> AAA ATG AGG AAC <br><br> - 119 - <br><br> GTT GAC TGG CTC Val Asp Trp Leu 625 <br><br> AAT ACC AGG GTG Asn Thr Arg Val 640 <br><br> CAG GAG AAG GAC Gin Glu Lys Asp 655 <br><br> AGC CTC GGA GCG Ser Leu Gly Ala 670 <br><br> ACG GTG GGC CGA Thr Val Gly Arg <br><br> GGG GCC GAC ATC Gly Ala Asp lie 705 <br><br> GAT GTC CTC CAC Asp Val Leu His 720 <br><br> CAG ATG CCT GTG Gin Met Pro Val 735 <br><br> CAG CCA GGC TGT Gin Pro Gly Cys 750 <br><br> ACA ATC ACA GAG Thr lie Thr Glu <br><br> TTT GTT GAC TCT Phe Val Asp Ser 785 <br><br> TGC ACT GAC TCC Cys Thr Asp Ser 800 <br><br> AAG AAC CGT TGT Lys Asn Arg Cys 815 <br><br> AAG AGG AAC AGC <br><br> CCC CTG GCC CAC Pro Leu Ala His <br><br> GTG GGA CAC AGC Val Gly His Ser 645 <br><br> GAT TTT TCT CTC Asp Phe Ser Leu 660 <br><br> CAC GTG GCC GGG His Val Ala Gly 675 <br><br> ATC ACA GGT TTG lie Thr Gly Leu 690 <br><br> CAC AAG AGG CTC His Lys Arg Leu <br><br> ACC TAC ACG CGT Thr Tyr Thr Arg 725 <br><br> GGC CAC ATT GAC Gly His lie Asp 740 <br><br> GGA CTC AAC GAT Gly Leu Asn Asp 755 <br><br> GTG GTA AAA TGT Val Val Lys Cys 770 <br><br> CTG GTG AAT CAG Leu Val Asn Gin <br><br> AAT CGC TTC AAA Asn Arg Phe Lys 805 <br><br> AAT AGC ATT GGC Asn Ser lie Gly 820 <br><br> AAA ATG TAC CTA <br><br> o o <br><br> - 120 - <br><br> Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met Tyr Leu <br><br> 825 830 835 <br><br> AAA ACC CGG GCA GGC ATG CCT TTC AGA 1035 <br><br> Lys Thr Arg Ala Gly Met Pro Phe Arg 840 845 <br><br> (2) INFORMATION FOR SEQ ID NO?10: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 345 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:10: <br><br> Met Ser Asn Ser Val Pro Leu Leu Cys Phe Trp Ser Leu Cys Tyr Cys <br><br> 1 5 10 15 <br><br> Phe Ala Ala Gly Ser Pro Val Pro Phe Gly Pro Glu Gly Arg Lieu Glu <br><br> 20 25 30 <br><br> Asp Lys Leu His Lys Pro Lys Ala Thr Gin Thr Glu Val Lys Pro Ser 35 40 45 <br><br> - 121 - <br><br> Val Arg Phe Asn Leu Arg Thr Ser Lys Asp Pro Glu His Glu Gly Cys <br><br> 50 55 60 <br><br> Tyr Leu Ser Val Gly His Ser Gin Pro Leu Glu Asp Cys Ser Phe Asn 65 70 75 80 <br><br> Met Thr Ala Lys Thr Phe Phe lie lie His Gly Trp Thr Met Ser Gly <br><br> 85 90 95 <br><br> lie Phe Glu Asn Trp Leu His Lys Leu Val Ser Ala Leu His Thr Arg <br><br> 100 105 110 <br><br> Glu Lys Asp Ala Asn Val Val Val Val Asp Trp Leu Pro Leu Ala His <br><br> 115 120 125 <br><br> Gin Leu Tyr Thr Asp Ala Val Asn Asn Thr Arg Val Val Gly His Ser <br><br> 130 135 140 <br><br> lie Ala Arg Met Leu Asp Trp Leu Gin Glu Lys Asp Asp Phe Ser Leu 145 150 155 160 <br><br> Gly Asn Val His Leu lie Gly Tyr Ser Leu Gly Ala His Val Ala Gly <br><br> 165 170 175 <br><br> Tyr Ala Gly Asn Phe Val Lys Gly Thr Val Gly Arg lie Thr Gly Leu <br><br> 180 185 190 <br><br> Asp Pro Ala Gly Pro Met Phe Glu Gly Ala Asp lie His Lys Arg Leu <br><br> 195 200 205 <br><br> Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His Thr Tyr Thr Arg <br><br> 210 215 220 <br><br> Ser Phe Gly Leu Ser lie Gly lie Gin Met Pro Val Gly His lie Asp 225 230 235 240 <br><br> lie Tyr Pro Asn Gly Gly Asp Phe Gin Pro Gly Cys Gly Leu Asn Asp <br><br> 245 250 255 <br><br> Val Leu Gly Ser lie Ala Tyr Gly Thr lie Thr Glu Val Val Lys Cys <br><br> 260 265 270 <br><br> Glu His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin <br><br> 275 280 285 <br><br> Asp Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys <br><br> 290 295 300 <br><br> Lys Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Ser lie Gly 305 310 315 320 <br><br> Tyr Asn Ala Lys Lys Met Arg Asn Lys Arg Asn Ser Lys Met Tyr Leu <br><br> 325 330 335 <br><br> Lys Thr Arg Ala Gly Met Pro Phe Arg 340 345 <br><br> (2) INFORMATION FOR SEQ ID NO: 11: <br><br> o o <br><br> - 122 - <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 225 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 1..225 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: <br><br> CTG GGA TCC ATC GCC TAT GGC ACG ATC GCG GAG GTG GTG AAG TGC GAG Leu Gly Ser lie Ala Tyr Gly Thr lie Ala Glu Val Val Lys Cys Glu <br><br> 350 355 360 <br><br> CAT GAG CGG GCC GTG CAT CTC TTT GTG GAC TCC CTG GTG AAC CAG GAC His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin Asp <br><br> 365 370 375 <br><br> AAG CCG AGC TTT GCC TTC CAG TGC ACA GAC TCC AAC CGC TTC AAA AAA Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys Lys <br><br> 380 385 390 <br><br> GGG ATC TGT CTC AGC TGC CGG AAG AAC CGC TGT AAC GGC ATC GGC TAC Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Gly lie Gly Tyr <br><br> 395 400 405 <br><br> AAT GCT AAG AAG ACG AGG AAT AAG AGG AAC ACC Asn Ala Lys Lys Thr Arg Asn Lys Arg Asn Thr 410 415 420 <br><br> (2) INFORMATION FOR SEQ ID NO: 12: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 75 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: <br><br> Leu Gly Ser lie Ala Tyr Gly Thr lie Ala Glu Val Val Lys Cys Glu <br><br> 15 10 15 <br><br> His Glu Arg Ala Val His Leu Phe Val Asp Ser Leu Val Asn Gin Asp <br><br> 20 25 30 <br><br> Lys Pro Ser Phe Ala Phe Gin Cys Thr Asp Ser Asn Arg Phe Lys Lys <br><br> 35 40 45 <br><br> Gly lie Cys Leu Ser Cys Arg Lys Asn Arg Cys Asn Gly lie Gly Tyr <br><br> - 123 - <br><br> 50 55 60 <br><br> Asn Ala Lys Lys Thr Arg Asn Lys Arg Asn Thr 65 70 75 <br><br> (2) INFORMATION FOR SEQ ID NO:13: <br><br> &lt;i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 472 amino acids <br><br> (B) TYPE: amino acid <br><br> (C) STRANDEDNESS: <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: <br><br> Met Glu Ser Lys Ala Leu Leu Val Leu Thr Leu Ala Val Trp Leu Gin 1 5 10 15 <br><br> Ser Leu Thr Ala Ser Arg Gly Gly Val Ala Ala Ala Asp Gin Arg Arg <br><br> 20 25 30 <br><br> Asp Phe lie Asp lie Glu Ser Lys Phe Ala Leu Arg Thr Pro Glu Asp <br><br> 35 40 45 <br><br> Thr Ala Glu Asp Thr Cys His Leu lie Pro Gly Val Ala Glu Ser Val <br><br> 50 55 60 <br><br> Ala Thr Cys His Phe Asn His Ser Ser Lys Thr Phe Met Val lie His 65 70 75 80 <br><br> Gly Trp Thr Val Thr Gly Met Tyr Glu Ser Trp Val Pro Lys Leu Val <br><br> 85 90 95 <br><br> Ala Ala Leu Tyr Lys Arg Glu Pro Asp Ser Asn Val lie Val Val Asp <br><br> 100 105 110 <br><br> Trp Leu Ser Arg Ala Gin Glu His Tyr Pro Val Ser Ala Gly Tyr Thr <br><br> 115 120 125 <br><br> Lys Val Gly Gin Asp Val Ala Arg Phe lie Asn Trp Met Glu Glu Glu <br><br> 130 135 140 <br><br> Phe Asn Tyr Pro Leu Asp Asn Val His Leu Leu Gly Tyr Ser Leu Gly 145 150 155 160 <br><br> Ala His Ala Ala Gly lie Ala Gly Ser Leu Thr Asn Lys Lys Val Asn <br><br> 165 170 175 <br><br> Arg lie Thr Gly Leu Asp Pro Ala Gly Pro Asn Phe Glu Tyr Ala Glu <br><br> 180 185 190 <br><br> Ala Pro Arg Leu Ser Pro Asp Asp Ala Asp Phe Val Asp Val Leu His <br><br> 195 200 205 <br><br> Thr Phe Thr Arg Gly Ser Pro Gly Arg Ser lie Gly lie Gin Lys Pro 210 215 220 <br><br> o o <br><br> - 124 - <br><br> Val Gly His Val Asp lie Tyr Pro Asn Gly Gly Thr Phe Gin Pro Gly 225 , 230 235 240 <br><br> Cys Asn lie Gly Glu Ala He Arg Val lie Ala Glu Arg Gly Leu Gly <br><br> 245 250 255 <br><br> Asp Val Asp Gin Leu Lys Cys Ser His Glu Arg Ser lie His Leu Phe <br><br> 260 265 270 <br><br> lie Asp Ser Leu Leu Asn Glu Glu Asn Pro Ser Lys Ala Tyr Arg Cys <br><br> 275 280 285 <br><br> Ser Ser Lys Glu Ala Phe Glu Lys Gly Leu Cys Leu Ser Cys Arg Lys <br><br> 290 295 300 <br><br> Asn Arg Cys Asn Asn Leu Gly Tyr Glu lie Asn Lys Val Arg Ala Lys 305 310 315 320 <br><br> Arg Ser Ser Lys Met Tyr Leu Lys Thr Arg Ser Gin Met Pro Tyr Lys <br><br> 325 330 335 <br><br> Val Phe His