WO2003051911A2 - Polypeptides et polynucleotides gmg-5 et utilisations de ceux-ci - Google Patents

Polypeptides et polynucleotides gmg-5 et utilisations de ceux-ci Download PDF

Info

Publication number
WO2003051911A2
WO2003051911A2 PCT/IB2002/004806 IB0204806W WO03051911A2 WO 2003051911 A2 WO2003051911 A2 WO 2003051911A2 IB 0204806 W IB0204806 W IB 0204806W WO 03051911 A2 WO03051911 A2 WO 03051911A2
Authority
WO
WIPO (PCT)
Prior art keywords
gmg
polypeptide
polypeptides
sequence
insulin
Prior art date
Application number
PCT/IB2002/004806
Other languages
English (en)
Other versions
WO2003051911A3 (fr
Inventor
Hiroaki Tanaka
Deno Dialynas
Aaron Scalia
Original Assignee
Genset S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genset S.A. filed Critical Genset S.A.
Priority to AU2002339697A priority Critical patent/AU2002339697A1/en
Publication of WO2003051911A2 publication Critical patent/WO2003051911A2/fr
Publication of WO2003051911A3 publication Critical patent/WO2003051911A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • 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

Definitions

  • the present invention relates to the field of metabolic research, in particular the discovery of compounds effective for reducing body mass and useful for treating metabolic-related diseases and disorders.
  • the metabolic-related diseases or disorders envisioned to be treated by the methods of the invention include, but are not limited to, hyperlipidemia, atherosclerosis, diabetes, and hypertension.
  • Obesity is a public health problem that is serious, widespread, and increasing. In the United States, 20 percent of the population is obese; in Europe, a slightly lower percentage is obese (Friedman (2000) Nature 404:632-634). Obesity is associated with increased risk of hypertension, cardiovascular disease, diabetes, and cancer as well as respiratory complications and osteoarthritis (Kopelman (2000) Nature 404:635-643). Even modest weight loss ameliorates these associated conditions.
  • leptin ob and its receptor (db)
  • pro-opiomelanocortin Pome
  • melanocortin-4-receptor Mc4r
  • agouti protein A>
  • carboxypeptidase E fat
  • 5-hydroxytryptamine receptor 2C Htr2c
  • nescient basic helix-loop-helix 2 Nhlhl
  • prohormone convertase 1 IPCSKl prohormone convertase 1 IPCSKl
  • tubby tubby protein
  • GMG-5 Genset Metabolic Gene-5
  • GMG-5 polypeptide is comprised of a C-terminal globular Clq homology domain preceded by an N-terminal amino acid sequence that lacks a recognizable signal peptide (SignalP).
  • the invention includes polypeptides encoded by GMG-5, which include both the full length polynucleotide sequence and polypeptide fragments thereof, preferably said fragments comprise all or part of the Clq homology domain.
  • the GMG-5 polypeptide fragments containing all or part of the Clq homology domain are believed to possess unexpected effects in vitro and in vivo, in terms of their increased biological activity as described herein, including utility for weight reduction, prevention of weight gain and control of blood glucose levels in humans and other mammals. More specifically, the biological activities of the GMG-5 polypeptides, especially fragments, include reduction of elevated free fatty acid levels caused by administration of epinephrine, t.v. injection of "intralipid", or administration of a high fat test meal, as well as increased fatty acid oxidation in muscle cells, reduction in glucose levels, modulation of energy expenditure, resistance to insulin and weight reduction in mammals consuming a high fat/high sucrose diet.
  • GMG-5 polypeptide lacks a recognizable signal peptide (SignalP)
  • the inventors believe that GMG-5 polypeptide and fragments thereof comprising the C-terminal Clq homology domain have novel extracellular utility for the treatment of obesity and diabetes as well as disorders associated therewith.
  • the instant invention therefore includes not only GMG-5 polypeptide, fragments of GMG-5 polypeptide, and polynucleotides encoding the GMG-5 polypeptide and fragments thereof, but also GMG-5 polypeptide and GMG-5 polypeptide fragments modified to include a recognizable signal peptide.
  • the invention further includes the polynucleotides that encode the GMG-5 polypeptide and GMG-5 polypeptide fragments modified to include a recognizable signal peptide. Modification by signal peptide is intended facilitate the extracellular localization of GMG-5 polypeptide or fragments thereof comprising the C-terminal Clq homology domain.
  • the invention is drawn to GMG-5 polypeptides, polynucleotides encoding said GMG- 5 polypeptides, vectors comprising said GMG-5 polynucleotides, and cells recombinant for said GMG-5 polynucleotides, as well as to pharmaceutical and physiologically acceptable compositions comprising said GMG-5 polypeptides and methods of administering said GMG-5 pharmaceutical and physiologically acceptable compositions in order to reduce body weight or to treat metabolic- related diseases and disorders.
  • Assays for identifying agonists and antagonists of metabolic-related activity are also part of the invention.
  • the invention features purified, isolated, or recombinant GMG-5 polypeptides that have lipid partitioning, lipid metabolism, and insulin-like activities.
  • GMG-5 polypeptides may include a recognizable signal peptide that has been added to the N-terminal end of a polypeptide of the invention or fragment thereof.
  • Preferred GMG-5 polypeptide fragments have the same or greater activity than a full-length GMG-5 polypeptide, wherein said activity is also selected from the group consisting of lipid partitioning, lipid metabolism, and insulin- like activity.
  • said polypeptide fragment comprises, consists essentially of, or consists of, at least 6 consecutive amino acids and not more than 158, 157, 178, 177, 163, 156 or 150 consecutive amino acids of SEQ ID NOs: 2, 4, 6, 8, 10, 12 or 14, respectively.
  • GMG-5 polypeptide fragments are said selected GMG-5 polypeptide fragments made resistant to dipeptidyl peptidase cleavage by N-termmal modification.
  • said polypeptide fragment comprises an ammo acid sequence at least
  • the invention further provides a purified or isolated polypeptide comprising, consisting of, or consisting essentially of an ammo acid sequence selected from the group consisting of: (a) a full length GMG-5 polypeptide of SEQ ID NOs: 2, 4, 6, 8, 10, 12 or 14; (b) a full length GMG-5 polypeptide of SEQ ID NOs: 2, 6, 8, 10, 12 or 14 absent the N-termmal Met; (c) a mature GMG-5 polypeptide of SEQ ID NOs: 6, 8, 10, 12 or 14 lacking signal peptide; (d) a GMG-5 polypeptide of
  • GMG-5 polypeptide is of any one integer in length between 6 amino acids and 158, 157, 178, 177, 163, 156 or 150 amino acids (full length) inclusive of SEQ ID NOs: 2, 4, 6, 8, 10, 12 or 14, respectively;
  • the invention further provides for fragments of the polypeptides of (a)-(f) above, such as those having biological activity or comprising biologically functional doma ⁇ n(s).
  • GMG-5 polypeptides are able to lower circulating (either blood, serum or plasma) levels (concentration) of: (l) free fatty acids, (n) glucose, and/or (in) tnglycendes.
  • Further preferred polypeptides of the invention demonstrating free fatty acid level lowenng activity, glucose level lowering activity, and/or tnglycende level lowering activity have an activity that is the same or greater than full length GMG-5 polypeptides at the same molar concentration, have the same or greater than transient activity and/or have a sustained activity.
  • GMG-5 polypeptides are those that significantly stimulate muscle lipid or free fatty acid oxidation. Further preferred GMG-5 polypeptides are those that significantly stimulate muscle lipid or free fatty acid oxidation.
  • GMG-5 polypeptides are those that cause C2C12 cells differentiated in the presence of said polypeptides to undergo at least 10%, 20%, 30% , 35%, or 40% more oleate oxidation as compared to untreated cells. Further preferred GMG-5 polypeptides are those that increase leptin uptake in a liver cell line (preferably BPRCL mouse liver cells (ATCC CRL-2217)).
  • GMG-5 polypeptides are those that significantly reduce the postprandial increase in plasma free fatty acids due to a high fat meal.
  • GMG-5 polypeptides are those that significantly reduce or eliminate ketone body production as the result of a high fat meal.
  • GMG-5 polypeptides are those that increase glucose uptake in skeletal muscle cells. Further preferred GMG-5 polypeptides are those that increase glucose uptake in adipose cells.
  • GMG-5 polypeptides are those that increase glucose uptake in neuronal cells. Further preferred GMG-5 polypeptides are those that increase glucose uptake in red blood cells.
  • Further preferred GMG-5 polypeptides are those that increase glucose uptake in the brain. Further preferred GMG-5 polypeptides are those that significantly reduce the postprandial increase in plasma glucose following a meal, particularly a high carbohydrate meal. Further preferred GMG-5 polypeptides are those that significantly prevent the postprandial increase in plasma glucose following a meal, particularly a high fat or a high carbohydrate meal. Further preferred GMG-5 polypeptides are those that increase insulin sensitivity. Further preferred GMG-5 polypeptide fragments are those that inhibit the progression from impaired glucose tolerance to insulin resistance. Further preferred GMG-5 polypeptides are those that form multimers (e.g., heteromultimers or homomultimers) in vitro and/or in vivo.
  • multimers e.g., heteromultimers or homomultimers
  • Preferred multimers are homodimers or homotrimers.
  • Other preferred multimers are homomultimers comprising at least 4, 6, 8, 9, 10 or 12 GMG-5 polypeptide subunits.
  • Other preferred mulimers are hetero multimers comprising a GMG-5 polypeptide of the invention. Further preferred embodiments include heterologous polypeptides comprising one of the
  • GMG-5 polypeptides of the invention are GMG-5 polypeptides of the invention.
  • the invention features purified, isolated, or recombinant polynucleotides encoding said GMG-5 polypeptides described in the first aspect, or the complement thereof.
  • a further preferred embodiment of the invention is a recombinant, purified or isolated polynucleotide comprising, or consisting of a mammalian genomic sequence, gene, or fragments thereof. In one aspect the sequence is derived from a human, mouse or other mammal.
  • the genomic sequence includes isolated, purified, or recombinant polynucleotides comprising a contiguous span of at least 12, 15, 18, 20, 22, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 400 or 500 nucleotides of any one of the polynucleotide sequences described in SEQ ID NOs: 1, 3, 5, 7, 9, 11 or 13, or the complements thereof, wherein said contiguous span comprises a nucleotide sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the corresponding nucleotide sequence of the Clq homology domains of SEQ ID NOs: 1, 3, 5, 7, 9, 11 or 13.
  • the polynucleotides are DNA, RNA, DNA/RNA hybrids, single- stranded, and double-stranded.
  • the invention features a recombinant vector comprising, consisting essentially of, or consisting of, said polynucleotide described in the second aspect.
  • the invention features a recombinant cell comprising, consisting essentially of, or consisting of, said recombinant vector described in the third aspect.
  • a further embodiment includes a host cell recombinant for a polynucleotide of the invention.
  • the invention features a pharmaceutical or physiologically acceptable composition
  • a pharmaceutical or physiologically acceptable composition comprising, consisting essentially of, or consisting of, said GMG-5 polypeptides described in the first aspect and, alternatively, a pharmaceutical or physiologically acceptable diluent.
  • the invention features a method of reducing body mass comprising providing or administering to individuals in need of reducing body mass said pharmaceutical or physiologically acceptable composition described in the fifth aspect.
  • the identification of said individuals in need of reducing body mass to be treated with said pharmaceutical or physiologically acceptable composition comprises genotyping GMG-5 single nucleotide polymorphisms (SNPs) or measuring metabolic polypeptide or mRNA levels in clinical samples from said individuals.
  • SNPs single nucleotide polymorphisms
  • said clinical samples are selected from the group consisting of plasma, urine, and saliva.
  • a GMG-5 polypeptide fragment of the present invention is administered to an individual with at least a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in blood, serum or plasma levels of full length any one or all of the GMG-5 polypeptides or the naturally proteolytically cleaved GMG-5 fragments as compared to healthy, non-obese patients.
  • the invention features a method of preventing or treating a metabolic- related disease or disorder comprising providing or administering to an individual in need of such treatment said pharmaceutical or physiologically acceptable composition described in the fifth aspect.
  • the identification of said individuals in need of such treatment to be treated with said pharmaceutical or physiologically acceptable composition comprises genotyping GMG-5 single nucleotide polymorphisms (SNPs) or measuring GMG-5 polypeptide or mRNA levels in clinical samples from said individuals.
  • SNPs single nucleotide polymorphisms
  • said clinical samples are selected from the group consisting of blood, serum, plasma, urine, and saliva.
  • said metabolic-related disease or disorder is selected from the group consisting of obesity, impaired glucose tolerance, insulin resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, non-insulin-dependent diabetes and Type II diabetes.
  • Type II diabetes-related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other metabolic-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • metabolic-related diseases or disorders of the invention include cachexia, wasting, AJDS-related weight loss, cancer-related weight loss, visceral obesity, anorexia, and bulimia.
  • said individual is a mammal, preferably a human.
  • embodiments of the present invention include methods of causing or inducing a desired biological response in an individual comprising the steps of: providing or administering to an individual a composition comprising a GMG-5 polypeptide, wherein said biological response is selected from the group consisting of:
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control blood glucose in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) in combination with insulin therapy.
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • IDDM Insulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control body weight, particularly visceral obesity, in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) in combination with insulin therapy.
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control body weight in some persons with Insulin Dependent Diabetes Mellitus (IDDM, Type I diabetes) in combination with insulin therapy.
  • IDDM Insulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control blood glucose in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) alone, without combination of insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control blood glucose in some persons with Insulin Dependent Diabetes Mellitus (IDDM, Type I diabetes) alone, without combination of insulin therapy.
  • IDDM Insulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control body weight in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) alone, without combination of insulin therapy.
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control body weight in some persons with Insulin Dependent Diabetes Mellitus (IDDM, Type I diabetes) alone, without combination of insulin therapy.
  • IDDM Insulin Dependent Diabetes Mellitus
  • the present invention may be used in complementary therapy of NIDDM patients to improve their weight or glucose control in combination with an insulin secretagogue (preferably oral form) or an insulin sensitising (preferably oral form) agent.
  • an insulin secretagogue preferably oral form
  • an insulin sensitising preferably oral form
  • the oral insulin secretagogue is 1,1 -dimethyl -2-(2-morpholino phenyl)guanidine fumarate (BTS67582) or a sulphonylurea selected from tolbutamide, tolazamide, chlorpropamide, glibenclamide, glimepiride, glipizide and glidazide.
  • the insulin sensitising agent is selected from metformin, ciglitazone, troglitazone and pioglitazone.
  • the present invention further provides a method of improving the body weight or glucose control of NIDDM patients alone, without an insulin secretagogue or an insulin sensitising agent.
  • the present invention may be used in complementary therapy of IDDM patients to improve their weight or glucose control in combination with an insulin secretagogue (preferably oral form) or an insulin sensitising (preferably oral form) agent.
  • the insulin secretagogue is 1,1 -dimethyl -2-(2-morpholino phenyl)guanidine fumarate
  • the insulin sensitising agent is selected from metformin, ciglitazone, troglitazone and pioglitazone.
  • the present invention further provides a method of improving the body weight or glucose control of IDDM patients alone, without an insulin secretagogue or an insulin sensitising agent.
  • the present invention may be administered either concomitantly or concurrently, with the insulin secretagogue or insulin sensitising agent for example in the form of separate dosage units to be used simultaneously, separately or sequentially (either before or after the secretagogue or either before or after the sensitising agent).
  • the present invention further provides for a composition of pharmaceutical or physiologically acceptable composition and an insulin secretagogue or insulin sensitising agent as a combined preparation for simultaneous, separate or sequential use for the improvement of body weight or glucose control in NIDDM or IDDM patients.
  • the present invention of said pharmaceutical or physiologically acceptable composition further provides a method for the use as an insulin sensitiser.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) in combination with insulin therapy.
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some persons with Insulin Dependent Diabetes Mellitus (IDDM, Type I diabetes) in combination with insulin therapy.
  • IDDM Insulin Dependent Diabetes Mellitus
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some persons with Noninsulin Dependent Diabetes Mellitus (NIDDM, Type II diabetes) without insulin therapy.
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • the invention features a method of making the GMG-5 polypeptides described in the first aspect, wherein said method is selected from the group consisting of: proteolytic cleavage, recombinant methodology and artificial synthesis.
  • the present invention provides a method of making a recombinant GMG-5 polypeptide fragment or a full length GMG-5 polypeptide, the method comprising providing a transgenic, non-human mammal whose milk contains said recombinant GMG-5 polypeptide fragment or full-length protein, and purifying said recombinant GMG-5 polypeptide fragment or said full-length GMG-5 polypeptide from the milk of said non-human mammal.
  • said non-human mammal is a cow, goat, sheep, rabbit, or mouse.
  • the method comprises purifying a recombinant full-length GMG-5 polypeptide from said milk, and further comprises cleaving said protein in vitro to obtain a desired GMG-5 polypeptide fragment.
  • the invention features a purified or isolated antibody capable of specifically binding to a polypeptide of the present invention.
  • the antibody is capable of binding to a polypeptide comprising at least 6 consecutive amino acids, at least 8 consecutive amino acids, or at least 10 consecutive amino acids of the sequence of one of the polypeptide sequences described in SEQ ID NOs: 2, 4, 6, 8, 10, 12 or 14.
  • the invention features a use of the polypeptide described in the first aspect for treatment of metabolic-related diseases and disorders and/or reducing or increasing body mass.
  • said metabolic-related diseases and disorders are selected from the group consisting of obesity, insulin resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, non-insulin-dependent diabetes and Type II diabetes.
  • Type II diabetes-related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other metabolic-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • Yet other metabolic-related diseases or disorders of the invention include cachexia, wasting, AIDS- related weight loss, anorexia, and bulimia.
  • said individual is a mammal, preferably a human.
  • the invention provides a polypeptide of the first aspect of the invention, or a composition of the fifth aspect of the invention, for use in a method of treatment of the human or animal body.
  • the invention features methods of reducing body weight for cosmetic purposes comprising providing to an individual said pharmaceutical or physiologically acceptable composition described in the fifth aspect, or a polypeptide described in the first aspect.
  • said individual has a BMI of at least 20 and no more than 25.
  • said individual preferably has a BMI of at least 15 and no more than 20.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect for reducing body mass and/or for treatment or prevention of metabolic-related diseases or disorders.
  • said metabolic-related disease or disorder is selected from the group consisting of obesity, impaired glucose tolerance, insulin resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, non-insulin-dependent diabetes and Type II diabetes.
  • Type ⁇ diabetes-related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other metabolic-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • metabolic-related diseases or disorders of the invention include cachexia, wasting, AIDS-related weight loss, cancer-related weight loss, visceral obesity, anorexia, and bulimia.
  • said individual is a mammal, preferably a human.
  • the identification of said individuals to be treated with said pharmaceutical or physiologically acceptable composition comprises genotyping
  • GMG-5 single nucleotide polymorphisms SNPs
  • measuring GMG-5 polypeptides or mRNA levels in clinical samples from said individuals are selected from the group consisting of blood, serum, plasma, urine, and saliva.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect for reducing body weight for cosmetic reasons.
  • the invention features methods of treating insulin resistance comprising providing to an individual said pharmaceutical or physiologically acceptable composition described in the fifth aspect, or a polypeptide described in the first aspect.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect in a method of treating individuals with normal glucose tolerance (NGT) who are obese or who have fasting hyperinsulinemia, or who have both.
  • NTT normal glucose tolerance
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect in a method of treating individuals with gestational diabetes.
  • Gestational diabetes refers to the development of diabetes in an individual during pregnancy, usually during the second or third trimester of pregnancy.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect in a method of treating individuals with impaired fasting glucose (IFG).
  • Impaired fasting glucose (IFG) is that condition in which fasting plasma glucose levels in an individual are elevated but not diagnostic of overt diabetes, i.e. plasma glucose levels of less than 126 mg/dl and less than or equal to 110 mg/dl.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect in a method of treating and preventing impaired glucose tolerance (IGT) in an individual.
  • IGT impaired glucose tolerance
  • the invention provides methods for reducing and/or preventing the appearance of Insulin-Resistance Syndrome.
  • the invention features the pharmaceutical or physiologically acceptable composition described in the fifth aspect in a method of treating a subject having polycystic ovary syndrome (PCOS).
  • PCOS is among the most common disorders of premenopausal women, affecting 5-10% of this population.
  • Insulin-sensitizing agents e.g., troglitazone
  • the invention provides methods for reducing insulin resistance, normalizing blood glucose thus treating and/or preventing PCOS.
  • the invention features the pharmaceutical or physiologically acceptable composition descnbed in the fifth aspect in a method of treating a subject having insulin resistance.
  • a subject having insulin resistance is treated according to the methods of the invention to reduce or cure the insulm-resistance.
  • prevention or reducing insulm resistance according to the methods of the invention may prevent or reduce infections and cancer.
  • the methods of the invention are used to prevent the development of insulm resistance in a subject, e g., those known to have an increased risk of developing insulin-resistance.
  • any of the above-descnbed tests or other tests known in the art can be used to determine that a subject is msuhn-resistant, which patient can then be treated according to the methods of the invention to reduce or cure the insulin-resistance.
  • the methods of the invention can also be used to prevent the development of insulm resistance in a subject, e g , those known to have an increased nsk of developing insulin-resistance.
  • the invention features a method of using a GMG-5 polypeptide fragment m a method of screening compounds for one or more antagonists of dipeptidyl peptidase cleavage said polypeptide fragment
  • said compound is selected from but is not restncted to small molecular weight organic or inorganic compound, protein, peptide, carbohydrate, or lipid.
  • said polypeptide fragment is a GMG-5 polypeptide fragment.
  • said antagonist of dipeptidyl peptidase cleavage of said GMG-5 polypeptide fragment is specific for said dipeptidyl peptidase. In further preferred embodiment, said antagonist of dipeptidyl peptidase cleavage of said
  • GMG-5 polypeptide fragment is specific for said GMG-5 polypeptide fragment.
  • the invention features a method of using a GMG-5 polypeptide fragment in a method of screening compounds for one or more antagonists of GMG-5 activity, wherein said activity is selected from but not restncted to lipid partitioning, lipid metabolism, and insulin-like activity.
  • said compound is selected from but is not restricted to small molecular weight organic or inorganic compound, protein, peptide, carbohydrate, or lipid.
  • the invention features a method of adding a signal peptide to the N- terminal end of a polypetide of the invention, wherein said signal peptide serves to facilitate secretion of said polypeptide.
  • said signal peptide is selected from ammo acids 1-20 of SEQ ID NOs: 6, 8, 10, 12 or 14 (MVRMVPVLLSLLLLLGPAVP).
  • the amount of GMG-5 polypeptide or polynucleotide administered to an individual is sufficient to bring circulating (blood, serum, or plasma) levels (concentration) of GMG-5 polypeptides to their normal levels (levels in non-obese individuals). "Normal levels" may be specified as the total concentration of all circulating GMG-5 polypeptides (full length GMG-5 proteins and fragments thereof) or the concentration of all circulating proteolytically cleaved GMG-5 polypeptides only.
  • weight loss is due in part or in whole to a decrease in mass of either a) subcutaneous adipose tissue and/or b) viseral
  • SEQ ID NO:l represents the cDNA sequence of GMG-5.
  • SEQ ID NO:2 represents the amino acid sequence encoded by the cDNA of SEQ ID NO: 1.
  • SEQ ID NO: 3 represents the cDNA sequence of GMG-5 minus the N-terminal Met.
  • SEQ ED NO:4 represents the amino acid sequence encoded by the cDNA of SEQ ID NO:3.
  • SEQ ED NO: 5 represents the cDNA sequence of a GMG-5 plus a recognizable signal sequence.
  • SEQ ID NO: 6 represents the amino acid sequence encoded by the cDNA of SEQ ID NO: 5.
  • SEQ ID NO: 7 represents the cDNA sequence of a GMG-5 minus the N-terminal Met and plus a recognizable signal sequence.
  • SEQ ED NO: 8 represents the amino acid sequence encoded by the cDNA of SEQ ED NO:7.
  • SEQ ED NO:9 represents the cDNA sequence of a GMG-5 fragment plus a recognizable signal sequence.
  • SEQ ED NO: 10 represents the amino acid sequence encoded by the cDNA of SEQ ED NO:S
  • SEQ ED NO: 11 represents the cDNA sequence of a GMG-5 fragment plus a recognizable signal sequence.
  • SEQ ED NO: 12 represents the amino acid sequence encoded by the cDNA of SEQ ED NO: l l.
  • SEQ ED NO: 13 represents the cDNA sequence of a GMG-5 fragment plus a recognizable signal sequence.
  • SEQ ID NO: 14 represents the amino acid sequence encoded by the cDNA of SEQ ED NO: 13.
  • oligonucleotides and “polynucleotides” and nucleic acid include RNA, DNA, or RNA/DNA hybrid sequences of more than one nucleotide in either single chain or duplex form.
  • modified nucleotides which comprise at least one modification, including by way of example and not limitation: (a) an alternative linking group, (b) an analogous form of purine, (c) an analogous form of pyrimidine, or (d) an analogous sugar.
  • polynucleotide sequences of the invention may be prepared by any known method, including synthetic, recombinant, ex vivo generation, or a combination thereof, as well as utilizing any purification methods known in the art.
  • polynucleotide construct, recombinant polynucleotide and recombinant polypeptide are used herein consistently with their use in the art.
  • upstream and downstream are also used herein consistently with their use in the art.
  • base paired and “Watson & Crick base paired” are used interchangeably herein and consistently with their use in the art.
  • base paired and “Watson & Crick base paired” are used interchangeably herein and consistently with their use in the art.
  • complementary and “complementary thereof , “complement”, “complementary polynucleotide”, “complementary nucleic acid” and “complementary nucleotide sequence” are used interchangeably herein and consistently with their use in the art.
  • purified is used herein to describe a polynucleotide or polynucleotide vector of the invention that has been separated from other compounds including, but not limited to, other nucleic acids, carbohydrates, lipids and proteins (such as the enzymes used in the synthesis of the polynucleotide). Purified can also refer to the separation of covalently closed polynucleotides from linear polynucleotides, or vice versa, for example.
  • a polynucleotide is substantially pure when at least about 50%, 60%, 75%, or 90% of a sample contains a single polynucleotide sequence. In some cases this involves a determination between conformations (linear versus covalently closed).
  • a substantially pure polynucleotide typically comprises about 50, 60, 70, 80, 90, 95, 99% weight/weight of a nucleic acid sample.
  • Polynucleotide purity or homogeneity may be indicated by a number of means well known in the art, such as agarose or polyacrylamide gel electrophoresis of a sample, followed by visualizing a single polynucleotide band upon staining the gel. For certain purposes higher resolution can be provided by using HPLC or other means well known in the art.
  • purified is used herein to describe a polypeptide of the invention that has been separated from other compounds including, but not limited to, nucleic acids, lipids, carbohydrates and other proteins.
  • a polypeptide is substantially pure when at least about 50%, 60%, 75%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 99.5% of the polypeptide molecules of a sample have a single amino acid sequence.
  • a substantially pure polypeptide typically comprises about 50%, 60%, 70%, 80%, 90% 95%, 96%, 97%, 98%, 99% or 99.5% weight/weight of a protein sample.
  • Polypeptide purity or homogeneity is indicated by a number of methods well known in the art, such as agarose or polyacrylamide gel electrophoresis of a sample, followed by visualizing a single polypeptide band upon staining the gel. For certain purposes higher resolution can be provided by using HPLC or other methods well known in the art.
  • purified does not require absolute purity; rather, it is intended as a relative definition. Purification of starting material or natural material to at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated. Alternatively, purification may be expressed as "at least" a percent purity relative to heterologous polynucleotides (DNA, RNA or both) or polypeptides. As a preferred embodiment, the polynucleotides or polypeptides of the present invention are at least; 10%, 20%, 30%,
  • polynucleotides or polypeptides have an "at least" purity ranging from any number, to the thousandth position, between 90% and 100% (e.g., at least 99.995% pure) relative to heterologous polynucleotides or polypeptides. Additionally, purity of the polynucleotides or polypeptides may be expressed as a percentage (as described above) relative to all materials and compounds other than the carrier solution. Each number, to the thousandth position, may be claimed as individual species of purity.
  • isolated requires that the material be removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated.
  • Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that the vector or composition is not part of its natural environment.
  • isolated are: naturally occurring chromosomes (e.g., chromosome spreads), artificial chromosome libraries, genomic libraries, and cDNA libraries that exist either as an in vitro nucleic acid preparation or as a transfected transformed host cell preparation, wherein the host cells are either an in vitro heterogeneous preparation or plated as a heterogeneous population of single colonies. Also specifically excluded are the above libraries wherein a 5' EST makes up less than 5% (or alternatively 1%, 2%, 3%, 4%, 10%, 25%, 50%, 75%, or 90%, 95%, or 99%) of the number of nucleic acid inserts in the vector molecules.
  • whole cell genomic DNA or whole cell RNA preparations including said whole cell preparations which are mechanically sheared or enzymatically digested.
  • whole cell preparations as either an in vitro preparation or as a heterogeneous mixture separated by electrophoresis (including blot transfers of the same) wherein the polynucleotide of the invention have not been further separated from the heterologous polynucleotides in the electrophoresis medium (e.g., further separating by excising a single band from a heterogeneous band population in an agarose gel or nylon blot).
  • primer denotes a specific oligonucleotide sequence which is complementary to a target nucleotide sequence and used to hybridize to the target nucleotide sequence.
  • a primer serves as an initiation point for nucleotide polymerization catalyzed by DNA polymerase, RNA polymerase, or reverse transcriptase.
  • probe denotes a defined nucleic acid segment (or nucleotide analog segment, e.g., PNA as defined hereinbelow) which can be used to identify a specific polynucleotide sequence present in a sample, said nucleic acid segment comprising a nucleotide sequence complementary to the specific polynucleotide sequence to be identified.
  • polypeptide refers to a polymer of amino acids without regard to the length of the polymer.
  • peptides, oligopeptides, and proteins are included within the definition of polypeptide. This term also does not specify or exclude post-expression modifications of polypeptides.
  • polypeptides that include the covalent attachment of glycosyl groups, acetyl groups, phosphate groups, lipid groups and the like are expressly encompassed by the term polypeptide.
  • polypeptides which contain one or more analogs of an amino acid (including, for example, non-naturally occurring amino acids, amino acids of a recognizable signal sequence, amino acids which only occur naturally in an unrelated biological system, modified amino acids from mammalian systems etc.), polypeptides with substituted linkages, as well as other modifications known in the art, both naturally occurring and non-naturally occurring.
  • the compounds/polypeptides of the invention are capable of modulating the partitioning of dietary lipids between the liver and peripheral tissues, and are thus believed to treat "diseases involving the partitioning of dietary lipids between the liver and peripheral tissues.
  • peripheral tissues is meant to include muscle and adipose tissue.
  • the compounds/polypeptides of the invention partition the dietary lipids toward the muscle. In alternative preferred embodiments, the dietary lipids are partitioned toward the adipose tissue. In other preferred embodiments, the dietary lipids are partitioned toward the liver. In yet other preferred embodiments, the compounds/polypeptides of the invention increase or decrease the oxidation of dietary lipids, preferably free fatty acids (FFA) by the muscle. Dietary lipids include, but are not limited to triglycerides and free fatty acids.
  • Preferred diseases believed to involve the partitioning of dietary lipids include obesity and obesity-related diseases and disorders such as obesity, impaired glucose tolerance, insulin resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, Noninsulin Dependent Diabetes Mellitus (NIDDM, or Type II diabetes) and Insulin Dependent Diabetes Mellitus (IDDM or Type I diabetes).
  • Diabetes-related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, retinopathy, neuropathy, and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other obesity-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • Yet other obesity-related diseases or disorders of the invention include cachexia, wasting, A DS-related weight loss, cancer-related weight loss, visceral obesity, anorexia, and bulimia.
  • heterologous when used herein, is intended to designate any polypeptide or polynucleotide other than a GMG-5 polypeptide or a polynucleotide encoding a GMG-5 polypeptide of the present invention.
  • host cell recombinant for a particular polynucleotide of the present invention, means a host cell that has been altered by the hands of man to contain said polynucleotide in a way not naturally found in said cell.
  • said host cell may be transiently or stably transfected or transduced with said polynucleotide of the present invention.
  • the term "obesity" as used herein is defined in the WHO classifications of weight (Kopelman (2000) Nature 404:635643). Underweight is less than 18.5 (thin); Healthy is 18.5-24.9 (normal); grade 1 overweight is 25.0-29.9 (overweight); grade 2 overweight is 30.0-39.0 (obesity); grade 3 overweight is greater than or equal to 40.0 BMI.
  • BMI body mass index (morbid obesity) and is kg/m 2 .
  • Waist circumference can also be used to indicate a risk of metabolic complications where in men a circumference of greater than or equal to 94 cm indicates an increased risk, and greater than or equal to 102 cm indicates a substantially increased risk.
  • 88 cm indicates an increased risk
  • the waist circumference is measured in cm at midpoint between lower border of ribs and upper border of the pelvis.
  • Other measures of obesity include, but are not limited to, skinfold thickness which is a measurement in cm of skinfold thickness using calipers, and bioimpedance, which is based on the principle that lean mass conducts current better than fat mass because it is primarily an electrolyte solution; measurement of resistance to a weak current (impedance) applied across extremities provides an estimate of body fat using an empirically derived equation.
  • diabetes as used herein is intended to encompass the usual diagnosis of diabetes made from any of the methods included, but not limited to, the following list: symptoms of diabetes (eg. polyuria, polydipsia, polyphagia) plus casual plasma glucose levels of greater than or equal to 200 mg/dl, wherein casual plasma glucose is defined any time of the day regardless of the timing of meal or drink consumption; 8 hour fasting plasma glucose levels of less than or equal to 126 mg/dl; and plasma glucose levels of greater than or equal to 200 mg/dl 2 hours following oral administration of 75 g anhydrous glucose dissolved in water.
  • symptoms of diabetes eg. polyuria, polydipsia, polyphagia
  • IGT impaired glucose tolerance
  • IGT is diagnosed by a procedure wherein an affected person's postprandial glucose response is determined to be abnormal as assessed by 2-hour postprandial plasma glucose levels.
  • a measured amount of glucose is given to the patient and blood glucose levels measured regular intervals, usually every half hour for the first two hours and every hour thereafter.
  • glucose levels rise during the first two hours to a level less than 140 mg/dl and then drop rapidly.
  • the blood glucose levels are higher and the drop-off level is at a slower rate.
  • Insulin-Resistance Syndrome is intended to encompass the cluster of abnormalities resulting from an attempt to compensate for insulin resistance that sets in motion a series of events that play an important role in the development of both hypertension and coronary artery disease (CAD), such as premature atherosclerotic vascular disease.
  • CAD coronary artery disease
  • the invention provides methods for reducing and/or preventing the appearance of insulin-resistance syndrome.
  • PCOS polycystic ovary syndrome
  • Hyperandrogenism also is a feature of a variety of diverse insulin-resistant states, from the type A syndrome, through leprechaunism and lipoatrophic diabetes, to the type B syndrome, when these conditions occur in premenopausal women. It has been suggested that hyperinsulinemia per se causes hyperandrogenism. Insulin- sensitizing agents, e.g., troglitazone, have been shown to be effective in PCOS and that, in particular, the defects in insulin action, insulin secretion, ovarian steroidogenosis and fibrinolysis are improved (Ehrman et al. (1997) J Clin Invest 100: 1230), such as in insulin-resistant humans.
  • Insulin- sensitizing agents e.g., troglitazone
  • insulin resistance is intended to encompass the usual diagnosis of insulin resistance made by any of a number of methods, including but not restricted to: the intravenous glucose tolerance test or measurement of the fasting insulin level. It is well known that there is an excellent correlation between the height of the fasting insulin level and the degree of insulm resistance. Therefore, one could use elevated fasting msulm levels as a surrogate marker for insulin resistance for the purpose of identifying which normal glucose tolerance (NGT) individuals have insulin resistance. Another way to do this is to follow the approach as disclosed in The New
  • the target of the treatment according to the present invention can be defined as NGT individuals who are obese or who have fasting hypermsulmemia, or who have both.
  • a diagnosis of insulin resistance can also be made using the euglycemic glucose clamp test.
  • This test involves the simultaneous administration of a constant insulin infusion and a vanable rate glucose infusion. Dunng the test, which lasts 3-4 hours, the plasma glucose concentration is kept constant at euglycemic levels by measuring the glucose level every 5-10 minutes and then adjusting the vanable rate glucose infusion to keep the plasma glucose level unchanged. Under these circumstances, the rate of glucose entry into the bloodstream is equal to the overall rate of glucose disposal in the body. The difference between the rate of glucose disposal in the basal state (no msulm infusion) and the insulin infused state, represents insulm mediated glucose uptake.
  • the term "agent acting on the partitioning of dietary lipids between the liver and penpheral tissues" refers to a compound or polypeptide of the invention that modulates the partitioning of dietary lipids between the liver and the penpheral tissues as previously descnbed.
  • the agent increases or decreases the oxidation of dietary lipids, preferably free fatty acids (FFA) by the muscle.
  • FFA free fatty acids
  • the agent decreases or increases the body weight of individuals or is used to treat or prevent an obesity-related disease or disorder such as obesity, impaired glucose tolerance, msulm resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, Noninsulin Dependent Diabetes Mellitus (NIDDM, or Type II diabetes) and Insulin Dependent Diabetes Mellitus ( DDM or Type I diabetes).
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • DDM Insulin Dependent Diabetes Mellitus
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other obesity-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • Yet other obesity-related diseases or disorders of the invention include cachexia, wasting, AIDS-related weight loss, cancer-related weight loss, visceral obesity, anorexia, and bulimia.
  • response to an agent acting on the partitioning of dietary lipids between the liver and peripheral tissues refer to drug efficacy, including but not limited to, ability to metabolize a compound, ability to convert a pro-drug to an active drug, and the pharmacokinetics (abso ⁇ tion, distribution, elimination) and the pharmacodynamics (receptor-related) of a drug in an individual.
  • side effects to an agent acting on the partitioning of dietary lipids between the liver and peripheral tissues refer to adverse effects of therapy resulting from extensions of the principal pharmacological action of the drug or to idiosyncratic adverse reactions resulting from an interaction of the drug with unique host factors.
  • Side effects to an agent acting on the partitioning of dietary lipids between the liver and peripheral tissues can include, but are not limited to, adverse reactions such as dermatologic, hematologic or hepatologic toxicities and further includes gastric and intestinal ulceration, disturbance in platelet function, renal injury, nephritis, vasomotor rhinitis with profuse watery secretions, angioneurotic edema, generalized urticaria, and bronchial asthma to laryngeal edema and bronchoconstriction, hypotension, and shock.
  • adverse reactions such as dermatologic, hematologic or hepatologic toxicities and further includes gastric and intestinal ulceration, disturbance in platelet function, renal injury, nephritis, vasomotor rhinitis with profuse watery secretions, angioneurotic edema, generalized urticaria, and bronchial asthma to laryngeal edema and bronchoconstriction, hypotension, and shock.
  • GMG-5 -related diseases and disorders refers to any disease or disorder comprising an aberrant functioning of GMG-5, or which could be treated or prevented by modulating GMG-5 levels or activity.
  • Aberrant functioning of GMG-5" includes, but is not limited to, aberrant levels of expression of GMG-5 (either increased or decreased, but preferably decreased), aberrant activity of GMG-5 (either increased or decreased), and aberrant interactions with ligands or binding partners (either increased or decreased).
  • aberrant is meant a change from the type, or level of activity seen in normal cells, tissues, or patients, or seen previously in the cell, tissue, or patient prior to the onset of the illness.
  • these GMG-5-related diseases and disorders include obesity and the metabolic-related diseases and disorders described previously.
  • Cosmetic treatments is meant to include treatments with compounds or polypeptides of the invention that increase or decrease the body mass of an individual where the individual is not clinically obese or clinically thin.
  • these individuals have a body mass index (BMI) below the cut-off for clinical obesity (e.g. below 25 kg/m 2 ) and above the cut-off for clinical thinness (e.g. above 18.5 kg/m 2 ).
  • these individuals are preferably healthy (e.g. do not have an metabolic-related disease or disorder of the invention).
  • Cosmetic treatments are also meant to encompass, in some circumstances, more localized increases in adipose tissue, for example, gains or losses specifically around the waist or hips, or around the hips and thighs, for example. These localized gains or losses of adipose tissue can be identified by increases or decreases in waist or hip size, for example.
  • preventing refers to administering a compound prior to the onset of clinical symptoms of a disease or condition so as to prevent a physical manifestation of aberrations associated with obesity or GMG-5.
  • treating refers to administering a compound after the onset of clinical symptoms.
  • in need of treatment refers to a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, etc in the case of humans; veterinarian in the case of animals, including non-human mammals) that an individual or animal requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, but that include the knowledge that the individual or animal is ill, or will be ill, as the result of a condition that is treatable by the compounds of the invention.
  • a caregiver e.g. physician, nurse, nurse practitioner, etc in the case of humans; veterinarian in the case of animals, including non-human mammals
  • the term “perceives a need for treatment” refers to a sub-clinical determination that an individual desires to reduce weight for cosmetic reasons as discussed under “cosmetic treatment” above.
  • the term “perceives a need for treatment” in other embodiments can refer to the decision that an owner of an animal makes for cosmetic treatment of the animal.
  • mice preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the term may specify male or female or both, or exclude male or female.
  • non-human animal refers to any non-human vertebrate, including birds and more usually mammals, preferably primates, animals such as swine, goats, sheep, donkeys, horses, cats, dogs, rabbits or rodents, more preferably rats or mice. Both the terms “animal” and “mammal” expressly embrace human subjects unless preceded with the term “non-human”. The inventors believe GMG-5 polypeptides are able to significantly reduce the postprandial response of plasma free fatty acids, glucose, and triglycerides in mice fed a high fat/sucrose meal, while not affecting levels of leptin, insulin or glucagon.
  • GMG-5 polypeptides modulate muscle free fatty acid oxidation in vitro and ex vivo, preferably increase oxidation. Further, GMG-5 polypeptides of the invention are believed to modulate weight gain in mice that are fed a high fat/sucrose diet.
  • the instant invention encompasses the use of GMG-5 polypeptides in the partitioning of free fatty acid (FFA) and as an important new tool to control energy homeostasis.
  • FFA free fatty acid
  • GMG-5 polypeptides that have measurable activity in vitro and in vivo have been identified.
  • activities include, but are not limited to, modulation, preferably reduction, of the postprandial response of plasma free fatty acids, glucose, and triglycerides in mice fed a high fat/sucrose meal (Example 6), change, preferably an increase, in muscle free fatty acid oxidation in vitro and ex vivo
  • Example 10 sustained weight loss in mice on a high fat/sucrose diet.
  • Other assays for GMG-5 polypeptide activity in vitro and in vivo are also provided (Examples 2, 5, 7, 9, 11, for example), and equivalent assays can be designed by those with ordinary skill in the art.
  • GMG-5 polypeptides includes both the “full-length” polypeptide and fragments of the "full-length” GMG-5 polypeptides (although each of the above species may be particularly specified).
  • GMG-5 polypeptide further includes polypeptides of the invention and fragments thereof that have been modified to contain a recognizable signal peptide that has been added to the N-terminal end of said polypeptides.
  • intact or full-length GMG-5 polypeptides as used herein is meant the full length polypeptide sequence of any GMG-5 polypeptide, from the N-terminal methionine to the C-terminal stop codon. Examples of intact or full length GMG-5 polypeptides are found in the sequence listing.
  • metabolic-related activity refers to at least one, and preferably all, of the activities described herein for GMG-5 polypeptides. Assays for the determination of these activities are provided herein (e.g. Examples 2, 5-7, 9-11), and equivalent assays can be designed by those with ordinary skill in the art.
  • metabolic-related activity can be selected from the group consisting of lipid partitioning, lipid metabolism, and insulin-like activity, or an activity within one of these categories.
  • lipid partitioning activity is meant the ability to effect the location of dietary lipids among the major tissue groups including, adipose tissue, liver, and muscle.
  • GMG-5 polypeptides of the invention play a role in the partitioning of lipids to the muscle, liver or adipose tissue.
  • lipid metabolism activity is meant the ability to influence the metabolism of lipids.
  • GMG-5 polypeptides of the invention have the ability to affect the level of free fatty acids in the plasma as well as to modulate, preferably increase, the metabolism of lipids in the muscle through free fatty acid oxidation experiments (Examples 2, 6, 8, 9, 10) and to transiently affect the levels of triglycerides in the plasma and the muscle (Examples 6, 8, 11).
  • insulin-like activity is meant the ability of GMG-5 polypeptides to modulate the levels of glucose in the plasma.
  • GMG-5 polypeptides do not significantly impact insulin levels but do impact glucose levels similarly to the effects of insulin (Examples 7 & 8). These effects may vary in the presence of the intact (full-length) GMG-5 polypeptides or are significantly greater in the presence of the GMG-5 polypeptide fragments compared with the full-length GMG-5 polypeptides.
  • the term "significantly greater" as used herein refers to a comparison of the activity of a GMG-5 polypeptide in an metabolic-related assay compared with untreated cells in the same assay.
  • “significantly” as used herein is meant statistically significant as it is typically determined by those with ordinary skill in the art. For example, data are typically calculated as a mean ⁇ SEM, and a p-value ⁇ 0.05 is considered statistically significant.
  • Statistical analysis is typically done using either the unpaired Student's t test or the paired Student's t test, as appropriate in each study.
  • Examples of a significant change in activity as a result of the presence of a GMG-5 polypeptide of the invention compared to untreated cells include an increase or a decrease in a given parameter of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75%.
  • One or more, but not necessarily all, of the measurable parameters will change significantly in the presence of GMG-5 polypeptide as compared to untreated cells.
  • Representative "metabolic-related assays" are provided in the Examples. These assays include, but are not limited to, methods of measuring the postprandial response, methods of measuring free fatty acid oxidation, and methods of measuring weight modulation.
  • the post-prandial response is measured in non-human animals, preferably mice.
  • changes in dietary lipids are measured, preferably free fatty acids and/or triglycerides.
  • other physiologic parameters are measured including, but not limited to, levels of glucose, insulin, and leptin.
  • free fatty acid oxidation is measured in cells in vitro or ex vivo, preferably in muscle cells or tissue of non-human animals, preferably mice.
  • weight modulation is measured in human or non-human animals, preferably rodents (rats or mice), primates, canines, felines or procines. on a high fat/sucrose diet.
  • measurable parameters relating to obesity and the field of metabolic research can be selected from the group consisting of free fatty acid levels, free fatty acid oxidation, triglyceride levels, glucose levels, insulin levels, leptin levels, food intake, weight, leptin and lipoprotein binding, uptake and degradation and lipolysis stimulated receptor (LSR) expression.
  • LSR lipolysis stimulated receptor
  • preferred GMG-5 polypeptides would cause a significant change in at least one of the measurable parameters selected from the group consisting of postprandial lipemia, free fatty acid levels, triglyceride levels, glucose levels, free fatty acid oxidation, and weight.
  • preferred GMG-5 polypeptides would have a significant change in at least one of the measurable parameters selected from the group consisting of an increase in LSR activity, an increase in leptin activity and an increase in lipoprotein activity.
  • LSR activity is meant expression of LSR on the surface of the cell, or in a particular conformation, as well as its ability to bind, uptake, and degrade leptin and lipoprotein.
  • LSR activity is meant its binding, uptake and degradation by LSR, as well as its transport across a blood brain barrier, and potentially these occurrences where LSR is not necessarily the mediating factor or the only mediating factor.
  • lipoprotein activity is meant its binding, uptake and degradation by LSR, as well as these occurrences where LSR is not necessarily the mediating factor or the only mediating factor.
  • the invention is drawn, inter alia, to isolated, purified or recombinant GMG-5 polypeptides.
  • GMG-5 polypeptides of the invention are useful for reducing or increasing (using antagonists of
  • GMG-5 polypeptides body weight either as a cosmetic treatment or for treatment or prevention of metabolic-related diseases and disorders.
  • GMG-5 polypeptides are also useful wter alia in screening assays for agonists or antagonists of GMG-5 polypeptide activity; in screening assays for antagonists of N-terminal dipeptidyl peptidase cleavage of GMG-5 fragments, preferably cleavage of the N-terminal LP dipeptide of GMG-5 polypeptide fragments 16-158, 23-158 of SEQ ID NO: 2,
  • the GMG-5 polypeptides of the present invention are preferably provided in an isolated form, and may be partially or substantially purified.
  • a recombinantly produced version of any one of the GMG-5 polypeptides can be substantially purified by the one-step method described by Smith et al. ((1988) Gene 67(1):31-40) or by the methods described herein or known in the art.
  • Polypeptides of the invention also can be purified from natural or recombinant sources using antibodies directed against the polypeptides of the invention by methods known in the art of protein purification.
  • GMG-5 polypeptides of the invention involving a partial purification of or selection for the GMG-5 polypeptides are also specifically contemplated. These crude preparations are envisioned to be the result of the concentration of cells expressing GMG-5 polypeptides with perhaps a few additional purification steps, but prior to complete purification of the fragment.
  • the cells expressing GMG-5 polypeptides are present in a pellet, they are lysed, or the crude polypeptide is lyophilized, for example.
  • GMG-5 polypeptides can be any integer in length from at least 6 consecutive amino acids to the number of amino acids of the full length GMG-5 polypeptide.
  • a GMG-5 polypeptide can be any integer of consecutive amino acids from 6 to 158, for example.
  • integers include, but are not limited to: 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 11
  • Each GMG-5 polypeptide as described above can be further specified in terms of its N- terminal and C-terminal positions. For example, every combination of a N-terminal and C-terminal position that fragments of from 6 contiguous amino acids to 1 amino acids less than the full length polypeptide of SEQ ED NO: 2 could occupy, on any given intact and contiguous full length polypeptide sequence of SEQ ID NO: 2 are included in the present invention. Thus, a 6 consecutive amino acid fragment could occupy positions selected from the group consisting of 1-6, 2-7, 3-8, 4-9,
  • the positions occupied by all the other fragments of sizes between 6 amino acids and 157, 178, 177, 163, 156 or 150 amino acids in SEQ ED NOs: 4, 6, 8, 10 ,12 or 14, respectively, are included in the present invention and can also be immediately envisaged based on these two examples and therefore, are not individually listed solely for the purpose of not unnecessarily lengthening the specification.
  • the positions occupied by fragments of 6 to next to the last amino acid consecutive amino acids in SEQ ED NOs: 2, 4, 6, 8, 10, 12 or 14 are included in the present invention and can also be immediately envisaged based on these two examples and therefore are not individually listed solely for the purpose of not unnecessarily lengthening the specification.
  • the GMG-5 polypeptides of the present invention may alternatively be described by the formula "n to c" (inclusive); where “n” equals the N-terminal most amino acid position (as defined by the sequence listing) and “c” equals the C-terminal most amino acid position (as defined by the sequence listing) of the polypeptide; and further where “n” equals an integer between 1 and the number of amino acids of the full length polypeptide sequence of the present invention minus 6 ; and where "c” equals an integer between 7 and the number of amino acids of the full length polypeptide sequence; and where "n” is an integer smaller then “c” by at least 6.
  • n is any integer selected from the list consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88,
  • n and c positions are included as specific embodiments of the invention.
  • the formula "n” to “c” may be modified as '"nl - n2" to "cl - c2"', wherein “nl - n2" and “cl - c2" represent positional ranges selected from any two integers above which represent amino acid positions of the sequence listing.
  • Alternative formulas include '"nl - n2" to c and n to cl - c2 .
  • the present invention also provides for the exclusion of any individual fragment specified by N-terminal and C-terminal positions or of any fragment specified by size in amino acid residues as described above.
  • any number of fragments specified by N-terminal and C-terminal positions or by size in amino acid residues as described above may be excluded as individual species.
  • any number of fragments specified by N-terminal and C-terminal positions or by size in amino acid residues as described above may make up a polypeptide fragment in any combination and may optionally include non-GMG-5 polypeptide sequences as well.
  • GMG-5 polypeptide fragments of the invention include variants, fragments, analogs and derivatives of the GMG-5 polypeptide fragments described above, including modified GMG-5 polypeptide fragments.
  • the full-length GMG-5 polypeptide is comprised of at least three distinct regions including: 1. an N-terminal putative signal sequence (MVRMVPVLLSLLLLLGPAVP) from amino acids 1-20 of SEQ ID NOs: 6, 8, 10, 12 or 14; 2. a unique region about from amino acids 2-26 of SEQ ID NO: 2, 1 -25 of SEQ ID NO: 4,
  • the invention further includes vanants of GMG-5 polypeptides that have metabolic- related activity as descnbed above.
  • vanants include GMG-5 polypeptide sequences with one or more ammo acid deletions, insertions, inversions, repeats, and substitutions either from natural mutations or human manipulation selected according to general rules known in the art so as to have little effect on activity. Guidance concerning how to make phenotypically silent amino acid substitutions is provided below.
  • Substantial modifications in function or immunological identity of the GMG-5 polypeptides are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side- chain properties:
  • hydrophobic norleucine, met, ala, val, leu, ile