Tyr Gin Val Lys lie His Phe Ser Gly Thr Glu Ser Glu <br><br> 340 345 350 <br><br> Thr His Thr Asn Gin Ala Phe Glu lie Ser Leu Tyr Gly Thr Val Ala <br><br> 355 360 365 <br><br> Glu Ser Glu Asn lie Pro Phe Thr Leu Pro Glu Val Ser Thr Asn Lys <br><br> 370 375 380 <br><br> Thr Tyr Ser Phe Leu lie Tyr Thr Glu Val Asp lie Gly Glu Leu Leu 385 390 395 400 <br><br> Met Leu Lys Leu Lys Trp Lys Ser Asp Ser Tyr Phe Ser Trp Ser Asp <br><br> 405 410 415 <br><br> Trp Trp Ser Ser Pro Gly Phe Ala lie Gin Lys lie Arg Val Lys Ala <br><br> 420 425 430 <br><br> Gly Glu Thr Gin Lys Lys Val lie Phe Cys Ser Arg Glu Lys Val Ser <br><br> 435 440 445 <br><br> His Leu Gin Lys Gly Lys Ala Pro Ala Val Phe Val Lys Cys His Asp <br><br> 450 455 460 <br><br> Lys Ser Leu Asn Lys Lys Ser Gly 465 470 <br><br> (2) INFORMATION FOR SEQ ID NO:14: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 499 amino acids <br><br> (B) TYPE: amino acid <br><br> (C) STRANDEDNESS: <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein <br><br> o <br><br> - 125 - <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: <br><br> Met Asp Thr Ser Pro Leu Cys Phe Ser lie Leu Leu Val Leu Cys lie 1 5 10 15 <br><br> Phe lie Gin Ser Ser Ala Leu Gly Gin Ser Leu Lys Pro Glu Pro Phe <br><br> 20 25 30 <br><br> Gly Arg Arg Ala Gin Ala Val Glu Thr Asn Lys Thr Leu His Glu Met <br><br> 35 40 45 <br><br> Lys Thr Arg Phe Leu Leu Phe Gly Glu Thr Asn Gin Gly Cys Gin lie <br><br> 50 55 60 <br><br> Arg lie Asn His Pro Asp Thr Leu Gin Glu Cys Gly Phe Asn Ser Ser 65 70 75 80 <br><br> Leu Pro Leu Val Met lie lie His Gly Trp Ser Val Asp Gly Val Leu <br><br> 85 90 95 <br><br> Glu Asn Trp lie Trp Gin Met Val Ala Ala Leu Lys Ser Gin Pro Ala <br><br> 100 105 110 <br><br> Gin Pro Val Asn Val Gly Leu Val Asp Trp lie Thr Leu Ala His Asp <br><br> 115 120 125 <br><br> His Tyr Thr lie Ala Val Arg Asn Thr Arg Leu Val Gly Lys Glu Val 130 135 140 <br><br> o o <br><br> - 126 - <br><br> Ala Ala Leu Leu Arg Trp Leu Glu Glu Ser Val Gin Leu Ser Arg Ser 145 150 155 160 <br><br> His Val His Leu lie Gly Tyr Ser Leu Gly Ala His Val Ser Gly Phe <br><br> 165 170 175 <br><br> Ala Gly Ser Ser lie Gly Gly Thr His Lys lie Gly Arg lie Thr Gly <br><br> 180 185 190 <br><br> Leu Asp Ala Ala Gly Pro Leu Phe Glu Gly Ser Ala Pro Ser Asn Arg <br><br> 195 200 205 <br><br> Leu Ser Pro Asp Asp Ala Asn Phe Val Asp Ala lie His Thr Phe Thr <br><br> 210 215 220 <br><br> Arg Glu His Met Gly Leu Ser Val Gly lie Lys Gin Pro He Gly His 225 230 235 240 <br><br> Tyr Asp Phe Tyr Pro Asn Gly Gly Ser Phe Gin Pro Gly Cys His Phe <br><br> 245 250 255 <br><br> Leu Glu Leu Tyr Arg His lie Ala Gin His Gly Phe Asn Ala lie Thr <br><br> 260 265 270 <br><br> Gin Thr lie Lys Cys Ser His Glu Arg Ser Val His Leu Phe lie Asp <br><br> 275 280 285 <br><br> Ser Leu Leu His Ala Gly Thr Gin Ser Met Ala Tyr Pro Cys Gly Asp <br><br> 290 295 300 <br><br> Met Asn Ser Phe Ser Gin Gly Leu Cys Leu Ser Cys Lys Lys Gly Arg 305 310 315 320 <br><br> Cys Asn Thr Leu Gly Tyr His Val Arg Gin Glu Pro Arg Ser Lys Ser <br><br> 325 330 335 <br><br> Lys Arg Leu Phe Leu Val Thr Arg Ala Gin Ser Pro Phe Lys Val Tyr <br><br> 340 345 350 <br><br> His Tyr Gin Leu Lys lie Gin Phe lie Asn Gin Thr Glu Thr Pro lie <br><br> 355 360 365 <br><br> Gin Thr Thr Phe Thr Met Ser Leu Leu Gly Thr Lys Glu Lys Met Gin <br><br> 370 375 380 <br><br> Lys lie Pro lie Thr Leu Gly Lys Gly lie Ala Ser Asn Lys Thr Tyr 385 390 395 400 <br><br> Ser Phe Leu lie Thr Leu Asp Val Asp lie Gly Glu Leu lie Met lie <br><br> 405 410 415 <br><br> Lys Phe Lys Trp Glu Asn Ser Ala Val Trp Ala Asn Val Trp Asp Thr <br><br> 420 425 430 <br><br> Val Gin Thr lie lie Pro Trp Ser Thr Gly Pro Arg His Ser Gly Leu <br><br> 435 440 445 <br><br> Val Leu Lys Thr lie Arg Val Lys Ala Gly Glu Thr Gin Gin Arg Met 450 455 460 <br><br> o o <br><br> - 127 - <br><br> Thr Phe Cys Ser Glu Asn Thr Asp Asp Leu Leu Leu Arg Pro Thr Gin 465 470 475 480 <br><br> Glu Lys lie Phe Val Lys Cys Glu lie Lys Ser Lys Thr Ser Lys Arg 485 490 495 <br><br> Lys lie Arg <br><br> (2) INFORMATION FOR SEQ ID NO:15: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 465 amino acids <br><br> (B) TYPE: amino acid <br><br> (C) STRANDEDNESS: <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:15: <br><br> Met Leu Pro Leu Trp Thr Leu Ser Leu Leu Leu Gly Ala Val Ala Gly 1 ' 5 10 15 <br><br> Lys Glu Val Cys Tyr Glu Arg Leu Gly Cys Phe Ser Asp Asp Ser Pro <br><br> 20 25 30 <br><br> Trp Ser Gly lie Thr Glu Arg Pro Leu His lie Leu Pro Trp Ser Pro <br><br> 35 40 45 <br><br> Lys Asp Val Asn Thr Arg Phe Leu Leu Tyr Thr Asn Glu Asn Pro Asn <br><br> 50 55 60 <br><br> Asn Phe Gin Glu Val Ala Ala Asp Ser Ser Ser lie Ser Gly Ser Asn 65 70 75 80 <br><br> Phe Lys Thr Asn Arg Lys Thr Arg Phe lie lie His Gly Phe lie Asp <br><br> 85 90 95 <br><br> Lys Gly Glu Glu Asn Trp Leu Ala Asn Val Cys Lys Asn Leu Phe Lys <br><br> 100 105 110 <br><br> Val Glu Ser Val Asn Cys lie Cys Val Asp Trp Lys Gly Gly Ser Arg <br><br> 115 120 125 <br><br> Thr Gly Tyr Thr Gin Ala Ser Gin Asn lie Arg lie Val Gly Ala Glu <br><br> 130 135 140 <br><br> Val Ala Tyr Phe Val Glu Phe Leu Gin Ser Ala Phe Gly Tyr Ser Pro 145 150 155 160 <br><br> Ser Asn Val His Val lie Gly His Ser Leu Gly Ala His Ala Ala Gly <br><br> 165 170 175 <br><br> Glu Ala Gly Arg Arg Thr Asn Gly Thr lie Gly Arg lie Thr Gly Leu <br><br> 180 185 190 <br><br> Asp Pro Ala Glu Pro Cys Phe Gin Gly Thr Pro Glu Leu Val Arg Leu 195 200 205 <br><br> o <br><br> - 128 - <br><br> Asp Pro Ser Asp Ala Lys Phe Val Asp Val lie His Thr Asp Gly Ala <br><br> 210 215 220 <br><br> Pro lie Val Pro Asn Leu Gly Phe Gly Met Ser Gin Val Val Gly His 225 230 235 240 <br><br> Leu Asp Phe Phe Pro Asn Gly Gly Val Glu Met Pro Gly Cys Lys Lys <br><br> 245 250 255 <br><br> Asn lie Leu Ser Gin lie Val Asp lie Asp Gly lie Trp Glu Gly Thr <br><br> 260 265 270 <br><br> Arg Asp Phe Ala Ala Cys Asn His Leu Arg Ser Tyr Lys Tyr Tyr Thr <br><br> 275 280 285 <br><br> Asp Ser lie Val Asn Pro Asp Gly Phe Ala Gly Phe Pro Cys Ala Ser <br><br> 290 295 300 <br><br> Tyr Asn Val Phe Thr Ala Asn Lys Cys Phe Pro Cys Pro Ser Gly Gly 305 310 315 320 <br><br> Cys Pro Gin Met Gly His Tyr Ala Asp Arg Tyr Pro Gly Lys Thr Asn <br><br> 325 330 335 <br><br> Asp Val Gly Gin Lys Phe Tyr Leu Asp Thr Gly Asp Ala Ser Asn Phe <br><br> 340 345 350 <br><br> Ala Arg Trp Arg Tyr Lys Val Ser Val Thr Leu Ser Gly Lys Lys Val <br><br> 355 360 365 <br><br> Thr Gly His lie Leu Val Ser Leu Phe Gly Asn Lys Gly Asn Ser Lys <br><br> 370 375 380 <br><br> Gin Tyr Glu lie Phe Lys Gly Thr Leu Lys Pro Asp Ser Thr His Ser 385 390 395 400 <br><br> Asn Glu Phe Asp Ser Asp Val Asp Val Gly Asp Leu Gin Met Val Lys <br><br> 405 410 415 <br><br> Phe lie Trp Tyr Asn Asn Val lie Asn Pro Thr Leu Pro Arg Val Gly <br><br> 420 425 430 <br><br> Ala Ser Lys lie lie Val Glu Thr Asn Val Gly Lys Gin Phe Asn Phe <br><br> 435 440 445 <br><br> Cys Ser Pro Glu Thr Val Arg Glu Glu Val Leu Leu Thr Leu Thr Pro 450 455 460 <br><br> Cys 465 <br><br> (2) INFORMATION FOR SEQ ID NO:16: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 17 amino acids &lt;B) TYPE: amino acid (C) STRANDEDNESS: <br><br> o <br><br> - 129 - <br><br> (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16: <br><br> Gly Pro Glu Gly Arg Leu Glu Asp Lys Leu His Lys Pro Lys Ala Thr 15 10 15 <br><br> Cys <br><br> (2) INFORMATION FOR SEQ ID NO:17: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 13 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17: <br><br> TTTTTTTTTT TGA <br><br> (2) INFORMATION FOR SEQ ID NO:18: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 10 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18: <br><br> GATCAATCGC <br><br> (2) INFORMATION FOR SEQ ID NO:19: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 23 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: <br><br> TAGGACATGC ACAGTGTAAT CTG <br><br> - 130 - <br><br> (2) INFORMATION FOR SEQ ID NO:20: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 20 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:20: GATTGTGCTG GCCACTTCTC <br><br> (2) INFORMATION FOR SEQ ID NO:21: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 19 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: GACACTCCAG GGACTGAAG <br><br> (2) INFORMATION FOR SEQ ID NO:22: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 48 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> (B) LOCATION: 36 <br><br> (D) OTHER INFORMATION: /mod_base= i (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> (B) LOCATION: 37 <br><br> (D) OTHER INFORMATION: /mod_base= i (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> o <br><br> - 131 - <br><br> (B) LOCATION: 41 <br><br> (D) OTHER INFORMATION: /mod_base= i (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> (B) LOCATION: 42 <br><br> (D) OTHER INFORMATION: /mod_base= i (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> (B) LOCATION: 46 <br><br> (D) OTHER INFORMATION: /mod_base= i (ix) FEATURE: <br><br> (A) NAME/KEY: modified_base <br><br> (B) LOCATION: 47 <br><br> (D) OTHER INFORMATION: /mod_base= i (xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: CUACUACUAC UAGGCCACGC GTCGACTAGT ACGGGNNGGG NNGGGNNG <br><br> (2) INFORMATION FOR SEQ ID NO:23: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 28 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: CACACACAGG CCACGCGTCG ACTAGTAC <br><br> (2) INFORMATION FOR SEQ ID NO:24: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 24 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: ACCACCATGG AGAGCAAAGC CCTG <br><br> (2) INFORMATION FOR SEQ ID NO:25: <br><br> - 132 - <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 25 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single &lt;D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:25: CCAGTTTCAG CCTGACTTCT TATTC <br><br> (2) INFORMATION FOR SEQ ID NO:26: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 21 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26: GGCTGTGGAC TCAACGATGT C <br><br> (2) INFORMATION FOR SEQ ID NO:27: <br><br> (!) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 22 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide' (xi) SEQUENCE DESCRIPTION: SEQ ID NO;27: CCGGGTGGGT AGGTACATTT TG <br><br> (2) INFORMATION FOR SEQ ID NO:28: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 25 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide1 <br><br> - 133 - <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:28: GGGGGTGACT TCCAGCCAGG CTGTG <br><br> (2) INFORMATION FOR SEQ ID NO:29: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 25 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:29: AACTCTGAAA GGCATGCCTG CCCGG <br><br> (2) INFORMATION FOR SEQ ID NO:30: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 26 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:30: TGAAGGTCGG AGTCAACGGA TTTGGT <br><br> (2) INFORMATION FOR SEQ ID NO:31: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 24 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: other nucleic acid <br><br> (A) DESCRIPTION: /desc = "Oligonucleotide" (xi) SEQUENCE DESCRIPTION: SEQ ID NO:31: CATGTGGGCC ATGAGGTCCA CCAC <br><br></p> </div>