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Such substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites.
  • Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-cham beyond the beta-carbon and is less likely to alter the main chain conformation of the vanant [Cunningham and Wells, Science, 244: 1081-1085 (1989)]. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both Primad and exposed positions [Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976)]. If alanine substitution does not yield adequate amounts of vanant, an isotenc amino acid can be used.
  • Ammo acids in the GMG-5 polypeptide sequences of the invention that are essential for function can also be identified by methods known in the art, such as site-directed mutagenesis or alanme-scanmng mutagenesis (see, e g., Cunningham, et al. (1989) Science 244(4908): 1081-5). The latter procedure introduces single alanme mutations at every residue in the molecule. The resulting mutant molecules are then tested for metabolic-related activity using assays as descnbed above. Of special interest are substitutions of charged ammo acids with other charged or neutral ammo acids that may produce proteins with highly desirable improved charactenstics, such as less aggregation.
  • Aggregation may not only reduce activity but also be problematic when preparing pharmaceutical or physiologically acceptable formulations, because aggregates can be lmmunogenic (see, e g., Pmckard, et al., (1967) Clm. Exp Immunol 2:331-340; Robbms, et al., (1987) Diabetes Jul,36(7):838-41; and Cleland, et al., (1993) Cnt Rev Ther Drug Carner Syst. 10(4):307-77).
  • the fragment, denvative, analog, or homolog of the GMG-5 polypeptides of the present invention may be, for example: (I) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved ammo acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code (i.e.
  • the ammo acid residues may be a non-naturally occurring amino acid); or (n) one in which one or more of the ammo acid residues includes a substituent group; or (in) one m which the GMG-5 polypeptides are fused with another compound, such as a compound to increase the half-life of the fragment (for example, polyethylene glycol); or (IV) one in which the additional ammo acids are fused to the above form of the fragment , such as an IgG Fc fusion region peptide or leader or secretory sequence or a sequence which is employed for purification of the above form of the fragment or a pro-protein sequence.
  • Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
  • a further embodiment of the invention relates to a polypeptide which comprises the amino acid sequence of GMG-5 polypeptides having an amino acid sequence which contains at least one conservative amino acid substitution, but not more than 50 conservative amino acid substitutions, not more than 40 conservative amino acid substitutions, not more than 30 conservative amino acid substitutions, and not more than 20 conservative amino acid substitutions. Also provided are polypeptides which comprise the amino acid sequence of a GMG-5 fragment, having at least one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 conservative amino acid substitutions.
  • a further embodiment of the invention relates to a GMG-5 polypeptide fragment made resistant to dipeptidyl peptidase cleavage through N-terminal modification of said polypeptide fragment.
  • said dipeptidyl peptidase cleavage leads to removal of the N- terminal dipeptide LP by dipeptidyl peptidase from said preferred fragment.
  • said dipeptidyl peptidase is selected from but not restricted to human CD26 and human Attractin.
  • said dipeptidyl peptidase is selected from soluble human CD26 or soluble human Attractin.
  • said N-terminal modification is selected from but not restricted to glycation [Harte (2001) Regulatory Peptides 96:95-104 which disclosure is hereby inco ⁇ orated by reference in its entirety], N-methylation, alpha-methylation, desamidation [Gallwitz (2000) Regulatory Peptides 86: 103-111 which disclosure is hereby inco ⁇ orated by reference in its entirety], or alternation of the chirality of one or more N-terminal amino acids [Siegel (1999) European Journal of Clinical Investigation 29:610-614 which disclosure is hereby inco ⁇ orated by reference in its entirety].
  • the invention also encompasses a GMG-5 polypeptide fragment or a variant thereof that has been made resistant to dipeptidyl peptidase cleavage through N-terminal modification of said polypeptide fragment.
  • amino acids have chirality within the body of either L or D.
  • it is preferable to alter the chirality of one or more amino acid in order to render the GMG-5 polypeptide fragment resistant to dipeptidyl peptidase cleavage [Siegel (1999) European Journal of Clinical Investigation 29:610-614 which disclosure is hereby inco ⁇ orated by reference in its entirety].
  • one or more of the amino acids are preferably in the L configuration. In other embodiments, one or more of the amino acids are preferably in the D configuration.
  • polypeptides of the present invention also include polypeptides having an amino acid sequence at least 50% identical, at least 60% identical, or 70%, 80%, 85%, 90%, 91%, 92%, 93%,
  • a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a GMG-5 polypeptide amino acid sequence is meant that the amino acid sequence is identical to the GMG-5 polypeptide sequence except that it may include up to five amino acid alterations per each 100 amino acids of the GMG-5 polypeptide amino acid sequence.
  • the reference sequence is the GMG-5 polypeptide with a sequence corresponding to the sequences provided in SEQ ID NOs: 2, 4, 6, 8, 10, 12 or 14.
  • polypeptide having an amino acid sequence at least 95% identical to a GMG-5 polypeptide amino acid sequence up to 5% (5 of 100) of the amino acid residues in the sequence may be inserted, deleted, or substituted with another amino acid compared with the GMG- 5 polypeptide sequence. These alterations may occur at the amino or carboxy termini or anywhere between those terminal positions, interspersed either individually among residues in the sequence or in one or more contiguous groups within the sequence.
  • any particular polypeptide is a percentage identical to a GMG-5 polypeptide can be determined conventionally using known computer programs.
  • Such algorithms and programs include, but are by no means limited to, TBLASTN, BLASTP, FASTA, TFASTA, and CLUSTALW (Pearson and Lipman, (1988) Proc Natl Acad Sci USA Apr;85(8):2444- 8; Altschul et al., (1990) J. Mol. Biol. 215(3):403-410; Thompson et al., (1994) Nucleic Acids Res. 22(2):4673-4680; Higgins et al., (1996) Meth. Enzymol.
  • BLAST Basic Local Alignment Search Tool
  • BLASTP and BLAST3 compare an amino acid query sequence against a protein sequence database
  • BLASTX compares the six-frame conceptual translation products of a query nucleotide sequence (both strands) against a protein sequence database
  • TBLASTN compares a query protein sequence against a nucleotide sequence database translated in all six reading frames (both strands); and (5) TBLASTX compares the six-frame translations of a nucleotide query sequence against the six-frame translations of a nucleotide sequence database.
  • the BLAST programs identify homologous sequences by identifying similar segments, which are referred to herein as "high-scoring segment pairs," between a query amino or nucleic acid sequence and a test sequence which is preferably obtained from a protein or nucleic acid sequence database.
  • High-scoring segment pairs are preferably identified (t.e., aligned) by means of a scoring matrix, many of which are known in the art.
  • the scoring matrix used is the BLOSUM62 matrix (see, Gonnet et al., (1992) Science Jun 5;256(5062): 1443-5; Henikoff and Henikoff (1993)
  • PAM or PAM250 matrices may also be used (See, e.g., Schwartz and Dayhoff, eds, (1978) Matrices for Detecting Distance Relationships: Atlas of
  • the BLAST programs evaluate the statistical significance of all high-scoring segment pairs identified, and preferably selects those segments which satisfy a user-specified threshold of significance, such as a user-specified percent homology.
  • a user-specified threshold of significance such as a user-specified percent homology.
  • the statistical significance of a high-scoring segment pair is evaluated using the statistical significance formula of Karlin (See, e.g., Karlin and Altschul, (1990) Proc Natl Acad Sci USA Mar;87(6):2264-8).
  • the BLAST programs may be used with the default parameters or with modified parameters provided by the user. Preferably, the parameters are default parameters.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence can be determined using the FASTDB computer program based on the algorithm of Brutiag et al. (1990) Comp. App. Biosci. 6:237-245.
  • a sequence alignment the query and subject sequences are both amino acid sequences.
  • the result of said global sequence alignment is in percent identity.
  • the results, in percent identity must be manually corrected because the FASTDB program does not account for N- and C-terminal truncations of the subject sequence when calculating global percent identity.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C- terminal of the subject sequence, that are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This final percent identity score is what is used for the pu ⁇ oses of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the pu ⁇ oses of manually adjusting the percent identity score. That is, only query amino acid residues outside the farthest N- and C-terminal residues of the subject sequence.
  • a 90 amino acid residue subject sequence is aligned with a 100-residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not match/align with the first residues at the
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90-residue subject sequence is compared with a 100-residue query sequence. This time the deletions are internal so there are no residues at the N- or C-termini of the subject sequence, which are not matched/aligned with the query. In this case, the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected. No other manual corrections are made for the pu ⁇ oses of the present invention.
  • GMG-5 polypeptides are discussed, GMG-5 fragments, variants and derivatives are specifically intended to be included as a preferred subset of GMG-5 polypeptides.
  • GMG-5 polypeptides are preferably isolated from human or mammalian tissue samples or expressed from human or mammalian genes in human or mammalian cells.
  • the GMG-5 polypeptides of the invention can be made using routine expression methods known in the art.
  • the polynucleotide encoding the desired polypeptide is ligated into an expression vector suitable for any convenient host. Both eukaryotic and prokaryotic host systems are used in forming recombinant polypeptides.
  • the polypeptide is then isolated from lysed cells or from the culture medium and purified to the extent needed for its intended use. Purification is by any technique known in the art, for example, differential extraction, salt fractionation, chromatography, centrifugation, and the like. See, for example, Methods in Enzymology for a variety of methods for purifying proteins.
  • the polypeptides of the invention are isolated from milk.
  • the polypeptides can be purified as full length GMG-5 polypeptides, which can then be cleaved, if appropriate, in vitro to generate a GMG-5 fragment, or, alternatively, GMG-5 fragments themselves can be purified from the milk.
  • Any of a large number of methods can be used to purify the present polypeptides from milk, including those taught in Protein Purification Applications, A Practical Approach (New Edition), Edited by Simon Roe, AEA Technology Products and Systems, Biosciences, Harwell; Clark (1998) J Mammary Gland Biol Neoplasia 3:337-50; Wilkins and
  • milk is centrifuged, e.g. at a relatively low speed, to separate the lipid fraction, and the aqueous supernatant is then centrifuged at a higher speed to separate the casein in the milk from the remaining, "whey" fraction.
  • whey aqueous supernatant
  • biomedical proteins are found in this whey fraction, and can be isolated from this fraction using standard chromatographic or other procedures commonly used for protein purification, e.g. as described elsewhere in the present application.
  • GMG-5 polypeptides are purified using antibodies specific to GMG-5 polypeptides, e.g. using affinity chromatography.
  • methods can be used to isolate particular GMG-5 fragments, e.g. electrophoretic or other methods for isolating proteins of a particular size.
  • the GMG-5 polypeptides isolating using these methods can be naturally occurring, as GMG-5 polypeptides have been discovered to be naturally present in the milk of mammals, or can be the result of the recombinant production of the protein in the mammary glands of a non-human mammal, as described infra.
  • the GMG-5 is produced as a fusion protein with a heterologous, antigenic polypeptide sequence, which antigenic sequence can be used to purify the protein, e.g., using standard immuno-affinity methodology.
  • shorter protein fragments may be produced by chemical synthesis.
  • the proteins of the invention are extracted from cells or tissues of humans or non- human animals.
  • Methods for purifying proteins include the use of detergents or chaotropic agents to disrupt particles followed by differential extraction and separation of the polypeptides by ion exchange chromatography, affinity chromatography, sedimentation according to density, and gel electrophoresis.
  • Any GMG-5 cDNA can be used to express GMG-5 polypeptides.
  • the nucleic acid encoding the GMG-5 to be expressed is operably linked to a promoter in an expression vector using conventional cloning technology.
  • the GMG-5 cDNA insert in the expression vector may comprise the coding sequence for: the full length GMG-5 polypeptide (to be later modified); from 6 amino acids to 6 amino acids any integer less than the full-length GMG-5 polypeptide; a GMG-5 fragment; or variants and % similar polypeptides.
  • the expression vector is any of the mammalian, yeast, insect or bacterial expression systems known in the art, some of which are described herein.
  • Commercially available vectors and expression systems are available from a variety of suppliers including Genetics Institute (Cambridge, MA), Stratagene (La Jolla, California), Promega (Madison, Wisconsin), and Invitrogen (San Diego,
  • codon context and codon pairing of the sequence can be optimized for the particular expression organism into which the expression vector is introduced, as explained by Hatfield, et al., U.S. Patent No. 5,082,767, the disclosures of which are inco ⁇ orated by reference herein in their entirety.
  • nucleic acid encoding any one of the GMG-5 polypeptides lacks a methionine to serve as the initiation site, an initiating methionine can be introduced next to the first codon of the nucleic acid using conventional techniques.
  • an initiating methionine can be introduced next to the first codon of the nucleic acid using conventional techniques.
  • insert from the GMG-5 polypeptide cDNA lacks a poly
  • this sequence can be added to the construct by, for example, splicing out the Poly A signal from pSG5 (Stratagene) using Bgll and Sail restriction endonuclease enzymes and inco ⁇ orating it into the mammalian expression vector pXTl (Stratagene).
  • pXTl contains the LTRs and a portion of the gag gene from Moloney Murine Leukemia Virus. The position of the LTRs in the construct allow efficient stable transfection.
  • the vector includes the He ⁇ es Simplex Thymidine Kinase promoter and the selectable neomycin gene.
  • the nucleic acid encoding GMG-5 can be obtained by PCR from a vector containing the GMG-5 nucleotide sequence using oligonucleotide primers complementary to the desired GMG-5 cDNA and containing restriction endonuclease sequences for Pst I inco ⁇ orated into the 5' primer and BglEI at the 5' end of the corresponding cDNA 3' primer, taking care to ensure that the sequence encoding the GMG-5 is positioned properly with respect to the poly A signal.
  • the purified polynucleotide obtained from the resulting PCR reaction is digested with Pstl, blunt ended with an exonuclease, digested with Bgl Ef, purified and ligated to pXTl, now containing a poly A signal and digested with Bgi ⁇ .
  • Transfection of a GMG-5 expressing vector into mouse NTH 3T3 cells is one embodiment of introducing polynucleotides into host cells.
  • Introduction of a polynucleotide encoding a polypeptide into a host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods.
  • polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • a polypeptide of this invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention can also be recovered from: products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells.
  • a prokaryotic or eukaryotic host including, for example, bacterial, yeast, higher plant, insect, and mammalian cells.
  • the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
  • the polypeptides of the invention are non-glycosylated.
  • polypeptides of the invention may also include an initial modified methionine residue, m some cases as a result of host-mediated processes.
  • an initial modified methionine residue m some cases as a result of host-mediated processes.
  • the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation m all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the ammo acid to which the N-termmal methionine is covalently linked.
  • the invention also encompasses pnmary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian ongm, that have been engineered to delete or replace endogenous genetic material (e g., coding sequence), and/or to include genetic material (e.g., heterologous polynucleotide sequences) that is operably associated with the polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous polynucleotides.
  • endogenous genetic material e.g., coding sequence
  • genetic material e.g., heterologous polynucleotide sequences
  • heterologous control regions e g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • heterologous control regions e g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • polypeptides of the invention can be chemically synthesized using techniques known m the art (See, e g., Creighton, 1983 Proteins. New York, New York: W.H. Freeman and Company; and Hunkapiller et al., (1984) Nature Jul 12-18;310(5973):105-11).
  • a relative short fragment of the invention can be synthesized by use of a peptide synthesizer.
  • nonclassical ammo acids or chemical amino acid analogs can be introduced as a substitution or addition into the fragment sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-d ⁇ ammobutync acid, a-amino lsobuty ⁇ c acid, 4-am ⁇ noburync acid, Abu, 2-am ⁇ no butyric acid, g-Abu, e-Ahx, 6-am ⁇ no hexanoic acid, Aib, 2-am ⁇ no lsobuty ⁇ c acid, 3-am ⁇ no propiomc acid, ornithine, norleucine, norvalme, hydroxyprolme, sarcosine, citrulline, homocitrulhne, cysteic acid, t-butylglycme, t-butylalanme, phenylglycine, cyclohexylalanine, b-alamne, fluoroamino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl
  • the invention encompasses polypeptides which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed by the invention include, for example, N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression.
  • the polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the polypeptide.
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the polypeptides may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog).
  • polyethylene glycol molecules should be attached to the polypeptide with consideration of effects on functional or antigenic domains of the polypeptide.
  • attachment methods available to those skilled in the art, e.g., EP 0 401 384, herein inco ⁇ orated by reference (coupling PEG to G-CSF), see also Malik et al. (1992) Exp
  • polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, a free amino or carboxyl group.
  • Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues, glutamic acid residues and the C-terminal amino acid residue.
  • Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules.
  • Preferred for therapeutic pu ⁇ oses is attachment at an amino group, such as attachment at the N-terminus or lysine group.
  • polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
  • the method of obtaining the N-terminally pegylated preparation i.e., separating this moiety from other monopegylated moieties if necessary
  • Selective proteins chemically modified at the N-terminus may be accomplished by reductive alkylation, which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • the invention features a method of reducing body mass comprising providing or administering to individuals in need of reducing body mass said pharmaceutical or physiologically acceptable composition described in the fifth aspect in combination with provision or administration of an antagonist of dipeptidyl peptidase cleavage of GMG-5 polypeptide fragment of the first aspect.
  • Preferred said antagonist is a peptidyl derivative of a diester of alpha- aminoalkylphosphonic acid (US Patent Number 5,543,396 which disclosure is hereby inco ⁇ orated by reference in its entirety). More preferred said peptidyl derivative is selected from Ala-Pro P (OZ) 2 , AcOH.Ala-Pip p (Oph) 2 , HCl.Ala-Pro p (Oph-4Cl) 2 , HCl.Ala-Pip p (Oph-4Cl) 2 , or 2HCl.Lys-Pip p (Oph- 4C1) 2 , where Z represents an aryl group, a substituted aryl group or a highly flourinated alkyl group, Pro p represents a proline phosphonate derivative, and Pip p represents piperidyl phosphonate (US Patent Number 5,543,396 which disclosure is hereby inco ⁇ orated by reference in its entirety).
  • Xaa is an amino acid
  • Z is a protecting group
  • Y' is one of various types of ring structures
  • Z may or may not be present and represents a protecting group, such as benzyloxycarbonyl
  • Xaa represents alanine, methionine, arginine, phenylalanine, aspartic acid, proline, asparagine, serine, cysteine, threonine, glycine, tyrosine, glutamic acid, tryptophan, glutamine, valine, isoleucine, lysine, leucine, L-thioproline, L-homoproline, L- l,2,3,4,tetrahydroisoquinoline-3-carboxylic acid (Tic), L-2,3-dihydroindol-2-carboxylic acid, L- naphthylglycine, L-phenylglycine, L-4-phenylproline, O-benzyl tyrosine, omega-Z lysine, or omega- acetyl lysine; and Y'
  • N-(substituted glycyl)-2-cyanopyrrolidine (US Patent Number 6,166,063). More preferred said N-(substituted glycyl)-2-cyanopyrrolidine is selected from pyrrolidine, l-[[(3,5-dimethyl-l-adamantyl)amino]-acetyl]-2-cyano-, (S)-; pyrrolidine, l-[[(3-ethyl- l-adamantyl)amino]-acetyl]-2-cyano-, (S)-; pyrrolidine, l-[[(3-methoxy-l-adamantyl)amino]- acetyl]-2-cyano-, (S)-; pyrrolidine, l-[[[3-[[[(t-butylamino)carbonyl]oxy]-l-adamantyl]amino]- acetyrolidine,
  • tetrahydroisoquinoline 3-carboxamide derivative of formula ##STR1## (US Patent Number 6,172,081). More preferred is said derivative and pharmaceutically acceptable salts thereof wherein X is CH 2 , S, O, or C(CH 3 ) 2 ; Rj and R 2 are independently hydrogen, hydroxy, alkyl, alkoxy, aralkoxy, or halogen (US Patent Number 6,172,081).
  • valine-pyrrolidide (Deacon (2001) Diabetes 50: 1588- 1597 which disclosure is hereby inco ⁇ orated by reference in its entirety].
  • the polypeptides of the invention may be in monomers or multimers (i.e., dimers, trimers, tetramers and higher multimers). Accordingly, the present invention relates to monomers and multimers of the polypeptides of the invention, their preparation, and compositions (preferably, pharmaceutical or physiologically acceptable compositions , ) containing them.
  • the polypeptides of the invention are monomers, dimers, trimers or tetramers.
  • the multimers of the invention are at least dimers, at least trimers, or at least tetramers. Multimers encompassed by the invention may be homomers or heteromers.
  • homomer refers to a multimer containing only polypeptides corresponding to the GMG-5 polypeptides of the invention (including polypeptide fragments, variants, splice variants, and fusion proteins corresponding to these polypeptide fragments as described herein). These homomers may contain polypeptide fragments having identical or different amino acid sequences. In a specific embodiment, a homomer of the invention is a multimer containing only polypeptide fragments having an identical amino acid sequence. In another specific embodiment, a homomer of the invention is a multimer containing polypeptide fragments having different amino acid sequences.
  • the multimer of the invention is a homodimer (e.g., containing polypeptides having identical or different amino acid sequences) or a homotrimer (e.g., containing polypeptides having identical and/or different amino acid sequences).
  • the homomeric multimer of the invention is at least a homodimer, at least a homotrimer, or at least a homotetramer.
  • heteromer refers to a multimer containing one or more heterologous polypeptides (i.e., corresponding to different proteins or polypeptides thereof) in addition to the polypeptides of the invention.
  • the multimer of the invention is a heterodimer, a heterotrimer, or a heterotetramer.
  • the heteromeric multimer of the invention is at least a heterodimer, at least a heterotrimer, or at least a heterotetramer.
  • Multimers of the invention may be the result of hydrophobic, hydrophilic, ionic and/or covalent associations and/or may be indirectly linked, by for example, liposome formation.
  • multimers of the invention such as, for example, homodimers or homotrimers, are formed when polypeptides of the invention contact one another in solution.
  • heteromultimers of the invention such as, for example, heterotrimers or heterotetramers, are formed when polypeptides of the invention contact antibodies to the polypeptides of the invention (including antibodies to the heterologous polypeptide sequence in a fusion protein of the invention) in solution.
  • multimers of the invention are formed by covalent associations with and/or between the polypeptides of the invention.
  • covalent associations may involve one or more amino acid residues contained in the polypeptide sequence (e.g., that recited in the sequence listing, or contained in the polypeptide encoded by a deposited clone).
  • the covalent associations are cross-linking between cysteine residues located within the polypeptide sequences, which interact in the native (i.e., naturally occurring) polypeptide.
  • the covalent associations are the consequence of chemical or recombinant manipulation.
  • such covalent associations may involve one or more amino acid residues contained in the heterologous polypeptide sequence in a fusion protein of the invention.
  • covalent associations are between the heterologous sequence contained in a fusion protein of the invention (see, e.g., US Patent Number 5,478,925).
  • the covalent associations are between the heterologous sequence contained in an Fc fusion protein of the invention (as described herein).
  • covalent associations of fusion proteins of the invention are between heterologous polypeptide sequence from another protein that is capable of forming covalently associated multimers, such as for example, oseteoprotegerin (see, e.g.,
  • polypeptide linkers are joined through peptide linkers. Examples include those peptide linkers described in U.S. Pat. No.
  • Proteins comprising multiple polypeptides of the invention separated by peptide linkers may be produced using conventional recombinant DNA technology.
  • Another method for preparing multimer polypeptides of the invention involves use of polypeptides of the invention fused to a leucine zipper or isoleucine zipper polypeptide sequence.
  • Leucine zipper and isoleucine zipper domains are polypeptides that promote multimerization of the proteins in which they are found. Leucine zippers were originally identified in several DNA-binding proteins, and have since been found in a variety of different proteins (Landschulz et al., (1988) Genes Dev. Jul;2(7):786-800).
  • leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize.
  • leucine zipper domains suitable for producing soluble multimeric proteins of the invention are those described in PCT application WO 94/10308, hereby inco ⁇ orated by reference.
  • Recombinant fusion proteins comprising a polypeptide of the invention fused to a polypeptide sequence that dimerizes or trimerizes in solution are expressed in suitable host cells, and the resulting soluble multimeric fusion protein is recovered from the culture supernatant using techniques known in the art.
  • Trimeric polypeptides of the invention may offer the advantage of enhanced biological activity.
  • Preferred leucine zipper moieties and isoleucine moieties are those that preferentially form trimers.
  • One example is a leucine zipper derived from lung surfactant protein D (SPD), as described in Hoppe et al. FEBS Letters (1994) May 16;344(2-3):191-5. and in U.S. patent application Ser. No. 08/446,922, hereby inco ⁇ orated by reference.
  • Other peptides derived from naturally occurring trimeric proteins may be employed in preparing trimeric polypeptides of the invention.
  • proteins of the invention are associated by interactions between Flag® & polypeptide sequence contained in fusion proteins of the invention containing Flag® polypeptide sequence.
  • proteins of the invention are associated by interactions between heterologous polypeptide sequence contained in Flag® fusion proteins of the invention and anti Flag® antibody.
  • the multimers of the invention may be generated using chemical techniques known in the art.
  • polypeptides desired to be contained in the multimers of the invention may be chemically cross-linked using linker molecules and linker molecule length optimization techniques known in the art (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • multimers of the invention may be generated using techniques known in the art to form one or more inter-molecule cross-links between the cysteine residues located within the sequence of the polypeptides desired to be contained in the multimer (see, e.g., US Patent
  • polypeptides of the invention may be routinely modified by the addition of cysteine or biotin to the C-terminus or
  • N-terminus of the polypeptide and techniques known in the art may be applied to generate multimers containing one or more of these modified polypeptides (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety). Additionally, at least 30 techniques known in the art may be applied to generate liposomes containing the polypeptide components desired to be contained in the multimer of the invention (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety). Alternatively, multimers of the invention may be generated using genetic engineering techniques known in the art.
  • polypeptides contained in multimers of the invention are produced recombinantly using fusion protein technology described herein or otherwise known in the art (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • polynucleotides coding for a homodimer of the invention are generated by ligating a polynucleotide sequence encoding a polypeptide of the invention to a sequence encoding a linker polypeptide and then further to a synthetic polynucleotide encoding the translated product of the polypeptide in the reverse orientation from the original C-terminus to the N-terminus (lacking the leader sequence) (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • recombinant techniques described herein or otherwise known in the art are applied to generate recombinant polypeptides of the invention which contain a transmembrane domain (or hyrophobic or signal peptide) and which can be inco ⁇ orated by membrane reconstitution techniques into liposomes (See, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • Preferred polynucleotides are those that encode GMG-5 polypeptides of the invention.
  • the recombinant polynucleotides encoding GMG-5 polypeptides can be used in a variety of ways, including, but not limited to, expressing the polypeptides in recombinant cells for use in screening assays for antagonists and agonists of its activity as well as to facilitate its purification for use in a variety of ways including, but not limited to screening assays for agonists and antagonists of its activity, diagnostic screens, and raising antibodies, as well as treatment and/or prevention of metabolic-related diseases and disorders and/or to reduce body mass.
  • the invention relates to the polynucleotides encoding GMG-5 polypeptides and variant polypeptides thereof as described herein. These polynucleotides may be purified, isolated, and/or recombinant. In all cases, the desired GMG-5 polynucleotides of the invention are those that encode GMG-5 polypeptides of the invention having metabolic-related activity as described and discussed herein. Fragments
  • a polynucleotide fragment is a polynucleotide having a sequence that entirely is the same as part, but not all, of the full length GMG-5 polypeptide or a specified GMG-5 polypeptide nucleotide sequence. Such fragments may be "free-standing", i.e. not part of or fused to other polynucleotides, or they may be comprised within another non-GMG-5 (heterologous) polynucleotide of which they form a part or region. However, several GMG-5 polynucleotide fragments may be comprised within a single polynucleotide.
  • the GMG-5 polynucleotides of the invention comprise from 18 consecutive bases to 18 consecutive bases less than the full length polynucleotide sequences encoding the intact GMG-5 polypeptides, for example the full length GMG-5 polypeptide polynucleotide sequences in SEQ ED NOs: 1, 3, 5, 7, 9, 11 or 13.
  • the polynucleotide comprises at least 18, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 305, 310, 315, 320, 325, 330, 335, 340, 345, 350, 355, 360, 365, 370, 375, 380, 385, 390, 395, 400, 405, 410, 415, 420, 425, 430, 435, 440, 445, 450, 455, 460, 465, 470, 475, 480
  • nucleic acids comprise at least 18 nucleotides, wherein "at least 18" is defined as any integer between 18 and the integer representing 18 nucleotides less than the 3' most nucleotide position of the intact GMG-5 polypeptides cDNA as set forth in SEQ ID NOs: 1, 3, 5, 7, 9, 11 or 13 or elsewhere herein.
  • nucleic acid fragments at least 18 nucleotides in length, as described above, that are further specified in terms of their 5' and 3' position.
  • the 5' and 3' positions are represented by the position numbers set forth in the sequence listing below.
  • position 1 is defined as the 5' most nucleotide of the ORF, i.e., the nucleotide "A" of the start codon (ATG) with the remaining nucleotides numbered consecutively.
  • every combination of a 5' and 3' nucleotide position that a polynucleotide fragment, at least 18 contiguous nucleotides in length, could occupy on an intact GMG-5 polypeptide encoding a polynucleotide of the present invention is included in the invention as an individual species.
  • the polynucleotide fragments specified by 5' and 3' positions can be immediately envisaged and are therefore not individually listed solely for the pu ⁇ ose of not unnecessarily lengthening the specification.
  • polynucleotide fragments of the present invention may alternatively be described by the formula "x to y"; where "x" equals the 5 ' most nucleotide position and “y” equals the 3' most nucleotide position of the polynucleotide; and further where "x” equals an integer between 1 and the number of nucleotides of the polynucleotide sequence of the present invention minus 18, and where "y” equals an integer between 19 and the number of nucleotides of the polynucleotide sequence of the present invention minus 18 nucleotides; and where "x" is an integer less than "y" by at least 18.
  • the present invention also provides for the exclusion of any species of polynucleotide fragments of the present invention specified by 5' and 3' positions or polynucleotides specified by size in nucleotides as described above. Any number of fragments specified by 5' and 3' positions or by size in nucleotides, as described above, may be excluded.
  • the GMG-5 polynucleotide fragments of the invention comprise from 18 consecutive bases to the full length polynucleotide sequence encoding the GMG-5 fragments described in Section II of the Preferred Embodiments of the Invention.
  • the polynucleotide comprises at least 18, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, 280, 285, 290, 295, 300, 305, 310, 315, 320, 325, 330, 335, 340, 345, 350, 355, 360, 365, 370, 375,
  • nucleic acids comprise at least 18 nucleotides, wherein "at least 18" is defined as any integer between 18 and the integer corresponding to the 3 ' most nucleotide position of a GMG-5 fragment cDNA herein.
  • nucleic acid fragments at least 18 nucleotides in length, as described above, that are further specified in terms of their 5' and 3' position.
  • the 5' and 3' positions are represented by the position numbers set forth in the sequence listing below.
  • position 1 is defined as the 5' most nucleotide of the open reading frame (ORF), i.e., the nucleotide "A" of the start codon (ATG) with the remaining nucleotides numbered consecutively.
  • polynucleotide fragments of the present invention may alternatively be described by the formula "x to y"; where "x" equals the 5' most nucleotide position and “y” equals the 3' most nucleotide position of the polynucleotide; and further where "x” equals an integer between 1 and the number of nucleotides of the GMG-5 polynucleotide sequences of the present invention minus 18, and where "y” equals an integer between 9 and the number of nucleotides of the GMG-5 polynucleotide sequences of the present invention; and where "x” is an integer smaller than "y” by at least 18. .
  • the present invention also provides for the exclusion of any species of polynucleotide fragments of the present invention specified by 5' and 3' positions or polynucleotides specified by size in nucleotides as described above. Any number of fragments specified by 5' and 3' positions or by size in nucleotides, as described above, may be excluded.
  • variants of GMG-5 polynucleotides encoding GMG-5 polypeptides are envisioned.
  • Variants of polynucleotides are polynucleotides whose sequence differs from a reference polynucleotide.
  • a variant of a polynucleotide may be a naturally occurring variant such as a naturally occurring allelic variant, or it may be a variant that is not known to occur naturally.
  • Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms. Generally, differences are limited so that the nucleotide sequences of the reference and the variant are closely similar overall and, in many regions, identical.
  • polynucleotide variants that comprise a sequence substantially different from those described above but that, due to the degeneracy of the genetic code, still encode GMG-5 polypeptides of the present invention are also specifically envisioned. It would also be routine for one skilled in the art to generate the degenerate variants described above, for instance, to optimize codon expression for a particular host (e.g., change codons in the human mRNA to those preferred by other mammalian or bacterial host cells). As stated above, variant polynucleotides may occur naturally, such as a natural allelic variant, or by recombinant methods.
  • an “allelic variant” is intended one of several alternate forms of a gene occupying a given locus on a chromosome of an organism (See, e.g., B. Lewin, (1990) Genes TV, Oxford University Press, New York).
  • Non-naturally occurring variants may be produced using art-known mutagenesis techniques.
  • Such nucleic acid variants include those produced by nucleotide substitutions, deletions, or additions. The substitutions, deletions, or additions may involve one or more nucleotides. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of GMG-5 polypeptides of the invention. Also preferred in this regard are conservative substitutions.
  • Nucleotide changes present in a variant polynucleotide are preferably silent, which means that they do not alter the amino acids encoded by the polynucleotide. However, nucleotide changes may also result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence.
  • preferred GMG-5 polypeptides include those that retain one or more metabolic-related activity as described in Section I of the Preferred Embodiments of the Invention.
  • the activity measured using the polypeptide encoded by the variant GMG-5 polynucleotide in assays is at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%, and not more than 101%, 102%, 103%, 104%, 105%, 110%, 115%, 120%) or 125% of the activity measured using a GMG-5 polypeptide described in the Examples Section herein.
  • the activity being "increased” is meant that the activity measured using the polypeptide encoded by the variant GMG-5 polynucleotide in assays is at least 125%, 130%, 135%, 140%, 145%, 150%, 155%., 160%, 170%, 180%, 190%, 200%, 225%, 250%, 275%, 300%, 325%, 350%, 375%, 400%o, 450%), or 500% of the activity measured using a GMG-5 polypeptide described in the Examples Section herein.
  • the activity being “decreased” is meant that the activity measured using the polypeptide encoded by the variant GMG-5 polynucleotide in assays is decreased by at least 25%, 30%, 35%, 40%, 45%, 50%, 75%, 80%, 90% or 95% of the activity measured using a GMG-5 polypeptidedescribed in the Examples Section herein
  • the present invention is further directed to nucleic acid molecules having sequences at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98% or 99% identical to the polynucleotide sequences of SEQ ID NOs: 1, 3, 5, 7, 9, 11 or 13 or fragments thereof that encode a polypeptide having metabolic-related activity as described in Section I of the Preferred Embodiments of the Invention.
  • nucleic acid molecules at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequences shown in SEQ ED NOs: 1, 3, 5, 7, 9, 11 or 13 or fragments thereof will encode a polypeptide having biological activity.
  • degenerate variants of these nucleotide sequences all encode the same polypeptide, this will be clear to the skilled artisan even without performing the above described comparison assay.
  • nucleic acid molecules that are not degenerate variants, a reasonable number will also encode a polypeptide having biological activity. This is because the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly affect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid), as further described previously in Section I of the Preferred Embodiments of the Invention.
  • nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the GMG-5 polypeptide.
  • up to 5% of the nucleotides in the reference sequence may be deleted, inserted, or substituted with another nucleotide.
  • the query sequence may be an entire sequence or any fragment specified as described herein.
  • the methods of determining and defining whether any particular nucleic acid molecule or polypeptide is at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be done by using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment can be determined using the FASTDB computer program based on the algorithm of Brutiag et al., ((1990) Comput Appl Biosci. Jul;6(3):237-45). In a sequence alignment the query and subject sequences are both DNA sequences.
  • RNA sequence can be compared by first converting U's to T's. The result of said global sequence alignment is in percent identity.
  • the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This corrected score is what is used for the pu ⁇ oses of the present invention. Only nucleotides outside the 5' and 3' nucleotides of the subject sequence, as displayed by the FASTDB alignment, which are not matched aligned with the query sequence, are calculated for the pu ⁇ oses of manually adjusting the percent identity score.
  • a 90-nucleotide subject sequence is aligned to a 100-nucleotide query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 nucleotides at
  • the 10 unpaired nucleotides represent 10% of the sequence (number of nucleotides at the 5' and 3' ends not matched/total number of nucleotides in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 nucleotides were perfectly matched the final percent identity would be 90%.
  • a 90 nucleotide subject sequence is compared with a 100 nucleotide query sequence. This time the deletions are internal deletions so that there are no nucleotides on the 5 ' or 3' of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected.
  • nucleotides 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are made for the pu ⁇ oses of the present invention.
  • polynucleotides encoding the polypeptides of the present invention that are fused in frame to the coding sequences for additional heterologous amino acid sequences.
  • nucleic acids encoding polypeptides of the present invention together with additional, non-coding sequences, including for example, but not limited to non-coding 5' and 3' sequences, vector sequence, sequences used for purification, probing, or priming.
  • heterologous sequences include transcribed, non- translated sequences that may play a role in transcription, and mRNA processing, for example, ribosome binding and stability of mRNA.
  • the heterologous sequences may alternatively comprise additional coding sequences that provide additional functionalities.
  • a nucleotide sequence encoding a polypeptide may be fused to a tag sequence, such as a sequence encoding a peptide that facilitates purification of the fused polypeptide.
  • the tag amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein (See, Gentz et al., (1989) Proc Natl Acad Sci USA Feb;86(3):821-4).
  • the "HA” tag is another peptide useful for purification which corresponds to an epitope derived from the influenza hemagglutinin protein (See, Wilson et al., (1984) Cell 37(3):767-78). As discussed above, other such fusion proteins include GMG-5 cDNA fused to Fc at the N- or C-terminus. El. Recombinant Vectors of the Invention
  • vector is used herein to designate either a circular or a linear DNA or RNA molecule, that is either double-stranded or single-stranded, and that comprises at least one polynucleotide of interest that is sought to be transferred in a cell host or in a unicellular or multicellular host organism.
  • the present invention relates to recombinant vectors comprising any one of the polynucleotides described herein.
  • the present invention encompasses a family of recombinant vectors that comprise polynucleotides encoding GMG-5 polypeptides of the invention.
  • a recombinant vector of the invention is used to amplify the inserted polynucleotide in a suitable cell host, this polynucleotide being amplified every time that the recombinant vector replicates.
  • the inserted polynucleotide can be one that encodes GMG-5 polypeptides of the invention.
  • a second preferred embodiment of the recombinant vectors according to the invention consists of expression vectors comprising polynucleotides encoding GMG-5 polypeptides of the invention.
  • expression vectors are employed to express a GMG-5 polypeptide of the invention, preferably a modified GMG-5 described in the present invention, which can be then purified and, for example, be used as a treatment for metabolic-related diseases, or simply to reduce body mass of individuals.
  • Expression requires that appropriate signals are provided in the vectors, said signals including various regulatory elements, such as enhancers/promoters from both viral and mammalian sources, that drive expression of the genes of interest in host cells.
  • signals including various regulatory elements, such as enhancers/promoters from both viral and mammalian sources, that drive expression of the genes of interest in host cells.
  • Dominant drug selection markers for establishing permanent, stable, cell clones expressing the products are generally included in the expression vectors of the invention, as they are elements that link expression of the drug selection markers to expression of the polypeptide.
  • the present invention relates to expression vectors which include nucleic acids encoding a GMG-5 polypeptide of the invention, or a modified GMG-5 as described herein, or variants or fragments thereof, under the control of a regulatory sequence selected among GMG-5 polypeptides, or alternatively under the control of an exogenous regulatory sequence.
  • preferred expression vectors of the invention are selected from the group consisting of : (a) a GMG-5 regulatory sequence and driving the expression of a coding polynucleotide operably linked thereto; and (b) a GMG-5 coding sequence of the invention, operably linked to regulatory sequences allowing its expression in a suitable cell host and/or host organism.
  • a recombinant vector according to the invention comprises, but is not limited to, a YAC (Yeast Artificial Chromosome), a BAC (Bacterial Artificial Chromosome), a phage, a phagemid, a cosmid, a plasmid, or even a linear DNA molecule which may consist of a chromosomal, non- chromosomal, semi-synthetic or synthetic DNA.
  • a recombinant vector can comprise a transcriptional unit comprising an assembly of :
  • Enhancers are cis-acting elements of DNA, usually from about 10 to 300 bp in length that act on the promoter to increase the transcription;
  • a structural or coding sequence which is transcribed into mRNA and eventually translated into a polypeptide, said structural or coding sequence being operably linked to the regulatory elements described in (1);
  • Structural units intended for use in yeast or eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell.
  • a recombinant protein when expressed without a leader or transport sequence, it may include a N-terminal residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final product.
  • recombinant expression vectors will include origins of replication, selectable markers permitting transformation of the host cell, and a promoter derived from a highly expressed gene to direct transcription of a downstream structural sequence.
  • the heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably a leader sequence capable of directing secretion of the translated protein into the periplasmic space or the extracellular medium.
  • preferred vectors will comprise an origin of replication in the desired host, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences, and 5 '-flanking non-transcribed sequences.
  • DNA sequences derived from the SV40 viral genome, for example SV40 origin, early promoter, enhancer, splice and polyadenylation sites may be used to provide the required non-transcribed genetic elements.
  • Promoters used in the expression vectors of the present invention are chosen taking into account the cell host in which the heterologous gene is expressed.
  • the particular promoter employed to control the expression of a nucleic acid sequence of interest is not believed to be important, so long as it is capable of directing the expression of the nucleic acid in the targeted cell.
  • a human cell it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell, such as, for example, a human or a viral promoter.
  • a suitable promoter may be heterologous with respect to the nucleic acid for which it controls the expression or alternatively can be endogenous to the native polynucleotide containing the coding sequence to be expressed. Additionally, the promoter is generally heterologous with respect to the recombinant vector sequences within which the construct promoter/coding sequence has been inserted.
  • Promoter regions can be selected from any desired gene using, for example, CAT (chloramphenicol transferase) vectors and more preferably pKR232-8 and pCM7 vectors.
  • CAT chloramphenicol transferase
  • Preferred bacterial promoters are the Lad, LacZ, the T3 or T7 bacteriophage RNA polymerase promoters, the gpt, lambda PR, PL and tip promoters (EP 0036776), the polyhedrin promoter, or the plO protein promoter from baculovirus (Kit Novagen) (Smith et al., (1983) Mol Cell Biol Dec;3(12):2156-65; O'Reilly et al., 1992), the lambda PR promoter or also the tie promoter.
  • Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late
  • promoters specific for a particular cell type may be chosen, such as those facilitating expression in adipose tissue, muscle tissue, or liver. Selection of a convenient vector and promoter is well within the level of ordinary skill in the art. The choice of a promoter is well within the ability of a person skilled in the field of genetic engineering. For example, one may refer to Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY, Vol. 1, 2, 3 (1989), or also to the procedures described by Fuller et al. (1996) Immunology in Current Protocols in Molecular Biology.
  • a cDNA insert where a cDNA insert is employed, one will typically desire to include a polyadenylation signal to effect proper polyadenylation of the gene transcript.
  • the nature of the polyadenylation signal is not believed to be crucial to the successful practice of the invention, and any such sequence may be employed such as human growth hormone and SV40 polyadenylation signals.
  • a terminator Also contemplated as an element of the expression cassette is a terminator. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences.
  • Vectors containing the appropriate DNA sequence as described above can be utilized to transform an appropriate host to allow the expression of the desired polypeptide or polynucleotide.
  • the selectable marker genes for selection of transformed host cells are preferably dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, TRP1 for S. cerevisiae or tetracycline, rifampicin or ampicillin resistance in E. coli, or levan saccharase for mycobacteria, this latter marker being a negative selection marker.
  • Bacterial vectors As a representative but non-limiting example, useful expression vectors for bacterial use can comprise a selectable marker and a bacterial origin of replication derived from commercially available plasmids comprising genetic elements of pBR322 (ATCC 37017). Such commercial vectors include, for example, pKR223-3 (Pharmacia, Uppsala, Sweden), and pGEMl (Promega Biotec, Madison, WI, USA).
  • bacterial vectors such as the following bacterial vectors : pQE70, pQE60, pQE-9 (Qiagen), pbs, pDIO, phagescript, psiX174, pbluescript SR, pbsks, pNH8A, pNH16A, pNH18A, pNH46A (Stratagene); ptrc99a, pRR223-3, pKK233-3, pDR540, pRIT5 (Pharmacia); pWLNEO, pSV2CAT, pOG44, pXTl, pSG (Stratagene); pSVK3, pBPV, pMSG, pSVL (Pharmacia); pQE-30 (QIAexpress).
  • Baculovirus vectors A suitable vector for the expression of polypeptides of the invention is a baculovirus vector that can be propagated in insect cells and in insect cell lines.
  • a specific suitable host vector system is the pVL1392/1393 baculovirus transfer vector (Pharmingen) that is used to transfect the SF9 cell line (ATCC N°CRL 1711) which is derived from Spodoptera frugiperda.
  • Suitable vectors for the expression of an Apml globular head polypeptide in a baculovirus expression system include those described by Chai et al. (1993; Biotechnol Appl Biochem. Dec;18 ( Pt 3):259-73); Vlasak et al. (1983; Eur J Biochem Sep l;135(l):123-6); and Lenhard et al. (1996; Gene Mar 9; 169(2): 187-90).
  • the vector is derived from an adenovirus.
  • adenovirus vectors according to the invention are those described by Feldman and Steg (1996; Semin Interv Cardiol Sep;l(3):203-8) or Ohno et al. (1994; Science Aug 5;265(5173):781-4).
  • Another preferred recombinant adenovirus according to this specific embodiment of the present invention is the human adenovirus type 2 or 5 (Ad 2 or Ad 5) or an adenovirus of animal origin (French patent application No. FR-93.05954).
  • Retrovirus vectors and adeno-associated virus vectors are generally understood to be the recombinant gene delivery systems of choice for the transfer of exogenous polynucleotides in vivo, particularly to mammals, including humans. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • retroviruses for the preparation or construction of retroviral in vitro or in vivo gene delivery vehicles of the present invention include retroviruses selected from the group consisting of Mink-Cell Focus Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma virus.
  • retroviruses selected from the group consisting of Mink-Cell Focus Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma virus.
  • Particularly preferred Murine Leukemia Viruses include the 4070A and the 1504A viruses, Abelson (ATCC No VR-999), Friend (ATCC No VR-245), Gross (ATCC No
  • Rous Sarcoma Viruses include Bryan high titer (ATCC Nos VR-334, VR-657, VR-726, VR-659 and VR-728).
  • Other preferred retroviral vectors are those described in Roth et al. (1996), PCT Application No WO 93/25234, PCT Application No WO 94/ 06920, Roux et al., ((1989) Proc Natl Acad Sci U S A Dec;86(23):9079-83), Julan et al., (1992) J. Gen. Virol. 3:3251-3255 and Neda et al., ((1991) J Biol Chem Aug 5;266(22): 14143-6).
  • AAV adeno-associated virus
  • the adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a he ⁇ es virus, as a helper virus for efficient replication and a productive life cycle (Muzyczka et al., (1992) Curr Top Microbiol Immunol;158:97-129).
  • these constructs In order to effect expression of the polynucleotides of the invention, these constructs must be delivered into a cell. This delivery may be accomplished in vitro, as in laboratory procedures for transforming cell lines, or in vivo or ex vivo, as in the treatment of certain disease states.
  • One mechanism is viral infection where the expression construct is encapsulated in an infectious viral particle.
  • non-viral methods for the transfer of polynucleotides into cultured mammalian cells include, without being limited to, calcium phosphate precipitation (Graham et al, (1973) Virology Aug;54(2):536-9; Chen et al, (1987) Mol Cell Biol Aug;7(8):2745-52), DEAE-dextran (Gopal, (1985) Mol Cell Biol May;5(5):l 188-90), electroporation (Tur-Kaspa et al, (1986) Mol Cell Biol Feb;6(2):716-8; Potter et al, (1984) Proc Natl Acad Sci USA Nov;81(22):7161-5.), direct microinjection (Harland et al, (1985) J Cell Biol Sep; 101(3): 1094-9), DNA-loaded liposomes (Nicolau et al, (1982) Biochim Biophys Acta Oct l l;721(2):185-90; Fra
  • the expression polynucleotide may be stably integrated into the genome of the recipient cell. This integration may be in the cognate location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non specific location (gene augmentation).
  • the nucleic acid may be stably maintained in the cell as a separate, episomal segment of DNA. Such nucleic acid segments or "episomes" encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle.
  • One specific embodiment for a method for delivering a protein or peptide to the interior of a cell of a vertebrate in vivo comprises the step of introducing a preparation comprising a physiologically acceptable carrier and a naked polynucleotide operatively coding for the polypeptide of interest into the interstitial space of a tissue comprising the cell, whereby the naked polynucleotide is taken up into the interior of the cell and has a physiological effect.
  • This is particularly applicable for transfer in vitro but it may be applied to in vivo as well.
  • compositions for use in vitro and in vivo comprising a "naked" polynucleotide are described in PCT application No. WO 90/11092 (Vical Inc.) and also in PCT application No. WO 95/11307 (Institut Pasteur, INSERM, Universite d'Ottawa) as well as in the articles of Tascon et al. (1996) Nature Medicine. 2(8):888-892 and of Huygen et al. ((1996) Nat Med Aug;2(8):893-8).
  • the transfer of a naked polynucleotide of the invention, including a polynucleotide construct of the invention, into cells may be proceeded with a particle bombardment (biolistic), said particles being DNA-coated microprojectiles accelerated to a high velocity allowing them to pierce cell membranes and enter cells without killing them, such as described by Klein et al. ((1990) Curr Genet Feb;17(2):97-103).
  • a particle bombardment biolistic
  • the polynucleotide of the invention may be entrapped in a liposome (Ghosh and Bacchawat, (1991) Targeted Diagn Ther;4:87-103; Wong et al, (1980) Gene 10:87-94; Nicolau et al, (1987) Methods Enzymol.; 149: 157-76).
  • liposomes may further be targeted to cells expressing LSR by inco ⁇ orating leptin, triglycerides, ACRP30, or other known LSR ligands into the liposome membrane.
  • the invention provides a composition for the in vivo production of a GMG-5 globular head polypeptide described herein. It comprises a naked polynucleotide operatively coding for this polypeptide, in solution in a physiologically acceptable carrier, and suitable for introduction into a tissue to cause cells of the tissue to express the said polypeptide.
  • the amount of vector to be injected to the desired host organism varies according to the site of injection. As an indicative dose, it will be injected between 0.1 and 100 ⁇ g of the vector in an animal body, preferably a mammal body, for example a mouse body.
  • the vector according to the invention may be introduced in vitro in a host cell, preferably in a host cell previously harvested from the animal to be treated and more preferably a somatic cell such as a muscle cell.
  • a somatic cell such as a muscle cell.
  • the cell that has been transformed with the vector coding for the desired GMG-5 globular head polypeptide or the desired fragment thereof is reintroduced into the animal body in order to deliver the recombinant protein within the body either locally or systemically.
  • Another object of the invention consists of host cells recombinant for, i.e., that have been transformed or transfected with one of the polynucleotides described herein, and more precisely a polynucleotide comprising a polynucleotide encoding a GMG-5 polypeptide of the invention such as any one of those described in "Polynucleotides of the Invention". These polynucleotides can be present in cells as a result of transient or stable transfection.
  • the invention includes host cells that are transformed (prokaryotic cells) or that are transfected (eukaryotic cells) with a recombinant vector such as any one of those described in "Recombinant Vectors of the Invention".
  • a recombinant host cell of the invention comprises at least one of the polynucleotides or the recombinant vectors of the invention that are described herein.
  • Preferred host cells used as recipients for the recombinant vectors of the invention are the following : a) Prokaryotic host cells : Escherichia coli strains (I.E. DH5- ⁇ strain), Bacillus subtilis, Salmonella typhimurium, and strains from species like Pseudomonas, Streptomyces and Staphylococcus, and b) Eukaryotic host cells : HeLa cells (ATCC N°CCL2; N°CCL2.1; N°CCL2.2), Cv 1 cells (ATCC N°CCL70), COS cells (ATCC N°CRL1650; N°CRL1651), Sf-9 cells (ATCC N°CRL1711), C127 cells (ATCC N° CRL-1804), 3T3 (ATCC N° CRL-6361), CHO (ATCC N° CCL-61), human kidney 293 (ATCC N° 45504; N° CRL-1573), BHK (ECACC N° 84100501; N°
  • the constructs in the host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the selected promoter is induced by appropriate means, such as temperature shift or chemical induction, and cells are cultivated for an additional period.
  • Microbial cells employed in the expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. Such methods are well known by the skilled artisan.
  • these recombinant cells can be created in vitro or in vivo in an animal, preferably a mammal, most preferably selected from the group consisting of mice, rats, dogs, pigs, sheep, cattle, and primates, not to include humans.