Claims (12)

<div class="application article clearfix printTableText" id="claims"> <p lang="en"> 134<br><br> WHAT WE CLAIM IS;<br><br>
1. A composition capable of lowering the expression of LIPG in a patient comprising a ribozyme which cleaves mRNA which encodes LIPG polypeptide.<br><br>
2. A composition capable of lowering the expression of LIPG in a patient comprising an expression vector which includes a DNA sequence encoding a ribozyme which cleaves mRNA which encodes LIPG polypeptide.<br><br>
3. The composition of Claim 2 wherein said expression vector is selected from the group consisting of retroviral vectors, adenoviral vectors, adeno-associated viral vectors, herpesviral vectors, and naked DNA vectors.<br><br>
4. The composition of Claim 1 wherein said ribozyme is a hammerhead ribozyme.<br><br>
5. Use of a ribozyme which cleaves mRNA which encodes LIPG polypeptide in the manufacture of a medicament for raising the level of high density lipoprotein (HDL) cholesterol and apolipoprotein AI in a patient.<br><br>
6. Use of an expression vector including a DNA sequence which encodes a ribozyme which cleaves mRNA which encodes LIPG polypeptide in the manufacture of a medicament for raising the level of high density lipoprotein (HDL) cholesterol and apolipoprotein AI in a patient.<br><br>
7. The use of Claim 6 wherein said expression vector is selected from the group consisting of retroviral vectors, adenoviral vectors and adeno-associated viral vectors, herpesviral vectors, and naked DNA vectors.<br><br>
8. A composition as claimed in claim 1 substantially as herein described with reference to any example thereof.<br><br>
9. A composition as claimed in claim 2 substantially as herein described with reference to any example thereof.<br><br> 208170_1.DOC ' ■ " -T « •' -V<br><br> 135<br><br>
10. A use as claimed in claim 5 substantially as herein described with reference to any example thereof.<br><br>
11. A use as claimed in claim 6 substantially as herein described with reference to any example thereof.<br><br>
12. A use as claimed in claim 7 substantially as herein described with reference to any example thereof.<br><br> Or HI.<br><br> ■&gt;'i± ii'V<br><br> ' 3 SEP 283J I D i<br><br> 208170_1.DOC<br><br> </p> </div>
NZ531180A 1999-03-26 2000-03-24 Compositions and methods for effecting the levels of cholesterol using the LIPG polypeptide NZ531180A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/277,401 US7008776B1 (en) 1996-12-06 1999-03-26 Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol

Publications (1)

Publication Number Publication Date
NZ531180A true NZ531180A (en) 2005-06-24

Family

ID=23060703

Family Applications (2)

Application Number Title Priority Date Filing Date
NZ531180A NZ531180A (en) 1999-03-26 2000-03-24 Compositions and methods for effecting the levels of cholesterol using the LIPG polypeptide
NZ51435000A NZ514350A (en) 1999-03-26 2000-03-24 Compositions and methods for effecting the levels of cholesterol

Family Applications After (1)

Application Number Title Priority Date Filing Date
NZ51435000A NZ514350A (en) 1999-03-26 2000-03-24 Compositions and methods for effecting the levels of cholesterol

Country Status (13)

Country Link
EP (1) EP1171078A4 (en)
JP (1) JP2002540127A (en)
KR (2) KR20050044812A (en)
AU (1) AU776684B2 (en)
BR (1) BR0009333A (en)
CA (1) CA2363486C (en)
HK (1) HK1043309A1 (en)
IL (2) IL145526A0 (en)
MX (1) MXPA01009727A (en)
NO (2) NO331779B1 (en)
NZ (2) NZ531180A (en)
WO (1) WO2000057837A2 (en)
ZA (1) ZA200107598B (en)

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7507808B2 (en) * 2002-12-12 2009-03-24 Isis Pharmaceuticals, Inc. Modulation of endothelial lipase expression
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
US8921329B2 (en) 2008-12-04 2014-12-30 Curna, Inc. Treatment of erythropoietin (EPO) related diseases by inhibition of natural antisense transcript to EPO
US8927511B2 (en) 2008-12-04 2015-01-06 Curna, Inc. Treatment of vascular endothelial growth factor (VEGF) related diseases by inhibition of natural antisense transcript to VEGF
US20110294870A1 (en) 2008-12-04 2011-12-01 Opko Curna, Llc Treatment of tumor suppressor gene related diseases by inhibition of natural antisense transcript to the gene
DK2396038T3 (en) 2009-02-12 2016-02-01 Curna Inc TREATMENT OF BRAIN-DERIVATED NEUROTROPHIC FACTOR- (BDNF) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENCE TRANSCRIPTION TO BDNF
EP2408919B1 (en) 2009-03-16 2017-10-18 CuRNA, Inc. Treatment of nuclear factor (erythroid-derived 2)-like 2 (nrf2) related diseases by inhibition of natural antisense transcript to nrf2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
WO2010129799A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of lipid transport and metabolism gene related diseases by inhibition of natural antisense transcript to a lipid transport and metabolism gene
WO2010129746A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
WO2010129861A2 (en) 2009-05-08 2010-11-11 Curna, Inc. Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
DK2432881T3 (en) 2009-05-18 2018-02-26 Curna Inc TREATMENT OF REPROGRAMMING FACTOR-RELATED DISEASES BY INHIBITING NATURAL ANTISENSE TRANSCRIPTS TO A REPROGRAMMING FACTOR
WO2010135695A2 (en) 2009-05-22 2010-11-25 Curna, Inc. TREATMENT OF TRANSCRIPTION FACTOR E3 (TFE3) and INSULIN RECEPTOR SUBSTRATE 2 (IRS2) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO TFE3
US8791085B2 (en) 2009-05-28 2014-07-29 Curna, Inc. Treatment of antiviral gene related diseases by inhibition of natural antisense transcript to an antiviral gene
KR101702689B1 (en) 2009-06-16 2017-02-06 큐알엔에이, 인크. Treatment of paraoxonase 1 (pon1) related diseases by inhibition of natural antisense transcript to pon1
EP2443237B1 (en) 2009-06-16 2017-02-22 CuRNA, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
WO2010151671A2 (en) 2009-06-24 2010-12-29 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (tnfr2) related diseases by inhibition of natural antisense transcript to tnfr2
EP2446037B1 (en) 2009-06-26 2016-04-20 CuRNA, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
US20120252869A1 (en) 2009-07-24 2012-10-04 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
WO2011017516A2 (en) 2009-08-05 2011-02-10 Curna, Inc. Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
CA2770104C (en) 2009-08-11 2019-03-19 Opko Curna, Llc Treatment of adiponectin (adipoq) related diseases by inhibition of natural antisense transcript to an adiponectin (adipoq)
JP5943836B2 (en) 2009-08-21 2016-07-05 カッパーアールエヌエー,インコーポレイテッド Treatment of CHIP-related diseases by inhibition of natural antisense transcripts on 'HSP70 interacting protein C-terminus' (CHIP)
CA2771172C (en) 2009-08-25 2021-11-30 Opko Curna, Llc Treatment of 'iq motif containing gtpase activating protein' (iqgap) related diseases by inhibition of natural antisense transcript to iqgap
NO2480669T3 (en) 2009-09-25 2018-04-07
JP6025567B2 (en) 2009-12-16 2016-11-16 カッパーアールエヌエー,インコーポレイテッド Treatment of MBTPS1-related diseases by inhibition of the natural antisense transcript against the membrane-bound transcription factor peptidase, site 1 (MBTPS1)
JP6031356B2 (en) 2009-12-23 2016-11-24 カッパーアールエヌエー,インコーポレイテッド Treatment of uncoupling protein 2 (UCP2) -related diseases by inhibition of natural antisense transcripts against UCP2.
EP2516648B1 (en) 2009-12-23 2017-11-08 CuRNA, Inc. Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
CA2785173A1 (en) 2009-12-29 2011-07-28 Curna, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
KR101853508B1 (en) 2009-12-29 2018-06-20 큐알엔에이, 인크. TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
CN102791862B (en) 2009-12-31 2017-04-05 库尔纳公司 IRS2 relevant diseases are treated by suppressing the natural antisense transcript of insulin receptor substrate2 (IRS2) and transcription factor E3 (TFE3)
NO2521784T3 (en) 2010-01-04 2018-05-05
CN102822342B (en) 2010-01-06 2017-05-10 库尔纳公司 Treatment of pancreatic developmental gene related diseases by inhibition of natural antisense transcript to a pancreatic developmental gene
US9200277B2 (en) 2010-01-11 2015-12-01 Curna, Inc. Treatment of sex hormone binding globulin (SHBG) related diseases by inhibition of natural antisense transcript to SHBG
US8946182B2 (en) 2010-01-25 2015-02-03 Curna, Inc. Treatment of RNASE H1 related diseases by inhibition of natural antisense transcript to RNASE H1
EP2539452B1 (en) 2010-02-22 2016-07-27 CuRNA, Inc. Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
KR101877065B1 (en) 2010-04-02 2018-07-10 큐알엔에이, 인크. Treatment of colony-stimulating factor 3 (csf3) related diseases by inhibition of natural antisense transcript to csf3
RU2610661C2 (en) 2010-04-09 2017-02-14 Курна, Инк. Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
JP2013525483A (en) 2010-05-03 2013-06-20 カッパーアールエヌエー,インコーポレイテッド Treatment of sirtuin (SIRT) related diseases by inhibition of natural antisense transcripts against sirtuin (SIRT)
TWI531370B (en) 2010-05-14 2016-05-01 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
US8895528B2 (en) 2010-05-26 2014-11-25 Curna, Inc. Treatment of atonal homolog 1 (ATOH1) related diseases by inhibition of natural antisense transcript to ATOH1
WO2011150007A2 (en) 2010-05-26 2011-12-01 Opko Curna Llc Treatment of methionine sulfoxide reductase a (msra) related diseases by inhibition of natural antisense transcript to msra
RU2588654C2 (en) 2010-06-23 2016-07-10 Курна, Инк. Treatment of sodium channel, voltage-gated, alpha subunit (scna) related diseases by inhibition of natural antisense transcript to scna
EP2593547B1 (en) 2010-07-14 2017-11-15 CuRNA, Inc. Treatment of discs large homolog (dlg) related diseases by inhibition of natural antisense transcript to dlg
CN103210086B (en) 2010-10-06 2017-06-09 库尔纳公司 NEU4 relevant diseases are treated by suppressing the natural antisense transcript of sialidase 4 (NEU4)
CN103180445B (en) 2010-10-22 2018-02-16 库尔纳公司 IDUA relevant diseases are treated by suppressing the natural antisense transcript of α L iduronases (IDUA)
WO2012068340A2 (en) 2010-11-18 2012-05-24 Opko Curna Llc Antagonat compositions and methods of use
JP6071893B2 (en) 2010-11-23 2017-02-01 カッパーアールエヌエー,インコーポレイテッド Treatment of NANOG-related diseases by inhibition of natural antisense transcripts to NANOG
CN103620036B (en) 2011-06-09 2016-12-21 库尔纳公司 FXN relevant disease is treated by the natural antisense transcript of suppression Frataxin (FXN)
CN103874486A (en) 2011-09-06 2014-06-18 库尔纳公司 Treatment of diseases related to alpha subunits of sodium channels, voltage-gated (scnxa) with small molecules
TR201815503T4 (en) 2012-03-15 2018-11-21 Curna Inc Treatment of brain-mediated neurotrophic factor (bknf) related diseases by inhibition of the native antisense transcript to bknf.
CN102628060A (en) * 2012-03-22 2012-08-08 北京济福霖生物技术有限公司 Production method of low-fat milk
JP6362268B2 (en) 2012-11-05 2018-07-25 塩野義製薬株式会社 Method for evaluating drug efficacy and screening method for EL activity inhibitory substance having therapeutic or preventive effect on diseases associated with EL activity
BR112015022009A2 (en) 2013-03-14 2017-08-29 Shionogi & Co MONOCLONAL ANTIBODY, INHIBITING THE ENZYME ACTIVITY OF VASCULAR ENDOTHELIAL LIPASE
TWI688575B (en) 2014-09-11 2020-03-21 日商塩野義製藥股份有限公司 Humanized monoclonal antibody, inhibiting the enzymatic activity of vascular endothelial lipase
WO2022165313A1 (en) 2021-02-01 2022-08-04 Regenxbio Inc. Gene therapy for neuronal ceroid lipofuscinoses
JPWO2023008337A1 (en) * 2021-07-26 2023-02-02

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU679525B2 (en) * 1992-12-04 1997-07-03 Innovir Laboratories, Inc. Regulatable nucleic acid therapeutic and methods of use thereof
DE69732789T2 (en) * 1996-12-06 2005-08-11 Aventis Pharmaceuticals Inc. HUMAN LIPASE-LIKE GENE GENE-CODED POLYPEPTIDES, AND COMPOSITIONS AND METHODS
AU1941899A (en) * 1997-12-19 1999-07-12 Progenitor, Inc. A lipase expressed in endothelial cells and methods for its use

Also Published As

Publication number Publication date
ZA200107598B (en) 2003-05-28
NO331779B1 (en) 2012-03-26
HK1043309A1 (en) 2002-09-13
CA2363486A1 (en) 2000-10-05
CA2363486C (en) 2012-12-18
KR20020029651A (en) 2002-04-19
MXPA01009727A (en) 2002-07-22
NO20014657L (en) 2001-11-21
EP1171078A4 (en) 2002-11-06
BR0009333A (en) 2002-01-08
KR20050044812A (en) 2005-05-12
IL145526A (en) 2010-11-30
NO20014657D0 (en) 2001-09-25
IL145526A0 (en) 2002-06-30
NO20100214L (en) 2001-11-21
NZ514350A (en) 2004-12-24
KR100887164B1 (en) 2009-03-10
WO2000057837A2 (en) 2000-10-05
AU3918700A (en) 2000-10-16
WO2000057837A8 (en) 2001-09-27
NO331784B1 (en) 2012-03-26
WO2000057837A3 (en) 2001-01-25
EP1171078A2 (en) 2002-01-16
WO2000057837A9 (en) 2001-10-18
JP2002540127A (en) 2002-11-26
AU776684B2 (en) 2004-09-16

Similar Documents

Publication Publication Date Title
AU776684B2 (en) Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI, very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
US7008776B1 (en) Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
US8343494B2 (en) Antibodies against LLG polypeptides of the triacylglycerol lipase family
WO1998024888A9 (en) Polypeptides encoded by a human lipase-like gene, compositions and methods

Legal Events

Date Code Title Description
ASS Change of ownership

Owner name: AVENTIS PHARMACEUTICALS INC, US

Free format text: OLD OWNER(S): AVENTIS PHARMACEUTICALS INC.; THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA

PSEA Patent sealed
S35A Application for proceedings under section 35 (restoration of lapsed patents)
RENW Renewal (renewal fees accepted)
S35R Proceedings under section 35 (restoration of lapsed patents): patent restored
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)

Free format text: PATENT RENEWED FOR 7 YEARS UNTIL 24 MAR 2020 BY CPA GLOBAL

Effective date: 20130211

EXPY Patent expired