  • Recombinant cells created in vitro can also be later surgically implanted in an animal, for example. Methods to create recombinant cells in vivo in animals are well-known in the art.
  • the present invention also encompasses primary, secondary, and immortalized homologously recombinant host cells of vertebrate origin, preferably mammalian origin and particularly human origin, that have been engineered to: a) insert exogenous (heterologous) polynucleotides into the endogenous chromosomal DNA of a targeted gene, b) delete endogenous chromosomal DNA, and/or c) replace endogenous chromosomal DNA with exogenous polynucleotides. Insertions, deletions, and/or replacements of polynucleotide sequences may be to the coding sequences of the targeted gene and/or to regulatory regions, such as promoter and enhancer sequences, operably associated with the targeted gene.
  • the present invention further relates to a method of making a homologously recombinant host cell in vitro or in vivo, wherein the expression of a targeted gene not normally expressed in the cell is altered.
  • the alteration causes expression of the targeted gene under normal growth conditions or under conditions suitable for producing the polypeptide encoded by the targeted gene.
  • the method comprises the steps of: (a) fransfecting the cell in vitro or in vivo with a polynucleotide construct, the polynucleotide construct comprising; (i) a targeting sequence; (ii) a regulatory sequence and/or a coding sequence; and (iii) an unpaired splice donor site, if necessary, thereby producing a transfected cell; and (b) maintaining the transfected cell in vitro or in vivo under conditions appropriate for homologous recombination.
  • the present invention further relates to a method of altering the expression of a targeted gene in a cell in vitro or in vivo wherein the gene is not normally expressed in the cell, comprising the steps of: (a) fransfecting the cell in vitro or in vivo with a polynucleotide construct, the polynucleotide construct comprising: (i) a targeting sequence; (ii) a regulatory sequence and/or a coding sequence; and (iii) an unpaired splice donor site, if necessary, thereby producing a transfected cell; and (b) maintaining the transfected cell in vitro or in vivo under conditions appropriate for homologous recombination, thereby producing a homologously recombinant cell; and (c) maintaining the homologously recombinant cell in vitro or in vivo under conditions appropriate for expression of the gene.
  • the present invention further relates to a method of making a polypeptide of the present invention by altering the expression of a targeted endogenous gene in a cell in vitro or in vivo wherein the gene is not normally expressed in the cell, comprising the steps of: a) fransfecting the cell in vitro with a polynucleotide construct, the polynucleotide construct comprising: (i) a targeting sequence; (ii) a regulatory sequence and/or a coding sequence; and (iii) an unpaired splice donor site, if necessary, thereby producing a transfected cell; (b) maintaining the transfected cell in vitro or in vivo under conditions appropriate for homologous recombination, thereby producing a homologously recombinant cell; and c) maintaining the homologously recombinant cell in vitro or in vivo under conditions appropriate for expression of the gene thereby making the polypeptide.
  • the present invention further relates to a polynucleotide construct that alters the expression of a targeted gene in a cell type in which the gene is not normally expressed. This occurs when a polynucleotide construct is inserted into the chromosomal DNA of the target cell, wherein the polynucleotide construct comprises: a) a targeting sequence; b) a regulatory sequence and/or coding sequence; and c) an unpaired splice-donor site, if necessary.
  • compositions may be produced, and methods performed, by techniques known in the art, such as those described in U.S.
  • GMG-5s in mammalian, and typically human, cells may be rendered defective, or alternatively it may be enhanced, with the insertion of a GMG-5 genomic or cDNA sequence with the replacement of the GMG-5 gene counte ⁇ art in the genome of an animal cell by a GMG-5 polynucleotide according to the invention.
  • GMG-5 genomic or cDNA sequence with the replacement of the GMG-5 gene counte ⁇ art in the genome of an animal cell by a GMG-5 polynucleotide according to the invention.
  • These genetic alterations may be generated by homologous recombination events using specific DNA constructs that have been previously described.
  • mammalian zygotes such as murine zygotes.
  • murine zygotes may undergo microinj ection with a purified DNA molecule of interest, for example a purified DNA molecule that has previously been adjusted to a concentration range from 1 ng/ml -for BAC inserts- 3 ng/ ⁇ l -for PI bacteriophage inserts- in 10 mM Tris-HCl, pH 7.4, 250 ⁇ M EDTA containing 100 mM NaCl, 30 ⁇ M spermine, and 70 ⁇ M spermidine.
  • a purified DNA molecule of interest for example a purified DNA molecule that has previously been adjusted to a concentration range from 1 ng/ml -for BAC inserts- 3 ng/ ⁇ l -for PI bacteriophage inserts- in 10 mM Tris-HCl, pH 7.4, 250 ⁇ M EDTA containing 100 mM NaCl, 30 ⁇ M spermine, and 70 ⁇ M
  • ES cell lines are derived from pluripotent, uncommitted cells of the inner cell mass of pre-implantation blastocysts.
  • Preferred ES cell lines are the following: ES-E14TG2a (ATCC No.CRL-1821), ES-D3
  • feeder cells are primary embryonic fibroblasts that are established from tissue of day 13- day 14 embryos of virtually any mouse strain, that are maintained in culture, such as described by Abbondanzo et al. (1993; Methods
  • the constructs in the host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the selected promoter is induced by appropriate means, such as temperature shift or chemical induction, and cells are cultivated for an additional period.
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in the expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. Such methods are well known by the skilled artisan.
  • the present invention also provides methods and compositions for the generation of non- human animals and plants that express the recombinant GMG-5 polypeptides, of the present invention.
  • the animals or plants can be transgenic, i.e. each of their cells contains a gene encoding a GMG-5 polypeptide, or, alternatively, a polynucleotide encoding a GMG-5 polypeptide can be introduced into somatic cells of the animal or plant, e.g. into mammary secretory epithelial cells of a mammal.
  • the non-human animal is a mammal such as a cow, sheep, goat, pig, or rabbit.
  • transgenic mammals can be produced, e.g., by fransfecting a pluripotential stem cell such as an ES cell with a polynucleotide encoding a polypeptide of interest. Successfully transformed ES cells can then be introduced into an early stage embryo which is then implanted into the uterus of a mammal of the same species.
  • a pluripotential stem cell such as an ES cell with a polynucleotide encoding a polypeptide of interest.
  • Successfully transformed ES cells can then be introduced into an early stage embryo which is then implanted into the uterus of a mammal of the same species.
  • the transformed (“transgenic") cells will comprise part of the germ line of the resulting animal, and adult animals comprising the transgenic cells in the germ line can then be mated to other animals, thereby eventually producing a population of transgenic animals that have the fransgene in each of their cells, and which can stably transmit the fransgene to each of their offspnng.
  • Other methods of introducing the polynucleotide can be used, for example introducing the polynucleotide encoding the polypeptide of interest into a fertilized egg or early stage embryo via microinjection.
  • the fransgene may be introduced into an animal by infection of zygotes with a retrovirus containing the fransgene (Jaenisch, R. (1976) Proc. Natl. Acad. Sci. USA 73, 1260-1264).
  • transgenic mammals are described, e.g., in Wall et al. (1992) J Cell Biochem
  • the polynucleotides are microinjected into the fertilized oocyte.
  • fertilized oocytes are microinjected using standard techniques, and then cultured in vifrountil a "pre-implantation embryo" is obtained.
  • pre-implantation embryos preferably contain approximately 16 to 150 cells.
  • Methods for cultunng fertilized oocytes to the pre- implantation stage are descnbed, e.g., by Gordon et al. ((1984) Methods in Enzymology, 101, 414); Hogan et al. ((1986) in Manipulating the mouse embryo. A Laboratory Manual. Cold Spnng Harbor Laboratory Press, Cold Spring Harbor, N.Y) (for the mouse embryo); Hammer et al.
  • Pre-implantation embryos are then transferred to an appropriate female by standard methods to permit the birth of a transgenic or chimenc animal, depending upon the stage of development when the fransgene is introduced.
  • the detection of fransgene integration in pre-implantation embryos is often desirable using any of the herein-descnbed methods.
  • Any of a number of methods can be used to detect the presence of a fransgene in a pre-implantation embryo. For example, one or more cells may be removed from the pre-implantation embryo, and the presence or absence of the fransgene in the removed cell or cells can be detected using any standard method e.g. PCR. Alternatively, the presence of a fransgene can be detected in utero or post partum using standard methods.
  • transgenic mammals are generated that secrete recombinant GMG-5 polypeptides in their milk.
  • the mammary gland is a highly efficient protem-producing organ, such methods can be used to produce protein concenfrations in the gram per liter range, and often significantly more.
  • expression m the mammary gland is accomplished by operably linking the polynucleotide encoding the GMG-5 polypeptide to a mammary gland specific promoter and, optionally, other regulatory elements.
  • Suitable promoters and other elements include, but are not limited to, those derived from mammalian short and long WAP, alpha, beta, and kappa, casein, alpha and beta lactoglobu n, beta-CN 5 ' genes, as well as the mouse mammary tumor virus (MMTV) promoter.
  • MMTV mouse mammary tumor virus
  • Such promoters and other elements may be denved from any mammal, including, but not limited to, cows, goats, sheep, pigs, mice, rabbits, and guinea pigs.
  • Promoter and other regulatory sequences, vectors, and other relevant teachings are provided, e.g., by Clark (1998) J Mammary Gland Biol Neoplasm 3:337-50; Jost et al.
  • polypeptides of the invention can be produced in milk by introducing polynucleotides encoding the polypeptides into somatic cells of the mammary gland in vivo, e.g. mammary secreting epithelial cells.
  • plasmid DNA can be infused through the nipple canal, e.g. in association with DEAE-dexfran (see, e.g., Hens et al. (2000) Biochim. Biophys.
  • the polynucleotide may be operably linked to a mammary gland specific promoter, as descnbed above, or, alternatively, any strongly expressing promoter such as CMV or MoMLV LTR.
  • fransfecting cells such as mammary epithelial cells, e.g. MacT cells (bovine mammary epithelial cells) or GME cells (goat mammary epithelial cells), in vitro and assessing the efficiency of transfection and expression of the fransgene in the cells.
  • mammary epithelial cells e.g. MacT cells (bovine mammary epithelial cells) or GME cells (goat mammary epithelial cells
  • the polynucleotides can be administered in any suitable formulation, at any of a range of concentrations (e.g. 1-500 ⁇ g/ml, preferably 50-100 ⁇ g/ml), at any volume (e.g. 1-100 ml, preferably 1 to 20 ml), and can be administered any number of times (e.g. 1, 2, 3, 5, or 10 times), at any frequency (e.g. every 1, 2, 3, 5, 10, or any number of days).
  • concentrations, frequencies, modes of administration, etc. will depend upon the particular polynucleotide, vector, animal, etc, and can readily be determined by one of skill in the art.
  • a retroviral vector such as as Gibbon ape leukemia viral vector is used, as descnbed in Archer et al. ((1994) PNAS 91 :6840-6844).
  • retroviral infection typically requires cell division, cell division in the mammary glands can be stimulated in conjunction with the administration of the vector, e.g. using a factor such as esfrodiol benzoate, progesterone, rese ⁇ ine, or dexamethasone.
  • retroviral and other methods of infection can be facilitated using accessory compounds such as polybrene.
  • the quantity of milk obtained, and thus the quantity of GMG-5 polypeptides produced can be enhanced using any standard method of lactation induction, e.g. using hexesfrol, estrogen, and/or progesterone.
  • the polynucleotides used in such embodiments can either encode a full-length GMG-5 polypeptide or a GMG-5 fragment.
  • the encoded polypeptide will include a signal sequence to ensure the secretion of the protein into the milk.
  • the full length protein can, e.g., be isolated from milk and cleaved in vitro using a suitable protease.
  • a second, protease-encoding polynucleotide can be introduced into the animal or into the mammary gland cells, whereby expression of the protease results in the cleavage of the GMG-5 polypeptide in vivo, thereby allowing the direct isolation of GMG-5 fragments from milk.
  • the GMG-5 polypeptides of the invention can be administered to non-human animals and/or humans, alone or in pharmaceutical or physiologically acceptable compositions where they are mixed with suitable carriers or excipient(s).
  • the pharmaceutical or physiologically acceptable composition is then provided at a therapeutically effective dose.
  • a therapeutically effective dose refers to that amount of a GMG-5 polypeptide sufficient to result in prevention or amelioration of symptoms or physiological status of metabolic-related diseases or disorders as determined by the methods described herein.
  • a therapeutically effective dose can also refer to the amount of a GMG-5 polypeptide necessary for a reduction in weight or a prevention of an increase in weight or prevention of an increase in the rate of weight gain in persons desiring this affect for cosmetic reasons.
  • a therapeutically effective dosage of a GMG-5 polypeptide of the invention is that dosage that is adequate to promote weight loss or weight gain with continued periodic use or administration.
  • Techniques for formulation and administration of GMG-5 polypeptides may be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co, Easton, PA, latest edition.
  • Other diseases or disorders that GMG-5 polypeptides of the invention could be used to treat or prevent include, but are not limited to, obesity and obesity-related diseases and disorders such as obesity, impaired glucose tolerance, insulin resistance, atherosclerosis, atheromatous disease, heart disease, hypertension, stroke, Syndrome X, Noninsulin Dependent Diabetes Mellitus (NIDDM, or Type II diabetes) and Insulin Dependent Diabetes Mellitus (IDDM or Type I diabetes).
  • NIDDM Noninsulin Dependent Diabetes Mellitus
  • IDDM Insulin Dependent Diabetes Mellitus
  • Diabetes- related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, retinopathy, neuropathy, renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other obesity-related disorders to be treated by compounds of the invention include hyperlipidemia and hyperuricemia.
  • Yet other obesity-related diseases or disorders of the invention include cachexia, wasting, AEDS- related weight loss, cancer-related weight loss, visceral obesity, anorexia, and bulimia.
  • the GMG-5 polypeptides may also be used to enhance physical performance during work or exercise or enhance a feeling of general well-being. Physical performance activities include walking, running, jumping, lifting and/or climbing.
  • the GMG-5 polypeptides or antagonists thereof may also be used to treat dyslexia, attention-deficit disorder (ADD), attention-deficit/hyperactivity disorder (ADHD), and psychiatric disorders such as schizophrenia by modulating fatty acid metabolism, more specifically, the production of certain long-chain polyunsaturated fatty acids.
  • ADD attention-deficit disorder
  • ADHD attention-deficit/hyperactivity disorder
  • psychiatric disorders such as schizophrenia by modulating fatty acid metabolism, more specifically, the production of certain long-chain polyunsaturated fatty acids.
  • the GMG-5 polypeptides of the invention may be provided alone or in combination with other pharmaceutically or physiologically acceptable compounds.
  • Other compounds useful for the treatment of obesity and other diseases and disorders are currently well-known in the art.
  • the GMG-5 polypeptides are useful for, and used in, the treatment of insulin resistance and diabetes using methods described herein and known in the art.
  • a preferred embodiments relates to process for the therapeutic modification and regulation of glucose metabolism in an animal or human subject, which comprises administering to a subject in need of treatment (alternatively on a timed daily basis) GMG-5 polypeptide (or polynucleotide encoding said polypeptide) in dosage amount and for a period sufficient to reduce plasma glucose levels in said animal or human subject.
  • Further preferred embodiments relate to methods for the prophylaxis or treatment of diabetes comprising administering to a subject in need of treatment (alternatively on a timed daily basis) a GMG-5 polypeptide (or polynucleotide encoding said polypeptide) in dosage amount and for a period sufficient to reduce plasma glucose levels in said animal or human subject.
  • Suitable routes of administration include oral, nasal, rectal, transmucosal, or intestinal administration, parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as infrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, infrapulmonary (inhaled) or intraocular injections using methods known in the art.
  • a particularly useful method of administering compounds for promoting weight loss involves surgical implantation, for example into the abdominal cavity of the recipient, of a device for delivering GMG-5 polypeptidesover an extended period of time.
  • Other particularly preferred routes of administration are aerosol and depot formulation. Sustained release formulations, particularly depot, of the invented medicaments are expressly contemplated.
  • compositions and medicaments for use in accordance with the present invention may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries. Proper formulation is dependent upon the route of administration chosen.
  • the medicaments described herein will include a pharmaceutically or physiologically acceptable acceptable carrier and at least one polypeptide that is a GMG-5 polypeptide of the invention.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer such as a phosphate or bicarbonate buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer such as a phosphate or bicarbonate buffer.
  • penevers appropriate to the barrier to be permeated are used in the formulation. Such peneflops are generally known in the art.
  • compositions that can be taken orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such adminisfration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable gaseous propellant, e.g., carbon dioxide.
  • a suitable gaseous propellant e.g., carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin, for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and or dispersing agents.
  • Pharmaceutical or physiologically acceptable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
  • Aqueous suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dexfran.
  • the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder or lyophilized form for constitution with a suitable vehicle, such as sterile pyrogen-free water, before use.
  • a suitable vehicle such as sterile pyrogen-free water
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve their intended pu ⁇ ose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes or encompasses a concentration point or range shown to increase leptin or lipoprotein uptake or binding in an in vitro system. Such information can be used to more accurately determine useful doses in humans.
  • a therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms in a patient. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50, (the dose lethal to 50% of the test population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds that exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50, with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See, e.g., Fingl et al, 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1).
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active compound which are sufficient to maintain or prevent weight loss or gain, depending on the particular situation. Dosages necessary to achieve these effects will depend on individual characteristics and route of administration.
  • Dosage intervals can also be determined using the value for the minimum effective concentration.
  • Compounds should be administered using a regimen that maintains plasma levels above the minimum effective concentration for 10-90% of the time, preferably between 30-90%; and most preferably between 50-90%.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • a preferred dosage range for the amount of a GMG-5 polypeptide of the invention which can be administered on a daily or regular basis to achieve desired results, including a reduction in levels of circulating plasma friglyceride-rich lipoproteins, range from 0.05 - 1.0 mg/kg body mass.
  • a more preferred dosage range is from 0.1 - 5 mg/kg.
  • a more preferred dose is 0.25 - 2.5 mg/kg.
  • these daily dosages can be delivered or administered in small amounts periodically during the course of a day. It is noted that these dosage ranges are only preferred ranges and are not meant to be limiting to the invention.
  • the invention is drawn inter alia to methods of preventing or treating metabolic-related diseases and disorders comprising providing an individual in need of such treatment with a GMG- 5 polypeptide of the invention.
  • the GMG-5 polypeptide has metabolic-related activity either in vitro or in vivo.
  • the GMG-5 polypeptide is provided to the individual in a pharmaceutical composition that is preferably taken orally.
  • the individual is a mammal, and most preferably a human.
  • the metabolic-related disease or disorder is selected from the group consisting of atherosclerosis, cardiovascular disease, impaired glucose tolerance, insulin resistance, hypertension, stroke, Syndrome X, Type I diabetes, Type II diabetes and lipoatrophic diabetes.
  • Diabetes-related complications to be treated by the methods of the invention include microangiopathic lesions, ocular lesions, retinopathy, neuropathy and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other metabolic-related disorders to be freated by compounds of the invention include hyperlipidemia, hypertriglyceridemia, and hyperuricemia.
  • Yet other metabolic-related diseases or disorders of the invention include cachexia, wasting, AIDS-related weight loss, cancer-related weight loss, visceral obesity, neoplasia-related weight loss, anorexia, and bulimia.
  • GMG-5 polypeptides in pharmaceutical compositions are used to modulate body weight in healthy individuals for cosmetic reasons.
  • the invention also features a method of preventing or treating metabolic-related diseases and disorders comprising providing an individual in need of such treatment with a compound identified by assays of the invention (described in Section VI of the Preferred Embodiments of the Invention and in the Examples).
  • a compound identified by assays of the invention (described in Section VI of the Preferred Embodiments of the Invention and in the Examples).
  • these compounds antagonize or agonize effects of GMG-5 polypeptides in cells in vitro, muscles ex vivo, or in animal models.
  • these compounds agonize or antagonize the effects of GMG-5 polypeptides on leptin and/or lipoprotein uptake and/or binding.
  • these compounds prevent the interaction, binding, or uptake of GMG-5 polypeptides with LSR in vitro or in vivo.
  • the compound is provided to the individual in a pharmaceutical composition that is preferably taken orally.
  • the individual is a mammal, and most preferably a human.
  • the metabolic-related disease or disorder is selected from the group consisting of obesity and metabolic-related diseases and disorders such as atherosclerosis, heart disease, insulin resistance, hypertension, stroke, Syndrome X, Type I diabetes, Type II diabetes, and lipoatrophic diabetes.
  • Diabetes-related complications to be freated by the methods of the invention include microangiopathic lesions, ocular lesions, retinopathy, neuropathy and renal lesions.
  • Heart disease includes, but is not limited to, cardiac insufficiency, coronary insufficiency, and high blood pressure.
  • Other metabolic-related disorders to be treated by compounds of the invention include hyperlipidemia, hypertriglyceridemia, and hyperuricemia.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control blood glucose in some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance, in combination with insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control body weight in some 67
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some individuals, particularly those with Type II diabetes or insulin resistance,without insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition further provides a method for the use as an inhibitor of the progression from impaired glucose tolerance to insulin resistance.
  • the invention features methods of screening for one or more compounds that modulate the activity of GMG-5 in cells, which includes providing potential compounds to be tested to the cells,. Exemplary assays that may be used are described in the Examples section. To these assays would be added compounds to be tested for their inhibitory or stimulatory activity as compared to the effects of GMG-5 polypeptides alone. Other assays in which an effect is observed based on the addition of GMG-5 polypeptides can also be used to screen for modulators of GMG-5 polypeptide activity or effects of the presence of GMG-5 polypeptides on cells.
  • the essential step is to apply an unknown compound and then to monitor an assay for a change from what is seen when only GMG-5 polypeptides are applied to the cell.
  • a change is defined as something that is significantly different in the presence of the compound plus GMG-5 polypeptide compared to GMG-5 polypeptide alone. In this case, significantly different would be an "increase” or a "decrease” in a measurable effect of at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75%.
  • modulation refers to a measurable change in an activity. Examples include, but are not limited to, lipolysis stimulated receptor (LSR) modulation, leptin modulation, lipoprotein modulation, plasma FFA levels, FFA oxidation, TG levels, glucose levels, and weight. These effects can be in vitro or preferably in vivo. Modulation of an activity can be either an increase or a decrease in the activity. Thus, LSR activity can be increased or decreased, leptin activity can be increased or decreased, and lipoprotein activity can be increased or decreased. Similarly, FFA, TG, glucose levels and weight can be increased or decreased in vivo Free Fatty Acid oxidation can be increased or decreased in vivo or ex vivo.
  • LSR lipolysis stimulated receptor
  • LSR activity is meant expression of LSR on the surface of the cell, or in a particular conformation, as well as its ability to bind, uptake, and degrade leptin and lipoprotein.
  • leptin activity is meant its binding, uptake and degradation by LSR, as well as its transport across a blood brain barrier, and potentially these occurrences where LSR is not necessarily the mediating factor or the only mediating factor.
  • lipoprotein activity is meant its binding, uptake and degradation by LSR, as well as these occurrences where LSR is not necessarily the mediating factor or the only mediating factor. Exemplary assays are provided in the Examples.
  • increasing refers to the ability of a compound to increase the activity of GMG-5 polypeptides in some measurable way compared to the effect of GMG-5 polypeptides in its absence.
  • an increase in activity is at least 25%, 30%, 35%, 40%, 45%, 50%,
  • the term "decreasing" as used herein refers to the ability of a compound to decrease an activity in some measurable way compared to the effect of a GMG-5 polypeptide in its absence.
  • the presence of the compound decreases the plasma concenfrations of FFA, TG, and glucose in mice.
  • an decrease in activity is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 10%, or 75% as compared to the level of activity in the presence of the GMG-5 polypeptides alone.
  • the invention features a method for identifying a potential compound to decrease body mass in individuals in need of decreasing body mass comprising: a) contacting a cell with a GMG-5 polypeptide and a candidate compound; b) detecting a result selected from the group consisting of LSR modulation, leptin modulation, increase in glucose uptake or oxidation, decrease in blood lipid or triglyceride levels, increase in lipoprotein binding, uptake or degradation; FFA oxidation increase; and c) wherein said result identifies said potential compound if said result differs from said result when said cell is contacted with the GMG-5 polypeptide alone.
  • the invention features a method for identifying a potential compound to increase body mass in individuals in need of increasing body mass comprising: a) contacting a cell with a GMG-5 polypeptide and a candidate compound; b) detecting a result selected from the group consisting of LSR modulation, leptin modulation, decrease in glucose uptake or oxidation, increase in blood lipid or triglyceride levels, decrease in lipoprotein binding, uptake or degradation; FFA oxidation decrease; and c) wherein said result identifies said potential compound if said result differs from said result when said cell is contacted with the GMG-5 polypeptide alone.
  • said potential compound is selected from the group consisting of peptides, peptide libraries, non-peptide libraries, peptoids, fatty acids, lipoproteins, medicaments, antibodies, small molecules, proteases and protease inhibitors.
  • Epitopes and Antibody Fusions 66 individuals, particularly those with Type I diabetes, Type El diabetes, or insulin resistance, in combination with insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to control blood glucose in some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance, alone, without combination of insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to confrol body weight in some individuals, particularly those with Type II diabetes or insulin resistance, alone, without combination of insulin therapy.
  • the confrol of body weight is due in part or in whole to a decrease in mass of l)subcutaneous adipose tissue and/or 2)viseral (omental) adipose tissue.
  • the present invention may be used in complementary therapy, particularly in some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance, to improve their weight or glucose confrol in combination with an insulin secretagogue or an insulin sensitising agent.
  • the insulin secretagogue is l,l-dimethyl-2- (2-mo ⁇ holino phenyl)guanidine fumarate (BTS67582) or a sulphonylurea selected from tolbutamide, tolazamide, chlo ⁇ ropamide, glibenclamide, glimepiride, glipizide and glidazide.
  • the insulin sensitising agent is selected from metformin, ciglitazone, troglitazone and pioglitazone.
  • the present invention further provides a method of improving the body weight or glucose control of some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance,alone, without an insulin secretagogue or an insulin sensitising agent.
  • the present invention may be administered either concomitantly or concurrently, with the insulin secretagogue or insulin sensitising agent for example in the form of separate dosage units to be used simultaneously, separately or sequentially (either before or after the secretagogue or either before or after the sensitising agent).
  • the present invention further provides for a composition of pharmaceutical or physiologically acceptable composition and an oral insulin secretagogue or insulin sensitising agent as a combined preparation for simultaneous, separate or sequential use for the improvement of body weight or glucose confrol in some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance.
  • the present invention of said pharmaceutical or physiologically acceptable composition further provides a method for the use as an insulin sensitiser.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some individuals, particularly those with Type I diabetes, Type II diabetes, or insulin resistance, in combination with insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition can be used as a method to improve insulin sensitivity in some individuals, particularly those with Type II diabetes or insulin resistance,without insulin therapy.
  • the present invention of said pharmaceutical or physiologically acceptable composition further provides a method for the use as an inhibitor of the progression from impaired glucose tolerance to insulin resistance.
  • the invention features methods of screening for one or more compounds that modulate the activity of GMG-5 in cells, which includes providing potential compounds to be tested to the cells,. Exemplary assays that may be used are described in the Examples section. To these assays would be added compounds to be tested for their inhibitory or stimulatory activity as compared to the effects of GMG-5 polypeptides alone. Other assays in which an effect is observed based on the addition of GMG-5 polypeptides can also be used to screen for modulators of GMG-5 polypeptide activity or effects of the presence of GMG-5 polypeptides on cells.
  • the essential step is to apply an unknown compound and then to monitor an assay for a change from what is seen when only GMG-5 polypeptides are applied to the cell.
  • a change is defined as something that is significantly different in the presence of the compound plus GMG-5 polypeptide compared to GMG-5 polypeptide alone. In this case, significantly different would be an "increase” or a "decrease” in a measurable effect of at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75%.
  • modulation refers to a measurable change in an activity. Examples include, but are not limited to, lipolysis stimulated receptor (LSR) modulation, leptin modulation, lipoprotein modulation, plasma FFA levels, FFA oxidation, TG levels, glucose levels, and weight. These effects can be in vitro or preferably in vivo. Modulation of an activity can be either an increase or a decrease in the activity. Thus, LSR activity can be increased or decreased, leptin activity can be increased or decreased, and lipoprotein activity can be increased or decreased. Similarly, FFA, TG, glucose levels and weight can be increased or decreased in vivo Free Fatty Acid oxidation can be increased or decreased in vivo or ex vivo.
  • LSR lipolysis stimulated receptor
  • LSR activity is meant expression of LSR on the surface of the cell, or in a particular conformation, as well as its ability to bind, uptake, and degrade leptin and lipoprotein.
  • leptin activity is meant its binding, uptake and degradation by LSR, as well as its transport across a blood brain barrier, and potentially these occurrences where LSR is not necessarily the mediating factor or the only mediating factor.
  • lipoprotein activity is meant its binding, uptake and degradation by LSR, as well as these occurrences where LSR is not necessarily the mediating factor or the only mediating factor. Exemplary assays are provided in the Examples.
  • increasing refers to the ability of a compound to increase the activity of GMG-5 polypeptides in some measurable way compared to the effect of GMG-5 polypeptides in its absence.
  • an increase in activity is at least 25%, 30%, 35%, 40%, 45%, 50%,
  • the term "decreasing" as used herein refers to the ability of a compound to decrease an activity in some measurable way compared to the effect of a GMG-5 polypeptide in its absence.
  • the presence of the compound decreases the plasma concenfrations of FFA, TG, and glucose in mice.
  • an decrease in activity is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%), 70%, or 75% as compared to the level of activity in the presence of the GMG-5 polypeptides alone.
  • the invention features a method for identifying a potential compound to decrease body mass in individuals in need of decreasing body mass comprising: a) contacting a cell with a GMG-5 polypeptide and a candidate compound; b) detecting a result selected from the group consisting of LSR modulation, leptin modulation, increase in glucose uptake or oxidation, decrease in blood lipid or triglyceride levels, increase in lipoprotein binding, uptake or degradation; FFA oxidation increase; and c) wherein said result identifies said potential compound if said result differs from said result when said cell is contacted with the GMG-5 polypeptide alone.
  • the invention features a method for identifying a potential compound to increase body mass in individuals in need of increasing body mass comprising: a) contacting a cell with a GMG-5 polypeptide and a candidate compound; b) detecting a result selected from the group consisting of LSR modulation, leptin modulation, decrease in glucose uptake or oxidation, increase in blood lipid or triglyceride levels, decrease in lipoprotein binding, uptake or degradation; FFA oxidation decrease; and c) wherein said result identifies said potential compound if said result differs from said result when said cell is contacted with the GMG-5 polypeptide alone.
  • said potential compound is selected from the group consisting of peptides, peptide libraries, non-peptide libraries, peptoids, fatty acids, lipoproteins, medicaments, antibodies, small molecules, proteases and protease inhibitors.
  • a preferred embodiment of the present invention is directed to eiptope-bearing polypeptides and epitope-bearing polypeptide fragments. These epitopes may be "antigenic epitopes" or both an
  • an “immunogenic epitope” is defined as a part of a protein that elicits an antibody response in vivo when the polypeptide is the immunogen.
  • a region of polypeptide to which an antibody binds is defined as an "antigenic determinant" or "antigenic epitope.”
  • the number of immunogenic epitopes of a protein generally is less than the number of antigenic epitopes. See, e.g., Geysen, et al. (1983) Proc. Natl. Acad. Sci.
  • an epitope can comprise as few as 3 amino acids in a spatial conformation which is unique to the epitope. Generally an epitope consists of at least 6 such amino acids, and more often at least 8-10 such amino acids. In preferred embodiment, antigenic epitopes comprise a number of amino acids that is any integer between 3 and 50. Fragments which function as epitopes may be produced by any conventional means. See, e.g., Houghten, R. A, Proc. Natl. Acad. Sci. USA 82:5131-5135 (1985), further described in U.S.
  • Patent No. 4,631,211 Methods for determining the amino acids which make up an immunogenic epitope include x-ray crystallography, 2-dimensional nuclear magnetic resonance, and epitope mapping, e.g., the Pepscan method described by H. Mario Geysen et al. (1984); Proc. Natl. Acad. Sci. U.S.A. 81:3998-4002; PCT Publication No. WO 84/03564; and PCT Publication No. WO 84/03506.
  • Another example is the algorithm of Jameson and Wolf, Comp. Appl. Biosci. 4:181-186 (1988) (said references inco ⁇ orated by reference in their entireties).
  • the Jameson-Wolf antigenic analysis for example, may be performed using the computer program PROTEAN, using default parameters (Version 4.0 Windows, DNASTAR, Inc., 1228 South Park Street Madison, WI).
  • the epitope-bearing fragments of the present invention preferably comprises 6 to 50 amino acids (i.e. any integer between 6 and 50, inclusive) of a polypeptide of the present invention. Also, included in the present invention are antigenic fragments between the integers of 6 and the full length sequence of the sequence listing. All combinations of sequences between the integers of 6 and the full-length sequence of a polypeptide of the present invention are included.
  • the epitope- bearing fragments may be specified by either the number of contiguous amino acid residues (as a sub-genus) or by specific N-terminal and C-terminal positions (as species) as described above for the polypeptide fragments of the present invention. Any number of epitope-bearing fragments of the present invention may also be excluded in the same manner.
  • Antigenic epitopes are useful, for example, to raise antibodies, including monoclonal antibodies that specifically bind the epitope (See,Wilson et al, 1984; and Sutcliffe, J. G et al, 1983). The antibodies are then used in various techniques such as diagnostic and tissue/cell identification techniques, as described herein, and in purification methods. 70 Similarly, immunogenic epitopes can be used to induce antibodies according to methods well known in the art (See, Sutcliffe et al, supra; Wilson et al, supra; Chow, M. et al.;(1985) and
  • a preferred immunogenic epitope includes the polypeptides of the sequence listing.
  • the immunogenic epitopes may be presented together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse) if necessary.
  • Immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide (e.g., in Western blotting.).
  • Epitope-bearing polypeptides of the present invention are used to induce antibodies according to methods well known in the art including, but not limited to, in vivo immunization, in vitro immunization, and phage display methods (See, e.g., Sutcliffe, et al, supra; Wilson, et al, supra, and Bittle, et al, 1985). If in vivo immunization is used, animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling of the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin (RLH) or tetanus toxoid.
  • a macromolecular carrier such as keyhole limpet hemacyanin (RLH) or tetanus toxoid.
  • peptides containing cysteine residues may be coupled to a carrier using a linker such as - maleimidobenzoyl-N-hydroxysuccinimide ester (MBS), while other peptides may be coupled to carriers using a more general linking agent such as glutaraldehyde.
  • a linker such as - maleimidobenzoyl-N-hydroxysuccinimide ester (MBS)
  • MBS - maleimidobenzoyl-N-hydroxysuccinimide ester
  • glutaraldehyde a linker
  • Animals such as rabbits, rats and mice are immunized with either free or carrier-coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 ⁇ gs of peptide or carrier protein and Freund's adjuvant.
  • booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody, which can be detected, for example, by ELISA assay using free peptide adsorbed to a solid surface.
  • the titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti-peptide antibodies, for instance, by adso ⁇ tion to the peptide on a solid support and elution of the selected antibodies according to methods well known in the art.
  • polypeptides of the present invention including, but not limited to, polypeptides comprising an immunogenic or antigenic epitope can be fused to heterologous polypeptide sequences.
  • the polypeptides of the present invention may be fused with the constant region comprising portions of immunoglobulins (IgA, IgE, IgG, IgM), or portions of the constant region (CHI, CH2, CH3, any combination thereof including both entire domains and portions thereof) resulting in chimeric polypeptides.
  • IgA, IgE, IgG, IgM immunoglobulins
  • CHI constant region
  • CH2, CH3 any combination thereof including both entire domains and portions thereof
  • DNA shuffling may be employed to modulate the activities of polypeptides of the present invention thereby effectively generating agonists and antagonists of the polypeptides. See, for example, U.S. Patent Nos.: 5,605,793; 5,811,238; 5,834,252; 5,837,458; and Patten, P.A, et al,
  • one or more components, motifs, sections, parts, domains, fragments, etc, of coding polynucleotides of the invention, or the polypeptides encoded thereby may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules.
  • the present invention further relates to antibodies and T-cell antigen receptors (TCR), which specifically bind the polypeptides, and more specifically, the epitopes of the polypeptides of the present invention.
  • TCR T-cell antigen receptors
  • the antibodies of the present invention include IgG (including IgGl, IgG2, IgG3, and IgG4), IgA (including IgAl and IgA2), IgD, IgE, or IgM, and IgY.
  • antibody is meant to include whole antibodies, including single-chain whole antibodies, and antigen binding fragments thereof.
  • the antibodies are human antigen binding antibody fragments of the present invention include, but are not limited to, Fab, Fab' F(ab)2 and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a V L or V H domain.
  • the antibodies may be from any animal origin including birds and mammals.
  • the antibodies are human, murine, rabbit, goat, guinea pig, camel, horse, or chicken.
  • Antigen-binding antibody fragments may comprise the variable region(s) alone or in combination with the entire or partial of the following: hinge region, CHI, CH2, and CH3 domains. Also included in the invention are any combinations of variable region(s) and hinge region, CHI, CH2, and CH3 domains.
  • the present invention further includes chimeric, humanized, and human monoclonal and polyclonal antibodies, which specifically bind the polypeptides of the present invention.
  • the present invention further includes antibodies that are anti-idiotypic to the antibodies of the present invention.
  • the antibodies of the present invention may be monospecific, bispecific, and trispecific or have greater multispecificity. Multispecific antibodies may be specific for different epitopes of a polypeptide of the present invention or may be specific for both a polypeptide of the present invention as well as for heterologous compositions, such as a heterologous polypeptide or solid 72 support material. See, e.g., WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, A. et al. (1991); US Patents 5,573,920, 4,474,893, 5,601,819, 4,714,681, 4,925,648; Rostelny, S.A. et al.
  • Antibodies of the present invention may be described or specified in terms of the epitope(s) or epitope-bearing portion(s) of a polypeptide of the present invention, which are recognized or specifically bound by the antibody.
  • the antibodies may specifically bind a full-length protein encoded by a nucleic acid of the present invention, a mature protein (i.e., the protein generated by cleavage of the signal peptide) encoded by a nucleic acid of the present invention, a signal peptide encoded by a nucleic acid of the present invention, or any other polypeptide of the present invention.
  • the epitope(s) or epitope bearing polypeptide portion(s) may be specified as described herein, e.g., by N-terminal and C-terminal positions, by size in contiguous amino acid residues, or otherwise described herein.
  • Antibodies which specifically bind any epitope or polypeptide of the present invention may also be excluded as individual species. Therefore, the present invention includes antibodies that specifically bind specified polypeptides of the present invention, and allows for the exclusion of the same.
  • Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not specifically bind any other analog, ortholog, or homolog of the polypeptides of the present invention are included. Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using methods known in the art and described herein, eg, using FASTDB and the parameters set forth herein) to a polypeptide of the present invention are also included in the present invention.
  • antibodies which only bind polypeptides encoded by polynucleotides, which hybridize to a polynucleotide of the present invention under stringent hybridization conditions (as described herein).
  • Antibodies of the present invention may also be described or specified in terms of their binding affinity.
  • Preferred binding affinities include those with a dissociation constant or Rd value less than 5X10- 6 M, 10 _6 M, 5X10 7 M, 10- 7 M, 5X10 '8 M, 10 "8 M, 5X10 "9 M, 10 _9 M, 5X10 " 10 M, 10-'°M, 5X10- U M, 10-"M, 5X10 "12 M, 10 "12 M, 5X10 "13 M, 10 "13 M, 5X10- 14 M, 10 "14 M, 5X10 "15 M, and 10- 15 M.
  • Antibodies of the present invention have uses that include, but are not limited to, methods known in the art to purify, detect, and target the polypeptides of the present invention including both in vitro and in vivo diagnostic and therapeutic methods.
  • the antibodies have use in immunoassays for qualitatively and quantitatively measuring levels of the polypeptides of the present invention in biological samples (See, e.g., Harlow et al, 1988).
  • the antibodies of the present invention may be used either alone or in combination with other compositions.
  • the antibodies may further be recombinantly fused to a heterologous polypeptide at the N- or C-terminus or chemically conjugated (including covalent and non-covalent 73 conjugations) to polypeptides or other compositions.
  • antibodies of the present invention may be recombinantly fused or conjugated to molecules useful as labels in detection assays and effector molecules such as heterologous polypeptides, drugs, or toxins. See, e.g., WO
  • the antibodies of the present invention may be prepared by any suitable method known in the art.
  • a polypeptide of the present invention or an antigenic fragment thereof can be administered to an animal in order to induce the production of sera containing polyclonal antibodies.
  • the term “monoclonal antibody” is not limited to antibodies produced through hybridoma technology.
  • antibody refers to a polypeptide or group of polypeptides which are comprised of at least one binding domain, where a binding domain is formed from the folding of variable domains of an antibody molecule to form three-dimensional binding spaces with an internal surface shape and charge distribution complementary to the features of an antigenic determinant of an antigen, which allows an immunological reaction with the antigen.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technology.
  • Fab and F(ab')2 fragments may be produced, for example, from hybridoma-produced antibodies by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • antibodies of the present invention can be produced through the application of recombinant DNA technology or through synthetic chemistry using methods known in the art.
  • the antibodies of the present invention can be prepared using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of a phage particle, which carries polynucleotide sequences encoding them.
  • Phage with a desired binding property are selected from a repertoire or combinatorial antibody library (e.g. human or murine) by selecting directly with antigen, typically antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
  • Examples of phage display methods that can be used to make the antibodies of the present invention include those disclosed in Brinkman U. et al. (1995); Ames, R.S. et al. (1995); Rettleborough, CA. et al. (1994); Persic, L. et al. (1997); Burton, D.R. et al.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host including mammalian cells, insect cells, plant cells, yeast, and bacteria.
  • techniques to recombinantly produce Fab, Fab' F(ab)2 and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in WO 92/22324; Mullinax, R.L. et al. (1992); and Sawai,
  • Antibodies can be humanized using a variety of techniques including CDR-grafting (EP 0 239 400; WO 91/09967; US Patent 5,530,101; and 5,585,089), veneering or resurfacing, (EP 0 592 106; EP 0 519 596; Padlan E.A, 1991; Srudnicka GM. et al, 1994; Roguska M.A. et al, 1994), and chain shuffling (US Patent 5,565,332). Human antibodies can be made by a variety of methods known in the art including phage display methods described above.
  • antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to a polypeptide of the present invention may be specific for antigens other than polypeptides of the present invention.
  • antibodies may be used to target the polypeptides of the present invention to particular cell types, either in vitro or in vivo, by fusing or conjugating the polypeptides of the present invention to antibodies specific for particular cell surface receptors.
  • Antibodies fused or conjugated to the polypeptides of the present invention may also be used in in vitro immunoassays and purification methods using methods known in the art (See e.g., Harbor et al.
  • the present invention further includes compositions comprising the polypeptides of the present invention fused or conjugated to antibody domains other than the variable regions.
  • the polypeptides of the present invention may be fused or conjugated to an antibody Fc region, or portion thereof.
  • the antibody portion fused to a polypeptide of the present invention may comprise the hinge region, CHI domain, CH2 domain, and CH3 domain or any combination of whole domains or portions thereof.
  • the polypeptides of the present invention may be fused or 75 conjugated to the above antibody portions to increase the in vivo half-life of the polypeptides or for use in immunoassays using methods known in the art.
  • the polypeptides may also be fused or conjugated to the above antibody portions to form multimers.
  • Fc portions fused to the polypeptides of the present invention can form dimers through disulfide bonding between the Fc portions. Higher multimeric forms can be made by fusing the polypeptides to portions of IgA and
  • the invention further relates to antibodies that act as agonists or antagonists of the polypeptides of the present invention.
  • the present invention includes antibodies that disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully. Included are both receptor-specific antibodies and ligand-specific antibodies. Included are receptor-specific antibodies, which do not prevent ligand binding but prevent receptor activation.
  • Receptor activation may be determined by techniques described herein or otherwise known in the art. Also include are receptor-specific antibodies which both prevent ligand binding and receptor activation. Likewise, included are neutralizing antibodies that bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies that bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor. Further included are antibodies that activate the receptor. These antibodies may act as agonists for either all or less than all of the biological activities affected by ligand-mediated receptor activation. The antibodies may be specified as agonists or antagonists for biological activities comprising specific activities disclosed herein. The above antibody agonists can be made using methods known in the art.
  • antibodies of the polypeptides of the invention can, in turn, be utilized to generate anti-idiotypic antibodies that "mimic" polypeptides of the invention using techniques well known to those skilled in the art (See, e.g. Greenspan and Bona (1989);and Nissinoff (1991).
  • antibodies which bind to and competitively inhibit polypeptide multimerization or binding of a polypeptide of the invention to ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization or binding domain and, as a consequence, bind to and neutralize polypeptide or its ligand.
  • Such neutralization anti-idiotypic antibodies can be used to bind a polypeptide of the invention or to bind its ligands/receptors, and thereby block its biological activity, 76
  • the invention also concerns a purified or isolated antibody capable of specifically binding to a mutated full length or mature polypeptide of the present invention or to a fragment or variant thereof comprising an epitope of the mutated polypeptide.
  • the present invention concerns an antibody capable of binding to a polypeptide comprising at least 10 consecutive amino acids of a polypeptide of the present invention and including at least one of the amino acids which can be encoded by the trait causing mutations.
  • Non-human animals or mammals whether wild-type or transgenic, which express a different species of a polypeptide of the present invention than the one to which antibody binding is desired, and animals which do not express a polypeptide of the present invention (i.e. a knock out animal) are particularly useful for preparing antibodies.
  • Gene knock out animals will recognize all or most of the exposed regions of a polypeptide of the present invention as foreign antigens, and therefore produce antibodies with a wider array of epitopes.
  • smaller polypeptides with only 10 to 30 amino acids may be useful in obtaining specific binding to any one of the polypeptides of the present invention.
  • the humoral immune system of animals which produce a species of a polypeptide of the present invention that resembles the antigenic sequence will preferentially recognize the differences between the animal's native polypeptide species and the antigen sequence, and produce antibodies to these unique sites in the antigen sequence.
  • Such a technique will be particularly useful in obtaining antibodies that specifically bind to any one of the polypeptides of the present invention.
  • Antibody preparations prepared according to either protocol are useful in quantitative immunoassays which determine concentrations of antigen-bearing substances in biological samples; they are also used semi-quantitatively or qualitatively to identify the presence of antigen in a biological sample.
  • the antibodies may also be used in therapeutic compositions for killing cells expressing the protein or reducing the levels of the protein in the body.
  • the antibodies of the invention may be labeled by any one of the radioactive, fluorescent or enzymatic labels known in the art.
  • the invention is also directed to a method for detecting specifically the presence of a polypeptide of the present invention according to the invention in a biological sample, said method comprising the following steps: a) obtaining a biological sample suspected of containing a polypeptide of the present invention; b) contacting the biological sample with a polyclonal or monoclonal antibody that specifically binds a polypeptide of the present invention under conditions suitable for antigen- antibody binding; and c) detecting the antigen-antibody complex formed.
  • the invention also concerns a diagnostic kit for detecting in vitro the presence of a polypeptide of the present invention in a biological sample, wherein said kit comprises: 77 a) a polyclonal or monoclonal antibody that specifically binds a polypeptide of the present invention, optionally labeled; b) a reagent allowing the detection of the antigen-antibody complexes formed, said reagent carrying optionally a label, or being able to be recognized itself by a labeled reagent, more particularly in the case when the above-mentioned monoclonal or polyclonal antibody is not labeled by itself.
  • Monoclonal antibody to epitopes of any of the peptides identified and isolated as described can be prepared from murine hybridomas according to the classical method of Rohler, G. and
  • a mouse is repetitively inoculated with a few micrograms of the selected protein or peptides derived therefrom over a period of a few weeks. The mouse is then sacrificed, and the antibody producing cells of the spleen isolated. The spleen cells are fused by means of polyethylene glycol with mouse myeloma cells, and the excess unfused cells destroyed by growth of the system on selective media comprising aminopterin (HAT media). The successfully fused cells are diluted and aliquots of the dilution placed in wells of a microtiter plate where growth of the culture is continued.
  • HAT media aminopterin
  • Antibody-producing clones are identified by detection of antibody in the supernatant fluid of the wells by immunoassay procedures, such as Elisa, as originally described by Engvall, E, Meth. Enzymol. 70:419 (1980), and derivative methods thereof. Selected positive clones can be expanded and their monoclonal antibody product harvested for use. Detailed procedures for monoclonal antibody production are described in Davis, L. et al. Basic Methods in Molecular Biology Elsevier, New York. Section 21-2.
  • Polyclonal antiserum containing antibodies to heterogenous epitopes of a single protein can be prepared by immunizing suitable animals with the expressed protein or peptides derived therefrom described above, which can be unmodified or modified to enhance immunogenicity. Effective polyclonal antibody production is affected by many factors related both to the antigen and the host species. For example, small molecules tend to be less immunogenic than others and may require the use of carriers and adjuvant. Also, host animals vary in response to site of inoculations and dose, with both inadequate or excessive doses of antigen resulting in low titer antisera. Small doses (ng level) of antigen administered at multiple infradermal sites appears to be most reliable.
  • Booster injections can be given at regular intervals, and antiserum harvested when antibody titer thereof, as determined semi-quantitatively, for example, by double immunodiffusion in agar against known concenfrations of the antigen, begins to fall. See, for example, Ouchterlony, O. et al. 78 Chap. 19 in: Handbook of Experimental Immunology D. Wier (ed) Blackwell (1973). Plateau concentration of antibody is usually in the range of 0.1 to 0.2 mg/ml of serum (about 12 DM).
  • Affinity of the antisera for the antigen is determined by preparing competitive binding curves, as described, for example, by Fisher, D, Chap. 42 in: Manual of Clinical Immunology, 2d Ed. (Rose and Friedman, Eds.) Amer. Soc. For Microbiol, Washington, D.C. (1980).
  • Antibody preparations prepared according to either protocol are useful in quantitative immunoassays which determine concenfrations of antigen-bearing substances in biological samples; they are also used semi-quantitatively or qualitatively to identify the presence of antigen in a biological sample.
  • the antibodies may also be used in therapeutic compositions for killing cells expressing the protein or reducing the levels of the protein in the body.
  • the invention features methods of screening compounds for one or more antagonists of GMG-5 or GMG-5 polypeptide fragment activity, wherein said activity is selected from but not restricted to lipid partitioning, lipid metabolism, and insulin-like activity.
  • Preferred said compound is selected from but is not restricted to small molecular weight organic or inorganic compound, protein, peptide, carbohydrate, or lipid.
  • Preferred said polypeptide fragment is GMG-5 polypeptide fragment.
  • Preferred said polypeptide fragment is GMG-5 polypeptide fragment.
  • the invention further features methods of screening compounds for said antagonist of
  • GMG-5 or GMG-5 polypeptide fragment activity comprising: a) contacting said GMG-5 polypeptide fragment with or without said compound; b) detecting a result on the basis of activity, wherein said activity is selected from but not restricted to lipid partitioning, lipid metabolism, and insulin-like activity; and c) wherein said result identifies said antagonist as specific for GMG-5 polypeptide fragment if said result with compound differs from said result for said result without compound.
  • Exemplary assay that may be used is described in Examples 4 and 18.
  • RNA Strip-EZ kit from Ambion as per manufacture's instructions.
  • Hybridization of RNA probes to RNA blots is performed Ulfrahyb hybridization solution (Ambion). Briefly, blots are prehybridized for 30 min at 58°C (low-sfrigency) or 65°C (high stringency).
  • blots are hybridized overnight (14-24 hrs), and washed 2 x 20 min at 50°C with 2x SSC/0.1% SDS (low stringency), 2 x 20 min at 58°C with lx SSC/0.1%SDS (medium stringency) and 2 x 20 min at 65°C with lx SSC/0.1%SDS (high stringency). After washings are completed blots are exposed on the phosphoimager (Molecular
  • EXAMPLE 2 In Vitro Tests of Metabolic-related Activity The activity of various preparations and various sequence variants of GMG-5 polypeptides are assessed using various in vitro assays including those provided below. These assays are also exemplary of those that can be used to develop GMG-5 polypeptide antagonists and agonists. To do that, the effect of GMG-5 polypeptides in the above assays, e.g. on leptin and/or LSR activity, in the presence of the candidate molecules would be compared with the effect of GMG-5 polypeptides in the assays in the absence of the candidate molecules. Since GMG-5 polypeptides are believed to reduce body weight in mice on a high-cafeteria diet (Example 5), these assays also serve to identify candidate treatments for reducing (or increasing) body weight.
  • liver cell lines including for example, PLC, HepG2, Hep3B (human), Hepa 1-6, BPRCL (mouse), or MCA-RH777, MCA-RH8994 (rat).
  • BPRCL mouse liver cells (ATCC Repository) are plated at a density of 300,000 cells/well in 6-well plates (day 0) in DMEM (high glucose) containing glutamine and penicillin-streptomycin (Bihain & Yen, 1992). Media is changed on day 2. On day 3, the confluent monolayers are washed once with phosphate-buffered saline (PBS, pH 7.4) (2 mL/well).
  • PBS phosphate-buffered saline
  • Cells are incubated at 37°C for 30 min with increasing concentrations of recombinant AdipoQ (AQ) or globular AdipoQ (AQ-GH) in DMEM containing 0.2% (w/v) BSA, 5 mM Hepes, 2 mM CaCl 2 , 3.7 g/L sodium bicarbonate, pH 7.5. Incubations are continued for 3 h at 37°C after addition of 10 ng/mL 125 I-mouse leptin (specific activity, 22100 cpm/ng). Monolayers are washed 2 times consecutively with PBS containing 0.2% BSA, followed by 1 wash with PBS/BSA, and then 2 times consecutively with PBS. Cells are lysed 80 with 0.1 N NaOH containing 0.24 mM EDTA. Lysates are collected into tubes, and counted in a gamma-counter.
  • AQ recombinant AdipoQ
  • AQ-GH globular AdipoQ
  • the effect of GMG-5 polypeptides on leptin transport in the brain can be determined using brain-derived cells.
  • One method that is envisioned is to use the blood/brain barrier model described by Dehouck, et al (J Neurochem 54:1798-801, 1990; hereby inco ⁇ orated herein by reference in its entirety including any figures, tables, or drawings) that uses a co-culture of brain capillary endothelial cells and astrocytes to test the effects of GMG-5 polypeptides on leptin (or other molecules) fransport via LSR or other receptors.
  • This assay would be an indicator of the potential effect of GMG-5 polypeptides on leptin fransport to the brain and could be used to screen GMG-5 polypeptide variants for their ability to modulate leptin transport through LSR or other receptors in the brain.
  • putative agonists and antagonists of the effect of GMG-5 polypeptides on leptin transport through LSR or other receptors could also be screened using this assay. Increased transport of leptin across the blood/brain barrier would presumably increase its action as a satiety factor.
  • the effect of GMG-5 polypeptides on LSR can also be determined by measuring the level of LSR expression at the cell surface by flow surface cytometry, using anti-LSR antibodies and fluorescent secondary antibodies.
  • Flow cytometry is a laser-based technology that is used to measure characteristics of biological particles. The underlying principle of flow cytometry is that light is scattered and fluorescence is emitted as light from the excitation source strikes the moving particles. This is a high through-put assay that could be easily adapted to screen GMG-5 polypeptides and variants as well as putative agonists or antagonists of GMG-5 polypeptides. Two assays are provided below.
  • the antibody, cell-line and GMG-5 polypeptide analogs would vary depending on the experiment, but a human cell-line, human anti-LSR antibody and globular APMl could be used to screen for variants, agonists, and antagonists to be used to treat humans.
  • Assay 1 :
  • Cells are pretreated with either intact GMG-5 polypeptides (or untreated) before harvesting and analysis by FACS.
  • Cells are harvested using non-enzymatic dissociation solution (Sigma), and then are incubated for 1 h at 4°C with a 1:200 dilution of anti-LSR 8 IB or an irrelevant anti-serum in PBS containing 1% (w/v) BSA. After washing twice with the same buffer, goat anti-rabbit FITC- conjugated antibody (Rockland, Gilbertsville, PA) is added to the cells, followed by a further incubation for 30 min at 4 °C. After washing, the cells are fixed in 2% formalin. Flow cytometry analysis is done on a FACSCalibur cytometer (Becton-Dickinson, Franklin Lakes, NJ). 81 Assay 2:
  • FACs buffer lx PBS/2% FBS, filter sterilized
  • the cell suspension is transferred to a 15 mL conical tube and centrifuged at 1200 ⁇ m, 4°C for 5 minutes. Supernatant is discarded and cells are resuspended in 10 mL FACs buffer chilled to 4°C.
  • a cell count is performed and the cell density adjusted with FACs buffer to a concentration of 1 x 10 6 cells/ mL.
  • One milliliter of cell suspension was added to each well of a 48 well plate for analysis. Cells are centrifuged at 1200 ⁇ m for 5 minutes at 4°C.
  • GMG-5 Polypeptides as Detected by Fluorescence Microscopy Fluorecein isothiocyanate (FITC) conjugation of GMG-5 polypeptides: Purified GMG-5 proteins at 1 mg/mL concentration are labeled with FITC using Sigma 's FluoroTag FITC conjugation kit (Stock No. FITC-1). Protocol outlined in the Sigma Handbook for small scale conjugation is followed for GMG-5 protein labeling.
  • Cell Culture C2C 12 mouse skeletal muscle cells (ATCC, Manassas, VA CRL- 1772) and
  • Hepa-1-6 mouse hepatocytes (ATCC, Manassas, VA CRL-1830) are seeded into 6 well plates at a cell density of 2xl0 5 cells per well. C2C12 and Hepa-1-6 cells are cultured according to repository's instructions for 24-48 hours prior to analysis. Assay is performed when cells were 80% confluent.
  • C2C12 and Hepa 1-6 cells are incubated in the presence/absence of antibody directed against human LSR (81B: N-terminal sequence of human LSR; does not cross react with mouse LSR and 93 A: c-terminal 82 sequence, cross reacts with mouse LSR) or an antiserum directed against gClqr (953) for 1 hour at
  • LSR antibodies are added to the media at a concentration of 2 ⁇ g/mL.
  • the anti- gClqr antiserum is added to the media at a volume of 2.5 ⁇ L undiluted serum (high concentration) or 1:100 dilution (low concentration).
  • FITC-GMG-5 polypeptide 50 nM/mL is added to each cell culture well.
  • Cells are again incubated for 1 hour at 37°C, 5% C02.
  • Cells are washed 2x with PBS, cells are scraped from well into 1 mL of PBS. Cell suspension is transferred to an eppendorf tube and centrifuged at 1000 m for 2 minutes.
  • FITC- GMG-5 polypeptide is analyzed by fluorescence microscopy under 40X magnification. This assay may be useful for identifying agents that facilitate or prevent the uptake and/or binding of GMG-5 polypeptides to cells.
  • the effect of GMG-5 protein on the lipoprotein binding, internalizing and degrading activity of LSR can also be tested. Measurement of LSR as lipoprotein receptor is described in Bihain & Yen, ((1992) Biochemistry May 19;31(19):4628-36; hereby inco ⁇ orated herein in its entirety including any drawings, tables, or figures).
  • the effect of GMG-5 protein on the lipoprotein binding, internalizing and degrading activity of LSR (or other receptors) can be compared with that of intact GMG-5 protein, with untreated cells as an additional control. This assay can also be used to screen for active and inhibitory variants of GMG-5 protein, as well as agonists and antagonists of metabolic-related activity.
  • Human liver PLC cells (ATCC Repository) are plated at a density of 300,000 cells/well in 6- well plates (day 0) in DMEM (high glucose) containing glutamine and penicillin-streptomycin (Bihain & Yen, 1992). Media is changed on day 2. On day 3, the confluent monolayers are washed once with phosphate-buffered saline (PBS, pH 7.4) (2 mL/well).
  • PBS phosphate-buffered saline
  • GMG-5 protein leads to an increased activity of LSR as a lipoprotein receptor.
  • the oleate-induced binding and uptake of LDL would be more affected by
  • GMG-5 proteinas compared to the degradation. This increased LSR activity would potentially result 83 in an enhanced clearance of triglyceride-rich lipoproteins during the postprandial state. Thus, more dietary fat would be removed through the liver, rather than being deposited in the adipose tissue.
  • This assay could be used to determine the efficiency of a compound (or agonists or antagonists) to increase or decrease LSR activity (or lipoprotein uptake, binding and degradation through other receptors), and thus affect the rate of clearance of triglyceride-rich lipoproteins.
  • C2C12 cells (murine skeletal muscle cell line; ATCC CRL 1772, Rockville, MD) are seeded sparsely (about 15-20%) in complete DMEM (w/glutamine, pen/strep, etc) + 10% FCS. Two days later they become 80-90% confluent. At this time, the media is changed to DMEM+2% horse serum to allow differentiation. The media is changed daily. Abundant myotube formation occurs after 3-4 days of being in 2% horse serum, although the exact time course of C2C12 differentiation depends on how long they have been passaged and how they have been maintained, among other things.
  • gACRP30 (1 to 2.5 ⁇ g/mL) is added the day after seeding when the cells are still in DMEM w/ 10% FCS. Two days after plating the cells (one day after gACRP30 was first added), at about 80-90% confluency, the media is changed to DMEM+2% horse serum plus gACRP30.
  • C2C12 cells are differentiated in the presence or absence of 2 ⁇ g/mL GMG-5 proteinfor 4 days.
  • oleate oxidation rates are determined by measuring conversion of l- 14 C-oleate (0.2 mM) to 14 CO 2 for 90 min. This experiment can be used to screen for active polypeptides and peptides as well as agonists and antagonists or activators and inhibitors of GMG-5 polypeptides.
  • the effect of gACRP30 on the rate of oleate oxidation can be compared in differentiated C2C12 cells (murine skeletal muscle cells; ATCC, Manassas, VA CRL- 1772) and in a hepatocyte cell line (Hepal-6; ATCC, Manassas, VA CRL-1830). Cultured cells are maintained according to manufacturer's instructions.
  • the oleate oxidation assay is performed as previously described (Muoio et al (1999) Biochem J 338;783-791). Briefly, nearly confluent myocytes are kept in low serum differentiation media (DMEM, 2.5% Horse serum) for 4 days, at which time formation of myotubes became maximal.
  • DMEM low serum differentiation media
  • Hepatocytes are kept in the same DMEM medium supplemented with 10% FCS for 2 days.
  • preincubation media MEM, 2.5% Horse serum, 3 mM glucose, 4 mM Glutamine, 25 mM Hepes, 1% FFA free BSA, 0.25 mM Oleate, 5 ⁇ g/mL gentamycin
  • 14 C-Oleic acid l ⁇ Ci mL, American Radiolabeled Chemical Lnc, St. Louis, MO
  • cells are incubated for 90 min at 37°C in the absence/presence of 2.5 ⁇ g/mL gACRP30.
  • 0.75 mL of the media is removed and assayed for 14 C-oxidation products as described below for the muscle FFA oxidation experiment.
  • Triglyceride and Protein Analysis following Oleate Oxidaiton in cultured cells Following transfer of media for oleate oxidation assay, cells are placed on ice. To determine triglyceride and protein content, cells are washed with 1 mL of lx PBS to remove residual media. To each well 300 ⁇ L of cell dissociation solution (Sigma) is added and incubated at 37°C for 10 min. Plates are tapped to loosen cells, and 0.5 mL of lx PBS was added. The cell suspension is fransferred to an eppendorf tube, each well is rinsed with an additional 0.5 mL of lx PBS, and is transferred to appropriate eppendorf tube.
  • Samples are centrifuged at 1000 ⁇ m for 10 minutes at room temperature. Supernatant is discarded and 750 ⁇ L of lx PBS/2% chaps is added to cell pellet. Cell suspension is vortexed and placed on ice for 1 hour. Samples are then centrifuged at 13000 ⁇ m for 20 min at 4°C. Supernatants are fransferred to new tube and frozen at -20°C until analyzed. Quantitative measure of triglyceride level in each sample is determined using Sigma Diagnostics GPO-TRJNDER enzymatic kit.
  • L6 Muscle cells are obtained from the European Culture Collection (Porton Down) and are used at passages 7-11. Cells are maintained in standard tissue culture medium DMEM, and glucose uptake is assessed using [ 3 H]-2-deoxyglucose (2DG) with or without GMG-5 polypeptide fragment in the presence or absence of insulin (10 '8 M) as has been previously described (Walker, P.S. et al. (1990) Glucose transport activity in L6 muscle cells is regulated by the coordinate control of subcellular glucose transporter distribution, biosynthesis, and mRNA transcription. JBC 265(3): 1516-1523; and Rilp, A. et al. (1992) Stimulation of hexose fransport by metformin in L6 muscle cells in culture.
  • mice All mice are housed individually. The mice are maintained on a high fat diet throughout each expenment.
  • the high fat diet (cafeteria diet; D12331 from Research Diets, Lnc.) has the following composition: protein kcal% 16, sucrose kcal% 26, and fat kcal% 58.
  • the fat is pnma ⁇ ly composed of coconut oil, hydrogenated.
  • mice After the mice are fed a high fat diet for 6 days, micro-osmotic pumps are inserted using lsoflurane anesthesia, and are used to provide full-length GMG-5 polypeptides, GMG-5 polypeptide fragments, saline, and an irrelevant peptide to the mice subcutaneously (s.c.) for 18 days.
  • GMG-5 polypeptides are provided at doses of 100, 50, 25, and 2.5 ⁇ g/day and the irrelevant peptide is provided at 10 ⁇ g/day.
  • Body weight is measured on the first, third and fifth day of the high fat diet, and then daily after the start of treatment. Final blood samples are taken by cardiac puncture and are used to determine tnglycende (TG), total cholesterol (TC), glucose, leptin, and insulm levels. The amount of food consumed per day is also determined for each group.
  • GMG-5 polypeptides Tests of the efficacy of GMG-5 polypeptides in humans are performed in accordance with a physician's recommendations and with established guidelines. The parameters tested in mice are also tested in humans (e g. food intake, weight, TG, TC, glucose, insulm, leptin, FFA). It is expected that the physiological factors would show changes over the short term. Changes in weight gain might require a longer penod of time. Ln addition, the diet would need to be carefully monitored. GMG-5 polypeptides, preferably GMG-5 polypeptides compnsing the Clq homology domain, would be given in daily doses of about 6 mg protein per 70 kg person or about 10 mg per day. Other doses would also be tested, for instance 1 mg or 5 mg per day up to 20 mg, 50 mg, or 100 mg per day.
  • EXAMPLE 5 Tests of Metabolic-related Activity in a Murine Lipoatrophic Diabetes Model
  • leptin was reported to reverse insulin resistance and diabetes mellitus in mice with congenital hpodysfrophy (Shimomura et al. Nature 401 : 73-76 (1999); hereby inco ⁇ orated herein in its entirety including any drawings, figures, or tables).
  • Leptin was found to be less effective in a different hpodystrophic mouse model of lipoatrophic diabetes (Gavnlova et al Nature 403: 850 (2000); hereby inco ⁇ orated herein m its entirety including any drawings, figures, or tables).
  • the instant invention encompasses the use of GMG-5 polypeptides for reducing the insulin resistance and hyperglycaemia in this model either alone or in combination with leptin, the leptin peptide (US provisional application No 60/155,506), or other compounds.
  • Assays include that descnbed previously in Gavnlova et al. ((2000) Diabetes Nov,49(l l):1910-6; (2000) Nature Feb 24;403(6772):850) using A-ZEP/F-1 mice, except that GMG-5 polypeptides would be administered 86 using the methods previously described in Example 3 (or Examples 6-8).
  • the glucose and insulin levels of the mice would be tested, and the food intake and liver weight monitored, as well as other factors, such as leptin, FFA, and TG levels, typically measured in our experiments (see Example 3, above, or Examples 6-8).
  • mice used in this experiment are fasted for 2 hours prior to the experiment after which a baseline blood sample is taken. All blood samples are taken from the tail using EDTA coated capillary tubes (50 ⁇ L each time point).
  • a GMG-5 polypeptide is injected i.p. in 100 ⁇ L saline.
  • the same dose 25 ⁇ g/mL in lOO ⁇ L is again injected at 45 min and at 1 hr 45 min.
  • Control animals are injected with saline (3xl00 ⁇ L). Untreated and freated animals are handled in an alternating mode.
  • Plasma samples are taken in hourly intervals, and are immediately put on ice. Plasma is prepared by centrifugation following each time point. Plasma is kept at -20°C and free fatty acids (FFA), triglycerides (TG) and glucose are determined within 24 hours using standard test kits (Sigma and Wako). Due to the limited amount of plasma available, glucose is determined in duplicate using pooled samples. For each time point, equal volumes of plasma from all 8 animals per treatment group are pooled.
  • FFA free fatty acids
  • TG triglycerides
  • glucose is determined in duplicate using pooled samples. For each time point, equal volumes of plasma from all 8 animals per treatment group are pooled.
  • EXAMPLE 7 Effect of GMG-5 Polypeptides on Plasma Leptin and Insulin in C57 BL/6 Mice The effect of GMG-5 polypeptides on plasma leptin and insulm levels during postprandial lipemia (PPL) in normal C57BL6/J mice is tested.
  • the experimental procedure is the same as that described in Example 6, except that blood was drawn only at 0, 2 and 4 hours to allow for greater blood samples needed for the determination of leptin and insulin by RIA. Briefly, 16 mice are fasted for 2 hours prior to the experiment after which a baseline blood sample is taken. All blood samples are taken from the tail using EDTA coated capillary tubes (100 ⁇ L each time point).
  • 25 ⁇ g of a GMG-5 polypeptide is injected i.p. in 100 ⁇ L saline.
  • the same dose (25 ⁇ g in lOO ⁇ L) is again injected at 45 min and at 1 hr 45 min (freated group).
  • Confrol animals are injected with saline (3xl00 ⁇ L). Untreated and treated animals are handled in an alternating mode.
  • Plasma samples are immediately put on ice and plasma is prepared by centrifugation 87 following each time point. Plasma is kept at -20°C and free fatty acids (FFA) are determined within
  • EXAMPLE 8 Effect of GMG-5 Polypeptides on Plasma FFA. TG and Glucose in C57 BL/6 Mice The effect of GMG-5 polypeptides on plasma FFA, TG, glucose, leptin and insulin levels during postprandial lipemia (PPL) in normal C57BL6/J mice has been described. Weight loss resulting from GMG-5 polypeptides (2.5 ⁇ g/day) given to normal C57BL6/J mice on a high fat diet has also been shown (Example 3).
  • mice re fasted for 2 hours prior to the experiment after which a baseline blood sample is taken. All blood samples are taken from the tail using EDTA coated capillary tubes (50 ⁇ L each time point). At time
  • mice are injected 25 ⁇ g of a GMG-5 polypeptide i.p. in lOO ⁇ L saline.
  • the same dose 25 ⁇ g in lOO ⁇ L is again injected at 45 min and at
  • a second treatment group receives 3 times 50 ⁇ g GMG-5 polypeptide at the same intervals.
  • Control animals are injected with saline (3xl00 ⁇ L). Untreated and treated animals are handled in an alternating mode.
  • Plasma samples are immediately put on ice. Plasma is prepared by centrifugation following each time point. Plasma is kept at -20 °C and free fatty acids (FFA), triglycerides (TG) and glucose are determined within 24 hours using standard test kits (Sigma and Wako).
  • FFA free fatty acids
  • TG triglycerides
  • glucose are determined within 24 hours using standard test kits (Sigma and Wako).
  • mice plasma free fatty acids increase after intragastric adminisfration of a high fat sucrose test meal. These free fatty acids are mostly produced by the activity of lipolytic enzymes t.e. lipoprotein lipase (LPL) and hepatic lipase (HL). In this species, these enzymes are found in significant amounts both bound to endothelium and freely circulating in plasma.
  • LPL lipoprotein lipase
  • HL hepatic lipase
  • Another source of plasma free fatty acids is hormone sensitive lipase (HSL) that releases free fatty acids from adipose tissue after ⁇ -adrenergic stimulation.
  • HSL hormone sensitive lipase
  • mice are injected with epinephrine.
  • mice Two groups of mice are given epinephrine (5 ⁇ g) by intraperitoneal injection.
  • a freated group is injected with a GMG-5 polypeptide (25 ⁇ g) one hour before and again together with epinephrine, while control animals receive saline.
  • Plasma is isolated and free fatty acids and glucose 88 are measured as described above (Example 8).
  • Muscles are rinsed for 30 min in incubation media with oxygenation. The muscles are then fransferred to fresh media (1.5 mL) and incubated at 30°C in the presence of l ⁇ Ci/mL [1- 14 C] oleic acid (American Radiolabeled Chemicals). The incubation vials containing this media are sealed with a rubber septum from which a center well carrying a piece of Whatman paper (1.5 cm x 11.5 cm) is suspended.
  • the muscle is removed from the medium, and an aliquot of 0.5 mL medium is also removed.
  • the vials are closed again and 1 mL of 35% perchloric acid is injected with a syringe into the media by piercing through the rubber septum.
  • the C0 2 released from the acidified media is collected by the Solvable in the center well.
  • the Whatman paper is removed from the center well and placed in scintillation vials containing 15 mL of 89 scintillation fluid (HionicFlour, Packard Instruments, Meriden, CT).
  • the amount of 14 C radioactivity is quantitated by liquid scintillation counting.
  • the rate of oleate oxidation is expressed as nmol oleate produced in 90min/g muscle.
  • these proteins are added to the media at a final concentration of 2.5 ⁇ g/mL and maintained in the media throughout the procedure.
  • the hindlimb muscle and liver triglyceride content is measured after the GMG-5 polypeptide treatment of mice. Hind limb muscles as well as liver samples are removed from treated and unfreated animals and the triglyceride and free fatty acid concentration is determined following a standard lipid extraction method
  • mice Two groups of mice are intravenously (tail vein) injected with 30 ⁇ L bolus of Infralipid-20% (Clintec) to generate a sudden rise in plasma FFAs, thus by-passing intestinal abso ⁇ tion.
  • Infralipid is an intravenous fat emulsion used in nutritional therapy.
  • a freated group (GMG-5 polypeptide- treated) is injected with a GMG-5 polypeptide (25 ⁇ g) at 30 and 60 minutes before Infralipid is given, while control animals ( ⁇ confrol) received saline. Plasma is isolated and FFAs are measured as described previously. The effect of GMG-5 polypeptides on the decay in plasma FFAs following the peak induced by Infralipid injection is then monitored.
  • EXAMPLE 13 In vitro glucose uptake by muscle cells L6 Muscle cells are obtained from the European Culture Collection (Porton Down) and are used at passages 7-11. Cells are maintained in standard tissue culture medium DMEM, and glucose uptake is assessed using [.sup.3 H]-2-deoxyglucose (2DG) with or without GMG-5 polypeptides in the presence or absence of insulin (10.sup.-8 M) as has been previously described (Walker, P.S. et al. (1990) Glucose fransport activity in L6 muscle cells is regulated by the coordinate control of subcellular glucose transporter distribution, biosynthesis, and mRNA transcription. JBC
  • mice The homozygous animals, C57 BL/RsJ-db/db mice developed by Jackson Laboratory, US, are obese, hyperglycemic, hyperinsulinemic and insulin resistant (J. Clin. Invest, (1990) 85: 962-967), whereas heterozygous are lean and normoglycemic.
  • db/db model mouse progressively develops insulinopenia with age, a feature commonly observed in late stages of human type II diabetes when blood sugar levels are insufficiently controlled. The state of pancreas and its course vary according to the models. Since this model resembles that of type II diabetes mellitus, the compounds of the present invention are tested for blood sugar and triglycerides lowering activities.
  • fa/fa rats are severely obese, hyperinsulinemic, and insulin resistant (Coleman, Diabetes 31 : 1 , 1982; E. Shafrir, in Diabetes Mellitus; H. Rifkin and D. Porte, Jr. Eds. (Elsevier Science Publishing Co, Inc., New York, ed. 4, 1990), pp. 299-340), and the fa/fa mutation may be the rat equivalent of the murine db mutation (Friedman et al. Cell 69:217-220, 1992; Truett et al, Proc. Natl. Acad. Sci. USA 88:7806, 1991).
  • Tubby mice are characterized by obesity, moderate insulin resistance and hyperinsulinemia without significant hyperglycemia (Coleman et al, J. Heredity 81:424, 1990).
  • leptin was reported to reverse insulin resistance and diabetes mellitus in mice with congenital lipodysfrophy (Shimomura et al. Nature 401 : 73-76 (1999).
  • Leptin is found to be less effective in a different lipodystrophic mouse model of lipoatrophic diabetes (Grajova et al Nature 403: 850 (2000); hereby inco ⁇ orated herein in its entirety including any drawings, figures, or tables).
  • STZ streptozotocin
  • the monosodium glutamate (MSG) model for chemically-induced obesity (Olney, Science 164:719,
  • a non-chemical, non-genetic model for induction of obesity includes feeding rodents a high fat/high carbohydrate (cafeteria diet) diet ad libitum.
  • the instant invention encompasses the use of GMG-5 polypeptides for reducing the insulin resistance and hyperglycemia in any or all of the above rodent diabetes models or in humans with
  • the GSSP4 polypeptides may, if desired, be associated with other compatible pharmacologically-active antidiabetic agents such as insulin, leptin (US provisional application No 60/155,506), or troglitazone , either alone or in combination.
  • Assays include that described previously in Gavrilova et al. ((2000) Diabetes Nov;49(l 1): 1910-6; (2000) Nature Feb 24;403(6772):850) using A-ZIP/F-1 mice, except that GSSP4 polypeptides are administered infraperotineally, subcutaneously, intramuscularly or intravenously.
  • mice In Vivo Assay for Anti-hypergrycemic Activity of GMG-5 polypeptides
  • db/db male, 7-9 weeks old
  • mice/cage 7-9 mice/cage
  • Purina rodent chow and water ad libitum Prior to freatment, blood is collected from the tail vein of each animal and blood glucose concenfrations are determined using One Touch BasicGlucose Monitor System (Lifescan).
  • mice that have plasma glucose levels between 250 to 500 mg/dl are used.
  • Each treatment group consists of seven mice that are distributed so that the mean glucose levels are equivalent in each group at the start of the study, db/db mice are dosed by micro-osmotic pumps, inserted using isoflurane anesthesia, to provide GMG-5 polypeptides, saline, and an irrelevant peptide to the mice subcutaneously (s.c).
  • Blood is sampled from the tail vein hourly for 4 hours and at 24, 30 h post-dosing and analyzed for blood glucose concentrations. Food is withdrawn from 0-4 h post dosing and reinfroduced thereafter. Individual body weights and mean food consumption (each cage) are also measured after 24 h.
  • GMG-5 polypeptides or vehicle is administered through the jugular vein after complete recovery and for the following two days.
  • hyperinsulinemic-euglycemic clamps are performed. Rodents are placed in restrainers and a bolus of 4 .mu Ci [3-.sup.3 H] glucose (NEN) is administered, followed by a continuous infusion of the tracer at a dose of 0.2 .mu.Ci/min (20 .mu.l/min).
  • 3 blood samples 0.3 ml each
  • An insulin infusion is then started (5 mU/kg/min), and 100 .mu.l blood samples are taken every 10 min. to monitor plasma glucose.
  • a 30% glucose solution is infused using a second pump based on the plasma glucose levels in order to reach and maintain euglycemia.
  • 3 additional blood samples are obtained for measurements of glucose, [3- .sup.3 H] glucose and insulin (100-120 min.).
  • a higher dose of insulm 25 mU/kg/min.
  • glucose infusion rates are adjusted for the second euglycemic clamp and blood samples are taken at min. 220-240.
  • Glucose specific activity is determined in deproteinized plasma and the calculations of Rd and hepatic glucose output (HGO) are made, as described (Lang et al. Endocrinology 130:43, 1992). Plasma insulin levels at basal period and after 5 and 25 mU/kg/min. infusions are then determined and compared between GMG-5 treated and vehicle treated rodents. Insulin regulation of glucose homeostasis has two major components; stimulation of peripheral glucose uptake and suppression of hepatic glucose output. Using tracer studies in the glucose clamps, it is possible to determine which portion of the insulm response is affected by the GMG-5 polypeptides.
  • Dipeptidyl peptidase cleavage of GMG-5 polypeptide fragment is determined by ELISA using a monoclonal antibody that specifically binds to intact GMG-5 polypeptide fragment but not to said GMG-5 polypeptide fragment from which the N-terminal dipeptide EP has been removed.
  • Dipetidyl peptidase is selected from but not restricted to human plasma comprised of dipeptidyl peptidase, soluble human CD26, or soluble human Attractin.
  • GMG-5 (5 ⁇ l, 100 frnol) is added to plasma samples (95 ⁇ l) and incubated for 1 h at 37°C, after which the amount of GMG-5 polypeptide fragment that remains undegraded is determined by enzyme-linked immunosorbent assay (ELISA) using a monoclonal antibody specific for intact GMG-5 polypeptide fragment.
  • ELISA enzyme-linked immunosorbent assay
  • GMG-5 polypeptide fragment (5 ⁇ l, 100 frnol) is added to heat-inactivated plasma (95 ⁇ l), which contains 0.01 mmol/1 valine-pyrrolidide and 500 REE/ml aprotinin.
  • the extent of dipeptidyl peptidase cleavage of GMG-5 polypeptide fragment is calculated relative to the reference and expressed as a percentage.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Obesity (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Diabetes (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention s'inscrit dans le domaine de la recherche sur le métabolisme. Les troubles du métabolisme, tels que l'obésité, constituent un problème de santé public grave et étendu. On a identifié des polypeptides GMG-5 qui se révèlent être bénéfiques pour le traitement des troubles du métabolisme. Ces composés permettent de réduire la masse corporelle et ils permettent de traiter les maladies et les troubles liés au métabolisme, parmi lesquels l'hyperlipidémies, l'athérosclérose, les diabètes et l'hypertension.
PCT/IB2002/004806 2001-12-19 2002-10-29 Polypeptides et polynucleotides gmg-5 et utilisations de ceux-ci WO2003051911A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002339697A AU2002339697A1 (en) 2001-12-19 2002-10-29 Gmg-5 polynucleotides and polypeptides and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US34344001P 2001-12-19 2001-12-19
US60/343,440 2001-12-19

Publications (2)

Publication Number Publication Date
WO2003051911A2 true WO2003051911A2 (fr) 2003-06-26
WO2003051911A3 WO2003051911A3 (fr) 2004-06-17

Family

ID=23346124

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2002/004806 WO2003051911A2 (fr) 2001-12-19 2002-10-29 Polypeptides et polynucleotides gmg-5 et utilisations de ceux-ci

Country Status (2)

Country Link
AU (1) AU2002339697A1 (fr)
WO (1) WO2003051911A2 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064718A2 (fr) * 2000-03-02 2001-09-07 Lexicon Genetics Incorporated Nouvelles proteines humaines et polynucleotides codant pour lesdites proteines
WO2002055694A2 (fr) * 2001-01-16 2002-07-18 Genset Sa Polynucleotides et polypeptides de genes metaboliques et leurs utilisations

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001055355A1 (fr) * 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Acides nucleiques, proteines et anticorps

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064718A2 (fr) * 2000-03-02 2001-09-07 Lexicon Genetics Incorporated Nouvelles proteines humaines et polynucleotides codant pour lesdites proteines
WO2002055694A2 (fr) * 2001-01-16 2002-07-18 Genset Sa Polynucleotides et polypeptides de genes metaboliques et leurs utilisations

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE GSN [Online] 7 January 2002 (2002-01-07) ROSEN C.A. ET AL.: "Human polynucleotide SEQ ID NO:23" Database accession no. AAI99525 XP002244348 -& DATABASE GSP [Online] 7 January 2002 (2002-01-07) ROSEN C.A. ET AL.: "Human polypeptide SEQ ID NO:43" Database accession no. AAM99927 XP002244349 -& WO 01 55173 A (HUMAN GENOME SCIENCES INC) 2 August 2001 (2001-08-02) *
DATABASE SWALL [Online] 1 March 2001 (2001-03-01) WATANABE K. ET AL.: "Hypothetical protein FLJ22569" Database accession no. Q9H667 XP002244351 *

Also Published As

Publication number Publication date
AU2002339697A8 (en) 2003-06-30
AU2002339697A1 (en) 2003-06-30
WO2003051911A3 (fr) 2004-06-17

Similar Documents

Publication Publication Date Title
US6867189B2 (en) Use of adipsin/complement factor D in the treatment of metabolic related disorders
AU2002321768B2 (en) GMG-2 polynucleotides and polypeptides and uses thereof
AU2002321768A1 (en) GMG-2 polynucleotides and polypeptides and uses thereof
WO2003010197A2 (fr) Polynucleotides et polypeptides gmg-1 et leurs utilisations
US7402393B2 (en) Methods of screening for antagonists of APM1/GZIP
AU2002339686B2 (en) NGZIPA, NGZIPD, PGZIPA, and PGZIPD polynucleotides and polypeptides and uses thereof
US7348406B2 (en) Metabolic gene polynucleotides and polypeptides and uses thereof
AU2002339686A1 (en) NGZIPA, NGZIPD, PGZIPA, and PGZIPD polynucleotides and polypeptides and uses thereof
US7326678B2 (en) GMG-3 GMG-4 and GMG-6 polynucleotides and polypeptides and uses thereof
AU2002249521A1 (en) Discriminative nucleic acid analysis using clone sequence signatures
US20060058507A1 (en) Krib polynucleotides and polypeptides and uses thereof in the treatment of metabolic disorders
US20060258567A1 (en) Xcrf polynucleotides and polypeptides and uses thereof
AU2002249529A1 (en) GMG-3, GMG-4, and GMG-6 polynucleotides and polypeptides and uses thereof
WO2003051911A2 (fr) Polypeptides et polynucleotides gmg-5 et utilisations de ceux-ci
US20040235709A1 (en) Gssp3 polynucleotides and polypeptides and uses thereof
US20050075285A1 (en) Gssp4 polynucleotides and polypeptides and uses thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP