US20020119138A1 - Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha) - Google Patents

Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha) Download PDF

Info

Publication number
US20020119138A1
US20020119138A1 US09/794,715 US79471501A US2002119138A1 US 20020119138 A1 US20020119138 A1 US 20020119138A1 US 79471501 A US79471501 A US 79471501A US 2002119138 A1 US2002119138 A1 US 2002119138A1
Authority
US
United States
Prior art keywords
dgat2α
leu
gly
val
phe
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US09/794,715
Inventor
Sylvaine Cases
Robert Farese
Scot Stone
Ping Zhou
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Priority to US09/794,715 priority Critical patent/US20020119138A1/en
Assigned to REGENT OF THE UNIVERSITY OF CALIFORNIA, THE reassignment REGENT OF THE UNIVERSITY OF CALIFORNIA, THE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FARESE, ROBERT V., CASES, SYLVAINE, STONE, SCOT, ZHOU, PING
Priority to US10/046,924 priority patent/US7045326B2/en
Priority to EP02780736A priority patent/EP1373483A4/en
Priority to PCT/US2002/005474 priority patent/WO2002068595A2/en
Priority to AU2002306573A priority patent/AU2002306573A1/en
Publication of US20020119138A1 publication Critical patent/US20020119138A1/en
Priority to US10/286,581 priority patent/US20030161831A1/en
Priority to US10/446,441 priority patent/US7862819B2/en
Priority to US10/945,738 priority patent/US20050106697A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: J. DAVID GLADSTONE INSTITUTES
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/1029Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y203/00Acyltransferases (2.3)
    • C12Y203/01Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • C12Y203/0102Diacylglycerol O-acyltransferase (2.3.1.20)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y203/00Acyltransferases (2.3)
    • C12Y203/01Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • C12Y203/010222-Acylglycerol O-acyltransferase (2.3.1.22)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Nucleic acid compositions encoding mammalian DGAT2α polypeptide products with diglyceride acyltransferase activity, as well as the mammalian DGAT2α polypeptide products encoded thereby and methods for producing the same, are provided. The subject DGAT2α polypeptide and nucleic acid compositions find use in a variety of applications, including research, diagnostic, and therapeutic agent screening applications, as well as in treatment therapies and in the production of triacylglycerols.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • Pursuant to 35 U.S.C. § 119(e), this application claims priority to the filing date of the U.S. Provisional Patent Application Serial No. 60/______ filed Feb. 23, 2001 and bearing the title “DIACYLGLYCEROL O-ACYLTRANSFERASE 2α (DGAT2α) (Attorney Docket No. 6510-240PRV), the disclosure of which is herein incorporated by reference.[0001]
  • INTRODUCTION
  • 1. Field of the Invention [0002]
  • The field of the invention is enzymes, particularly acyltransferases. Background of the Invention Diacylglycerol O-Acyltransferase (EC 2.3.1.20), also known as diglyceride acyltransferase or DGAT, is a critical enzyme in triacylglycerol synthesis. Triacylglycerols are quantitatively the most important storage form of energy for eukaryotic cells. DGAT catalyzes the rate-limiting and terminal step in triacylglycerol synthesis using diacylglycerol and fatty acyl CoA as substrates. As such, DGAT plays a fundamental role in the metabolism of cellular diacylglycerol and is important in higher eukaryotes for intestinal fat absorption, lipoprotein assembly, fat storage in adipocytes, milk production and possibly egg production and sperm maturation. [0003]
  • Because of its central role in a variety of different processes, there is much interest in the identification of polynucleotides encoding proteins having DGAT activity, as well as the proteins encoded thereby. [0004]
  • 2. Relevant Literature [0005]
  • Of particular interest are: U.S. Pat. No. 6,100,077; and PCT Published Application Nos. WO 98/55631; WO 99/67268; WO 00/01713; WO 99/67403; WO 00/32793; WO 00/32756; WO 00/36114; WO 00/60095; WO 00/66749. [0006]
  • Also of interest are: Smith et al., Nat. Genet. 2000 (25), 87-90).Cases et al., “Identification of a gene encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacylglycerol synthesis,” Proc. Natl. Acad. Sci. USA (October 1998) 95:13018-13023; and Oelkers et al., “Characterization of Two Human Genes Encoding Acyl Coenzyme A: Cholesterol Acyltransferase-Related Enzymes,” J. Biol. Chem. (Oct. 9, 1998) 273:26765-71. [0007]
  • References describing the role DGAT plays in various biological processes include: Bell & Coleman, “Enzymes of Glycerolipid Synthesis in Eukaryotes,” Annu. Rev. Biochem. (1980) 49: 459-487; Lehner & Kuksis, “Biosynthesis of Triacylglycerols,” Prog. Lipid Res. (1996) 35: 169-201; Brindley, Biochemistry of Lipids, Lipoproteins and Membranes (eds. Vance & Vance)(Elsevier, Amsterdam)(1991) pp 171-203; Haagsman & Van Golde, “Synthesis and Secretion of Very Low Density Lipoproteins by Isolated Rat Hepatocytes in Suspension: Role of Diacylglycerol Acyltransferase,” Arch. Biochem. Biophys. (1981) 208:395-402; Coleman & Bell, “Triacylglycerol Synthesis in Isolated Fat Cells. Studies on the Microsomal Diacylglycerol Acyltransferase Activity Using Ethanol-Dispersed Diacylglycerols,” J. Biol. Chem. (1976) 251:4537-4543. [0008]
  • SUMMARY OF THE INVENTION
  • Nucleic acid compositions encoding polypeptide products with diglyceride acyltransferase activity, as well as the polypeptide products encoded thereby, i.e., mammalian DGAT2α polypeptide products, and methods for producing the same, are provided. Also provided are: methods and compositions for modulating DGAT2α activity; DGAT2α transgenic cells, animals and plants, as well as methods for their preparation; and methods for making triglycerides and triglyceride compositions, as well as the compositions produced by these methods. The subject methods and compositions find use in a variety of different applications, including research, medicine, agriculture and industry applications.[0009]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 provides a graphical representation of the results obtained from a pulse assay that demonstrates the existence of mouse DGAT2α. [0010]
  • FIG. 2 provides a hydrophobicity plot of mouse DGAT2α. [0011]
  • FIGS. 3A to [0012] 3C provide graphical results of various mouse DGAT2α activity assays.
  • FIG. 4 provides the expression profile for mouse DGAT2α. [0013]
  • FIG. 5 provides the results of an assay showing that mouse DGAT2α expression increases during 3T3-Lladipocyte differentiation. [0014]
  • FIGS. 6A and 6B provide the amino acid and nucleic acid sequences of mouse DGAT2α. [0015]
  • FIGS. 7A and 7B provide the amino acid and nucleic acid sequences of human DGAT2α. [0016]
  • FIG. 8 provides the amino acid and nucleic acid sequences of various mouse and human DGAT2α homologs.[0017]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Nucleic acid compositions encoding polypeptide products with diglyceride acyltransferase activity, as well as the polypeptide products encoded thereby, i.e., mammalian DGAT2α, and methods for producing the same, are provided. Also provided are: methods and compositions for modulating DGAT2α activity, e.g. in the treatment of disease conditions associated with DGAT2α activity, including obesity; DGAT2α transgenic cells, animals, plants and fungi, and methods for their preparation, e.g. for use in research, food production, industrial feedstock production, etc.; and methods for making triglycerides and triglyceride compositions, e.g. oils. The methods and compositions of the subject invention find use in a variety of different applications and fields, including research, medicine, agriculture and industry. [0018]
  • Before the subject invention is further described, it is to be understood that the invention is not limited to the particular embodiments of the invention described below, as variations of the particular embodiments may be made and still fall within the scope of the appended claims. It is also to be understood that the terminology employed is for the purpose of describing particular embodiments, and is not intended to be limiting. Instead, the scope of the present invention will be established by the appended claims. [0019]
  • In this specification and the appended claims, the singular forms “a,” “an,” and “the”include plural reference unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. [0020]
  • NUCLEIC ACID COMPOSITIONS
  • Nucleic acid compositions encoding polypeptide products, as well as fragments thereof, having diglyceride acetyltransferase activity are provided. Specifically, nucleic acid compositions encoding mammalian, e.g., human, mouse, etc., DGAT2α polypeptides having diglyceride acyltransferase activity are provided. By nucleic acid composition is meant a composition comprising a sequence of DNA having an open reading frame that encodes a DGAT2α polypeptide, i.e. a gene or genomic region encoding a polypeptide having diglyceride acyltransferase activity, and is capable, under appropriate conditions, of being expressed as a DGAT2α polypeptide. Also encompassed in this term are nucleic acids that are homologous or substantially similar or identical to the nucleic acids encoding DGAT2α polypeptides or proteins. Thus, the subject invention provides nucleic acids encoding mammalian DGAT2α, such as nucleic acids encoding human DGAT2α and homologs thereof and mouse DGAT2α and homologs thereof. The coding sequence of the human DGAT2α genomic sequence, i.e. the human cDNA encoding the human DGAT2α enzyme, includes or comprises a nucleic acid sequence substantially the same as or identical to that identified as SEQ ID NO:01, infra. The coding sequence of the mouse DGAT2α genomic sequence, i.e., the mouse cDNA encoding the mouse DGAT2α enzyme, includes or comprises a nucleic acid substantially the same as or identical to the sequence identified as SEQ ID NO:03, infra. [0021]
  • The source of homologous nucleic acids to those specifically listed above may be any species, including both animal and plant species, e.g., primate species, particularly human; rodents, such as rats and mice, canines, felines, bovines, ovines, equines, yeast, nematodes, etc. Between mammalian species, e.g., human and mouse, homologs have substantial sequence similarity, e.g. at least 75% sequence identity, usually at least 90%, more usually at least 95% between nucleotide sequences. Sequence similarity is calculated based on a reference sequence, which may be a subset of a larger sequence, such as a conserved motif, coding region, flanking region, etc. A reference sequence will usually be at least about 18 nt long, more usually at least about 30 nt long, and may extend to the complete sequence that is being compared. Algorithms for sequence analysis are known in the art, such as BLAST, described in Altschul et al. (1990), [0022] J. Mol. Biol. 215:403-10. Unless specified otherwise, all sequence identity values provided herein are determined using GCG (Genetics Computer Group, Wisconsin Package, Standard Settings, gap creation penalty 3.0, gap extension penalty 0.1). The sequences provided herein are essential for recognizing DGAT2α—related and homologous polynucleotides in database searches. Specific DGAT2α homologues of interest are provide in FIG. 8, i.e., SEQ ID NOs. 05, 07, 09, 11, 13 and 15.
  • Also provided are nucleic acids that hybridize to the above described specific nucleic acids, e.g., those nucleic acids having a sequence of SEQ ID NO:01 or 03, under stringent conditions. An example of stringent hybridization conditions is hybridization at 50° C. or higher and 0.1×SSC (15 mM sodium chloride/1.5 mM sodium citrate). Another example of stringent hybridization conditions is overnight incubation at 42° C. in a solution: 50% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 μg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0.1×SSC at about 65° C. Stringent hybridization conditions are hybridization conditions that are at least as stringent as the above representative conditions. Other stringent hybridization conditions are known in the art and may also be employed to identify nucleic acids of this particular embodiment of the invention. [0023]
  • Nucleic acids encoding the DGAT2α proteins and DGAT2α polypeptides of the subject invention may be cDNAs or genomic DNAs, i.e. portions of chromosomes that include both introns and exons, as well as promoter regions, etc., as well as fragments thereof. The term “DGAT2α—gene” shall be intended to mean the open reading frame encoding specific DGAT2α proteins and polypeptides, and DGAT2α introns, as well as adjacent 5′ and 3′ non-coding nucleotide sequences involved in the regulation of expression, up to about 20 kb beyond the coding region, but possibly further in either direction. The gene may be introduced into an appropriate vector for extrachromosomal maintenance or for integration into a host genome. [0024]
  • The term “cDNA” as used herein is intended to include all nucleic acids that share the arrangement of sequence elements found in native mature mRNA species, where sequence elements are exons and 3′ and 5′ non-coding regions. Normally mRNA species have contiguous exons, with the intervening introns, when present, being removed by nuclear RNA splicing, to create a continuous open reading frame encoding a DGAT2α protein. [0025]
  • A genomic sequence of interest comprises the nucleic acid present between the initiation codon and the stop codon, as defined in the listed sequences, including all of the introns that are normally present in a native chromosome. It may further include the 3′ and 5′ untranslated regions found in the mature mRNA. It may further include specific transcriptional and translational regulatory sequences, such as promoters, enhancers, etc., including about 1 kb, but possibly more, of flanking genomic DNA at either the 5′ or 3′ end of the transcribed region. The genomic DNA may be isolated as a fragment of 100 kbp or smaller; and substantially free of flanking chromosomal sequence. The genomic DNA flanking the coding region, either 3′ or 5′, or internal regulatory sequences as sometimes found in introns, contains sequences required for proper tissue and stage specific expression. [0026]
  • The nucleic acid compositions of the subject invention may encode all or a part of the subject DGAT2α proteins and polypeptides, described in greater detail infra. Double or single stranded fragments may be obtained from the DNA sequence by chemically synthesizing oligonucleotides in accordance with conventional methods, by restriction enzyme digestion, by PCR amplification, etc. For the most part, DNA fragments will be of at least 15 nt, usually at least 18 nt or 25 nt, and may be at least about 50 nt. [0027]
  • The DGAT2α—nucleic acids or genes of the subject invention are isolated and obtained in substantial purity, generally as other than an intact chromosome. Usually, the DNA will be obtained substantially free of other nucleic acid sequences that do not include a DGAT2α sequence or fragment thereof, generally being at least about 50%, usually at least about 90% pure and are typically “recombinant”, i.e. flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome. [0028]
  • In addition to the plurality of uses described in greater detail in following sections, the subject nucleic acid compositions find use in the preparation of all or a portion of the DGAT2α polypeptides, as described below. [0029]
  • POLYPEPTIDE COMPOSITIONS
  • Also provided by the subject invention are DGAT2α polypeptides having diglyceride acyltransferase activity, i.e., capable of catalyzing the acylation of diacylglycerol. The term polyeptide composition as used herein refers to both full length proteins as well [0030] ps portions or fragments thereof. Also included in this term are variations of the naturally occurring proteins, where such variations are homologous or substantially similar to the naturally occurring protein, as described in greater detail below, be the naturally occurring protein the human protein, mouse protein, or protein from some other mammalian species which naturally expresses a DGAT2α enzyme. In the following description of the subject invention, the term DGAT2α is used to refer not only to the human form of the enzyme, but also to homologs thereof expressed in non-human mammalian species.
  • The subject DGAT2α proteins are, in their natural environment, trans-membrane proteins. The subject proteins are characterized by the presence of at least one potential N-linked glycosylation site, at least one potential tyrosine phosphorylation site, and multiple hydrophobic domains, including 4 to 12, e.g., 6, hydrophobic domains capable of serving as trans-membrane regions. The proteins range in length from about 300 to 500, usually from about 325 to 475 and more usually from about 350 to 425 amino acid residues, and the projected molecular weight of the subject proteins based solely on the number of amino acid residues in the protein ranges from about 35 to 55, usually from about 37.5 to 47.5 and more usually from about 40 to 45 kDa, where the actual molecular weight may vary depending on the amount of glycolsylation of the protein and the apparent molecular weight may be considerably less because of SDS binding on gels. [0031]
  • The amino acid sequences of the subject proteins are characterized by having substantially no homology to the known DGAT enzymes. More specifically, the subject human DGAT2α enzymes have substantially no homology to the human DGAT enzyme described in Cases et al., “Identification of a gene encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacylglycerol synthesis,” Proc. Natl. Acad. Sci. U.S.A. 95 (22), 13018-13023 (1998). Likewise, the subject mouse DGAT2α enzymes have substantially no homology to the mouse DGAT enzyme described in Cases et al., “Identification of a gene encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacylglycerol synthesis,” Proc. Natl. Acad. Sci. U.S.A. 95 (22), 13018-13023 (1998). By substantially no homology is meant that the homology does not exceed about 20%, and usually will not exceed about 10% and more usually will not exceed about as determined using GCG (Genetics Computer Group, Wisconsin Package, Standard Settings, Gap Creation Penalty 3.0, Gap Extension Penalty 0.1). [0032]
  • Of particular interest in many embodiments are proteins that are non-naturally glycosylated. By non-naturally glycosylated is meant that the protein has a glycosylation pattern, if present, which is not the same as the glycosylation pattern found in the corresponding naturally occurring protein. For example, human DGAT2α of the subject invention and of this particular embodiment is characterized by having a glycosylation pattern, if it is glycosylated at all, that differs from that of naturally occurring human DGAT2α. Thus, the non-naturally glycosylated DGAT2α proteins of this embodiment include non-glycosylated DGAT2α proteins, i.e. proteins having no covalently bound glycosyl groups. [0033]
  • The sequence of the full length human DGAT2α protein is identified, infra, as SEQ ID NO:02. As such, DGAT2α proteins having an amino acid sequence that is substantially the same as or identical to the sequence of SEQ ID NO:2 are of interest. By substantially the same as is meant a protein having a region with a sequence that has at least about 75%, usually at least about 90% and more usually at least about 98% sequence identity with the sequence of SED ID NO:02, as measured by GCG, supra. Of particular interest in other embodiments is the mouse DGAT2α protein, where the mouse DGAT2α protein of the subject invention has an amino acid sequence that is substantially the same as or identical to the sequence appearing as SEQ ID NO:04, infra. [0034]
  • In addition to the specific mammalian DGAT2α proteins described above, homologs or proteins (or fragments thereof) from other species, i.e. other animal or plant species, are also provided, where such homologs or proteins may be from a variety of different types of species, including animals, such as mammals, e.g., rodents, such as rats, mice; domestic animals, e.g. horse, cow, dog, cat; humans, and the like. By homolog is meant a protein having at least about 35%, usually at least about 40% and more usually at least about 60% amino acid sequence identity the specific DGAT2α proteins as identified in SEQ ID NOS: 02 to 04, where sequence identity is determined using GCG, supra. Specific homologs of interest include [0035] human DC 2, human DC3, human DC4, human DC5, mouse DC2 and mouse DC3, the sequences of which are provided in FIG. 8 (i.e., SEQ ID NOs. 06, 08, 10, 12, 14 and 16).
  • The DGAT2α proteins of the subject invention (e.g. human DGAT2α or a homolog thereof; non-human DGAT2α proteins, e.g. mouse DGAT2α) are present in a non-naturally occurring environment, e.g. are separated from their naturally occurring environment. In certain embodiments, the subject DGAT2α is present in a composition that is enriched for DGAT2α as compared to DGAT2α in its naturally occurring environment. As such, purified DGAT2α is provided, where by purified is meant that DGAT2α is present in a composition that is substantially free of non DGAT2α proteins, where by substantially free is meant that less than 90%, usually less than 60% and more usually less than 50% of the composition is made up of non-DGAT2α proteins. For compositions that are enriched for DGAT2α proteins, such compositions will exhibit a DGAT2α activity of at least about 100, usually at least about 200 and more usually at least about 1000 pmol triglycerides formed/mg protein/min, where such activity is determined by the assay described in the Experimental Section, infra. [0036]
  • In certain embodiments of interest, the DGAT2α protein is present in a composition that is substantially free of the constituents that are present in its naturally occurring environment. For example, a human DGAT2α protein comprising composition according to the subject invention in this embodiment will be substantially, if not completely, free of those other biological constituents, such as proteins, carbohydrates, lipids, etc., with which it is present in its natural environment. As such, protein compositions of these embodiments will necessarily differ from those that are prepared by purifying the protein from a naturally occurring source, where at least trace amounts of the protein's constituents will still be present in the composition prepared from the naturally occurring source. [0037]
  • The DGAT2α of the subject invention may also be present as an isolate, by which is meant that the DGAT2α is substantially free of both non-DGAT2α proteins and other naturally occurring biologic molecules, such as oligosaccharides, polynucleotides and fragments thereof, and the like, where substantially free in this instance means that less than 70%, usually less than 60% and more usually less than 50% (dry weight) of the composition containing the isolated DGAT2α is a non-DGAT naturally occurring biological molecule. In certain embodiments, the DGAT2α is present in substantially pure form, where by substantially pure form is meant at least 95%, usually at least 97% and more usually at least 99% pure. [0038]
  • In addition to the naturally occurring DGAT2α proteins, DGAT2α polypeptides which vary from the naturally occurring DGAT2α proteins are also provided. By DGAT2α polypeptides is meant proteins having an amino acid sequence encoded by an open reading frame (ORF) of a DGAT2α gene, described supra, including the full length DGAT2α protein and fragments thereof, particularly biologically active fragments and/or fragments corresponding to functional domains; and including fusions of the subject polypeptides to other proteins or parts thereof. Fragments of interest will typically be at least about 10 aa in length, usually at least about 50 aa in length, and may be as long as 300 aa in length or longer, but will usually not exceed about 1000 aa in length, where the fragment will have a stretch of amino acids that is identical to a DGAT2α protein of SEQ ID NO:2, SEQ ID NO:04, or a homolog thereof; of at least about 10 aa, and usually at least about 15 aa, and in many embodiments at least about 50 aa in length. [0039]
  • PREPARATION OF DGAT2α POLYPEPTIDES
  • The subject DGAT2α proteins and polypeptides may be obtained from naturally occurring sources, but are preferably synthetically produced. Where obtained from naturally occurring sources, the source chosen will generally depend on the species from which the DGAT2α is to be derived. [0040]
  • The subject DGAT2α polypeptide compositions may be synthetically derived by expressing a recombinant gene encoding DGAT2α, such as the polynucleotide compositions described above, in a suitable host. For expression, an expression cassette may be employed. The expression vector will provide a transcriptional and translational initiation region, which may be inducible or constitutive, where the coding region is operably linked under the transcriptional control of the transcriptional initiation region, and a transcriptional and translational termination region. These control regions may be native to a DGAT2α gene, or may be derived from exogenous sources. [0041]
  • Expression vectors generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences encoding heterologous proteins. A selectable marker operative in the expression host may be present. Expression vectors may be used for the production of fusion proteins, where the exogenous fusion peptide provides additional functionality, i.e. increased protein synthesis, stability, reactivity with defined antisera, an enzyme marker, e.g. β-galactosidase, etc. [0042]
  • Expression cassettes may be prepared comprising a transcription initiation region, the gene or fragment thereof, and a transcriptional termination region. Of particular interest is the use of sequences that allow for the expression of functional epitopes or domains, usually at least about 8 amino acids in length, more usually at least about 15 amino acids in length, to about 25 amino acids, and up to the complete open reading frame of the gene. After introduction of the DNA, the cells containing the construct may be selected by means of a selectable marker, the cells expanded and then used for expression. [0043]
  • DGAT2α proteins and polypeptides may be expressed in prokaryotes or eukaryotes in accordance with conventional ways, depending upon the purpose for expression. For large scale production of the protein, a unicellular organism, such as [0044] E. coli, B. subtilis, S. cerevisiae, insect cells in combination with baculovirus vectors, or cells of a higher organism such as vertebrates, particularly mammals, e.g. COS 7 cells, may be used as the expression host cells. In some situations, it is desirable to express the DGAT2α coding sequence in eukaryotic cells, where the DGAT2α protein will benefit from native folding and post-translational modifications. Small peptides can also be synthesized in the laboratory. Polypeptides that are subsets of the complete DGAT2α sequence may be used to identify and investigate parts of the protein important for function.
  • Once the source of the protein is identified and/or prepared, e.g. a transfected host expressing the protein is prepared, the protein is then purified to produce the desired DGAT2α comprising composition. Any convenient protein purification procedures may be employed, where suitable protein purification methodologies are described in Guide to Protein Purification, (Deuthser ed.) (Academic Press, 1990). For example, a lysate may prepared from the original source, e.g. naturally occurring cells or tissues that express DGAT2α or the expression host expressing DGAT2α, and purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, and the like. [0045]
  • Specific expression systems of interest include bacterial, yeast, insect cell and mammalian cell derived expression systems. Representative systems from each of these categories is are provided below: [0046]
  • Bacteria. Expression systems in bacteria include those described in Chang et al., [0047] Nature (1978) 275:615; Goeddel et al., Nature (1979) 281:544; Goeddel et al., Nucleic Acids Res. (1980) 8:4057; EP 0 036,776; U.S. Pat. No. 4,551,433; DeBoer et al., Proc. Natl. Acad. Sci. (USA) (1983) 80:21-25; and Siebenlist et al., Cell (1980) 20:269.
  • Yeast. Expression systems in yeast include those described in Hinnen et al., [0048] Proc. Natl. Acad. Sci. (USA) (1978) 75:1929; Ito et al., J. Bacteriol. (1983) 153:163; Kurtz et al., Mol. Cell. Biol. (1986) 6:142; Kunze et al., J. Basic Microbiol. (1985) 25:141; Gleeson et al., J. Gen. Microbiol. (1986) 132:3459; Roggenkamp et al., Mol. Gen. Genet. (1986) 202:302; Das et al., J. Bacteriol. (1984) 158:1165; De Louvencourt et al., J. Bacteriol. (1983) 154:737; Van den Berg et al., Bio/Technology (1990) 8:135; Kunze et al., J. Basic Microbiol. (1985) 25:141; Cregg et al., Mol. Cell. Biol. (1985) 5:3376; U.S. Pat. Nos. 4,837,148 and 4,929,555; Beach and Nurse, Nature (1981) 300:706; Davidow et al., Curr. Genet. (1985) 10:380; Gaillardin et al., Curr. Genet. (1985) 10:49; Ballance et al., Biochem. Biophys. Res. Commun. (1983) 112:284-289; Tilburn et al., Gene (1983) 26:205-221; Yelton et al., Proc. Natl. Acad. Sci. (USA) (1984) 81:1470-1474; Kelly and Hynes, EMBO J. (1985) 4:475479; EP 0 244,234; and WO 91/00357.
  • Insect Cells. Expression of heterologous genes in insects is accomplished as described in U.S. Pat. No. 4,745,051; Friesen et al., “The Regulation of Baculovirus Gene Expression”, in: [0049] The Molecular Biology Of Baculoviruses (1986) (W. Doerfler, ed.); EP 0 127,839; EP 0 155,476; and Vlak et al., J. Gen. Virol. (1988) 69:765-776; Miller et al., Ann. Rev. Microbiol. (1988) 42:177; Carbonell et al., Gene (1988) 73:409; Maeda et al., Nature (1985) 315:592-594; Lebacq-Verheyden et al., Mol. Cell. Biol. (1988) 8:3129; Smith et al., Proc. Natl. Acad. Sci. (USA) (1985) 82:8844; Miyajima et al., Gene (1987) 58:273; and Martin et al., DNA (1988) 7:99. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts are described in Luckow et al., Bio/Technology (1988) 6:47-55, Miller et al., Generic Engineering (1986) 8:277-279, and Maeda et al., Nature (1985) 315:592-594.
  • Mammalian Cells. Mammalian expression is accomplished as described in Dijkema et al., [0050] EMBO J. (1985) 4:761, Gorman et al., Proc. Natl. Acad. Sci. (USA) (1982) 79:6777, Boshart et al., Cell (1985) 41:521 and U.S. Pat. No. 4,399,216. Other features of mammalian expression are facilitated as described in Ham and Wallace, Meth. Enz. (1979) 58:44, Barnes and Sato, Anal. Biochem. (1980) 102:255, U.S. Pat. Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655, WO 90/103430, WO 87/00195, and U.S. RE 30,985.
  • When any of the above host cells, or other appropriate host cells or organisms, are used to replicate and/or express the polynucleotides or nucleic acids of the invention, the resulting replicated nucleic acid, RNA, expressed protein or polypeptide, is within the scope of the invention as a product of the host cell or organism. The product is recovered by any appropriate means known in the art. [0051]
  • Once the gene corresponding to a selected polynucleotide is identified, its expression can be regulated in the cell to which the gene is native. For example, an endogenous gene of a cell can be regulated by an exogenous regulatory sequence as disclosed in U.S. Pat. No. 5,641,670. [0052]
  • METHODS AND COMPOSITIONS HAVING RESEARCH APPLICATION
  • Also provided by the subject invention are methods and compositions having research applications, such as in the study of the acylglycerol metabolism, in the identification of key components of the triglyceride synthesis pathway, in the identification of triglyceride synthesis modulatory agents, e.g. DGAT2α inhibitors or enhancers, and the like. [0053]
  • The subject nucleic acid compositions find use in a variety of research applications. Research applications of interest include: the identification of DGAT2α homologs; as a source of novel promoter elements; the identification of DGAT2α expression regulatory factors; as probes and primers in hybridization applications, e.g. PCR; the identification of expression patterns in biological specimens; the preparation of cell or animal models for DGAT2α function; the preparation of in vitro models for DGAT2α function; etc. Homologs of the specifically disclosed DGAT2α nucleic acids are identified by any of a number of methods. A fragment of the provided cDNA may be used as a hybridization probe against a cDNA library from the target organism of interest, where low stringency conditions are used. The probe may be a large fragment, or one or more short degenerate primers. Nucleic acids having sequence similarity are detected by hybridization under low stringency conditions, for example, at 50° C. and 6×SSC (0.9 M sodium chloride/0.09 M sodium citrate) and remain bound when subjected to washing at 55° C. in 1×SSC (0.15 M sodium chloride/0.015 M sodium citrate). Sequence identity may be determined by hybridization under stringent conditions, for example, at 50° C. or higher and 0.1×SSC (15 mM sodium chloride/01.5 mM sodium citrate). Nucleic acids having a region of substantial identity to the provided nucleic acid sequences bind to the provided sequences under stringent hybridization conditions. By using probes, particularly labeled probes of DNA sequences, one can isolate homologous or related genes. One can also use sequence information derived from the polynucleotide compositions of the subject invention to prepare electronic “probes” for use in searching of computer based sequence date, e.g. BLAST searches EST databases. [0054]
  • The sequence of the 5′ flanking region of the subject nucleic acid compositions may be utilized as a source for promoter elements, including enhancer binding sites, that provide for developmental regulation in tissues where DGAT2α is expressed. The tissue specific expression is useful for determining the pattern of expression, and for providing promoters that mimic the native pattern of expression. Naturally occurring polymorphisms in the promoter region are useful for determining natural variations in expression, particularly those that may be associated with disease. [0055]
  • Alternatively, mutations may be introduced into the promoter region to determine the effect of altering expression in experimentally defined systems. Methods for the identification of specific DNA motifs involved in the binding of transcriptional factors are known in the art, e.g. sequence similarity to known binding motifs, gel retardation studies, etc. For examples, see Blackwell et al. (1995), [0056] Mol. Med. 1:194-205; Mortlock et al. (1996), Genome Res. 6:327-33; and Joulin and Richard-Foy (1995), Eur. J. Biochem. 232:620-626.
  • The regulatory sequences may be used to identify cis acting sequences required for transcriptional or translational regulation of DGAT2 gene expression, especially in different tissues or stages of development, and to identify cis acting sequences and trans-acting factors that regulate or mediate DGAT2 gene expression. Such transcription or translational control regions may be operably linked to a DGAT2 gene in order to promote expression of wild type or altered DGAT2 or other proteins of interest in cultured cells, or in embryonic, fetal or adult tissues, and for gene therapy. [0057]
  • Small DNA fragments are useful as primers for PCR, hybridization screening probes, etc. Larger DNA fragments, i.e. greater than 100 nt are useful for production of the encoded polypeptide, as described in the previous section. For use in amplification reactions, such as PCR, a pair of primers will be used. The exact composition of the primer sequences is not critical to the invention, but for most applications the primers will hybridize to the subject sequence under stringent conditions, as known in the art. It is preferable to choose a pair of primers that will generate an amplification product of at least about 50 nt, preferably at least about 100 nt. Algorithms for the selection of primer sequences are generally known, and are available in commercial software packages. Amplification primers hybridize to complementary strands of DNA, and will prime towards each other. [0058]
  • The DNA may also be used to identify expression of the gene in a biological specimen. The manner in which one probes cells for the presence of particular nucleotide sequences, as genomic DNA or RNA, is well established in the literature. Briefly, DNA or mRNA is isolated from a cell sample. The mRNA may be amplified by RT-PCR, using reverse transcriptase to form a complementary DNA strand, followed by polymerase chain reaction amplification using primers specific for the subject DNA sequences. Alternatively, the mRNA sample is separated by gel electrophoresis, transferred to a suitable support, e.g. nitrocellulose, nylon, etc., and then probed with a fragment of the subject DNA as a probe. Other techniques, such as oligonucleotide ligation assays, in situ hybridizations, and hybridization to DNA probes arrayed on a solid chip may also find use. Detection of mRNA hybridizing to the subject sequence is indicative of DGAT2α gene expression in the sample. [0059]
  • The sequence of a DGAT2α gene or nucleic acid, including flanking promoter regions and coding regions, may be mutated in various ways known in the art to generate targeted changes in promoter strength, sequence of the encoded protein, etc. The DNA sequence or protein product of such a mutation will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one nucleotide or amino acid, respectively, and may differ by at least two but not more than about ten nucleotides or amino acids. The sequence changes may be substitutions, insertions, deletions, or a combination thereof. Deletions may further include larger changes, such as deletions of a domain or exon. Other modifications of interest include epitope tagging, e.g. with the FLAG system, HA, etc. For studies of subcellular localization, fusion proteins with green fluorescent proteins (GFP) may be used. [0060]
  • Techniques for in vitro mutagenesis of cloned genes are known. Examples of protocols for site specific mutagenesis may be found in Gustin et al. (1993), [0061] Biotechniques 14:22; Barany (1985), Gene 37:111-23; Colicelli et al. (1985), Mol. Gen. Genet. 199:537-9; and Prentki et al (1984), Gene 29:303-13. Methods for site specific mutagenesis can be found in Sambrook et al., Molecular Cloning: A Laboratory Manual, CSH Press 1989, pp. 15.3-15.108; Weiner et al. (1993), Gene 126:35-41; Sayers et al. (1992), Biotechniques 13:592-6; Jones and Winistorfer (1992), Biotechniques 12:528-30; Barton et al. (1990), Nucleic Acids Res 18:7349-55; Marotti and Tomich (1989), Gene Anal. Tech. 6:67-70; and Zhu (1989), Anal Biochem 177:120-4. Such mutated genes may be used to study structure-function relationships of DGAT2α, or to alter properties of the protein that affect its function or regulation.
  • The subject nucleic acids can be used to generate transgenic hosts, e.g non-human animals, such as mice, cows, rats, pigs etc., or site specific gene modifications in cell lines. Examples of transgenic hosts include hosts in which the naturally expressed DGAT2α gene has been disrupted, e.g. DGAT2α knock-outs, as well as hosts in which DGAT2α expression has been amplified, e.g. through introduction of additional DGAT2α copies, through introduction of strong promoter upstream of the DGAT2α gene, and the like. Using the nucleic acid compositions of the subject invention, standard protocols known to those of skill in the art may used to produce such transgenic hosts that have been genetically manipulated with respect to the DGAT2α gene, i.e. DGAT2α transgenic hosts. [0062]
  • Transgenic animals may be made through homologous recombination, where the normal DGAT2α locus is altered, e.g. as in DGAT2α knockouts. Alternatively, a nucleic acid construct is randomly integrated into the genome. Vectors for stable integration include plasmids, retroviruses and other animal viruses, YACs, and the like. DNA constracts for homologous recombination will comprise at least a portion of the DGAT2α gene native to the species of the host animal, wherein the gene has the desired genetic modification(s), and includes regions of homology to the target locus. DNA constructs for random integration need not include regions of homology to mediate recombination. Conveniently, markers for positive and negative selection are included. Methods for generating cells having targeted gene modifications through homologous recombination are known in the art. For various techniques for transfecting mammalian cells, see Keown et al. (1990), [0063] Meth. Enzymol. 185:527-537.
  • For embryonic stem (ES) cells, an ES cell line may be employed, or embryonic cells may be obtained freshly from a host, e.g. mouse, rat, guinea pig, cow, etc. Such cells are grown on an appropriate fibroblast-feeder layer or grown in the presence of leukemia inhibiting factor (LIF). When ES or embryonic cells have been transformed, they may be used to produce transgenic animals. After transformation, the cells are plated onto a feeder layer in an appropriate medium. Cells containing the construct may be detected by employing a selective medium. After sufficient time for colonies to grow, they are picked and analyzed for the occurrence of homologous recombination or integration of the construct. Those colonies that are positive may then be used for embryo manipulation and blastocyst injection. Blastocysts are obtained from 4 to 6 week old superovulated females. The ES cells are trypsinized, and the modified cells are injected into the blastocoel of the blastocyst. After injection, the blastocysts are returned to each uterine horn of pseudopregnant females. Females are then allowed to go to term and the resulting offspring screened for the construct. By providing for a different phenotype of the blastocyst and the genetically modified cells, chimeric progeny can be readily detected. [0064]
  • The resultant chimeric animals are screened for the presence of the modified gene and males and females having the modification are mated to produce homozygous progeny. If the gene alterations cause lethality at some point in development, tissues or organs can be maintained as allogeneic or congenic grafts or transplants, or in in vitro culture. The transgenic animals may be any non-human mammal, such as laboratory animals, domestic animals, etc. [0065]
  • Transgenic plants may be produced in a similar manner. Methods of preparing transgenic plant cells and plants are described in U.S. Pat. Nos. 5,767,367; 5,750,870; 5,739,409; 5,689,049; 5,689,045; 5,674,731; 5,656,466; 5,633,155; 5,629,470 ; 5,595,896; 5,576,198; 5,538,879; 5,484,956; the disclosures of which are herein incorporated by reference. Methods of producing transgenic plants are also reviewed in Plant Biochemistry and Molecular Biology (eds Lea & Leegood, John Wiley & Sons)(1993) pp 275-295. In brief, a suitable plant cell or tissue is harvested, depending on the nature of the plant species. As such, in certain instances, protoplasts will be isolated, where such protoplasts may be isolated from a variety of different plant tissues, e.g. leaf, hypoctyl, root, etc. For protoplast isolation, the harvested cells are incubated in the presence of cellulases in order to remove the cell wall, where the exact incubation conditions vary depending on the type of plant and/or tissue from which the cell is derived. The resultant protoplasts are then separated from the resultant cellular debris by sieving and centrifugation. Instead of using protoplasts, embryogenic explants comprising somatic cells may be used for preparation of the transgenic host. Following cell or tissue harvesting, exogenous DNA of interest is introduced into the plant cells, where a variety of different techniques are available for such introduction. With isolated protoplasts, the opportunity arise for introduction via DNA-mediated gene transfer protocols, including: incubation of the protoplasts with naked DNA, e.g. plasmids, comprising the exogenous coding sequence of interest in the presence of polyvalent cations, e.g. PEG or PLO; and electroporation of the protoplasts in the presence of naked DNA comprising the exogenous sequence of interest. Protoplasts that have successfully taken up the exogenous DNA are then selected, grown into a callus, and ultimately into a transgenic plant through contact with the appropriate amounts and ratios of stimulatory factors, e.g. auxins and cytokinins. With embryogenic explants, a convenient method of introducing the exogenous DNA in the target somatic cells is through the use of particle acceleration or “gene-gun” protocols. The resultant explants are then allowed to grow into chimera plants, cross-bred and transgenic progeny are obtained. Instead of the naked DNA approaches described above, another convenient method of producing transgenic plants is Agrobacterium mediated transformation. With Agrobacterium mediated transformation, co-integrative or binary vectors comprising the exogenous DNA are prepared and then introduced into an appropriate Agrobacterium strain, e.g. A. tumefaciens. The resultant bacteria are then incubated with prepared protoplasts or tissue explants, e.g. leaf disks, and a callus is produced. The callus is then grown under selective conditions, selected and subjected to growth media to induce root and shoot growth to ultimately produce a transgenic plant. [0066]
  • The modified cells, animals or plants are useful in the study of DGAT2α function and regulation. For example, a series of small deletions and/or substitutions may be made in the host's native DGAT2α gene to determine the role of different exons in various physiological processes. Specific constructs of interest include anti-sense DGAT2α, which will block DGAT2α expression, expression of dominant negative DGAT2α mutations, and over-expression of DGAT2α genes. Where a DGAT2α sequence is introduced, the introduced sequence may be either a complete or partial sequence of a DGAT2α gene native to the host, or may be a complete or partial DGAT2α sequence that is exogenous to the host animal, e.g., a human DGAT2α sequence. A detectable marker, such as lac Z may be introduced into the DGAT2α locus, where upregulation of DGAT2α gene expression will result in an easily detected change in phenotype. One may also provide for expression of the DGAT2α gene or variants thereof in cells or tissues where it is not normally expressed, at levels not normally present in such cells or tissues, or at abnormal times of development. The transgenic hosts, e.g. animals, plants, etc., may be used in functional studies, drug screening, etc., e.g. to determine the effect of a candidate drug on DGAT2α activity, to identify drugs that reduce serum triglyceride levels, etc. [0067]
  • The subject polypeptide compositions can be used to produce in vitro models of triglyceride synthesis, where such models will consist of the subject DGAT2α proteins and other components of triglyceride synthesis, e.g. substrates, such as diacylglycerol or metabolic precursors thereof, fatty acyl CoAs and the like, other components of the triacylglycerol synthetase complex, e.g. acyl CoA ligase, acyl CoA acyltransferase, monoacyl glycerol acyltransferase, etc. [0068]
  • Also provided by the subject invention are screening assays designed to find modulatory agents of DGAT2α activity, e.g. inhibitors or enhancers of DGAT2α activity, as well as the agents identified thereby, where such agents may find use in a variety of applications, including as therapeutic agents, as agricultural chemicals, etc. The screening methods will typically be assays which provide for qualitative/quantitative measurements of DGAT2α activity in the presence of a particular candidate therapeutic agent. For example, the assay could be an assay which measures the acylation activity of DGAT2α in the presence and absence of a candidate inhibitor agent. The screening method may be an in vitro or in vivo format, where both formats are readily developed by those of skill in the art. Depending on the particular method, one or more of, usually one of, the components of the screening assay may be labeled, where by labeled is meant that the components comprise a detectable moiety, e.g. a fluorescent or radioactive tag, or a member of a signal producing system, e.g. biotin for binding to an enzyme-streptavidin conjugate in which the enzyme is capable of converting a substrate to a chromogenic product. Where in vitro assays are employed, the various components of the in vitro assay, e.g. the substrate, the donor, the DGAT2α protein and the candidate inhibitor, etc. are combined in a assay mixture under conditions sufficient for DGAT2α activity to occur, as described in the experimental section, infra. [0069]
  • A variety of other reagents may be included in the screening assay and reaction mixture. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc that are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Reagents that improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc. may be used. [0070]
  • A variety of different candidate agents may be screened by the above methods. Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. [0071]
  • Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs. [0072]
  • Using the above screening methods, a variety of different therapeutic agents may be identified. Such agents may target the enzyme itself, or an expression regulator factor thereof. Such agents may inhibitors or promoters of DGAT2α activity, where inhibitors are those agents that result in at least a reduction of DGAT2α activity as compared to a control and enhancers result in at least an increase in DGAT2α activity as compared to a control. Such agents may be find use in a variety of therapeutic applications, as described in greater detail below. [0073]
  • METHODS AND COMPOSITIONS HAVING MEDICAL APPLICATIONS
  • The methods and compositions of the subject invention also have broad ranging applications in a variety of medical applications, including diagnostic screening, therapeutic treatments of pathological conditions, in the regulation of DGAT2α activity in desirable ways, and the like. [0074]
  • The subject invention provides methods of screening individuals for a predisposition to a disease state or the presence of disease state, where such screening may focus on the presence of one or more markers, such as a mutated DGAT2α gene or expression regulatory element thereof, observed levels of DGAT2α; the expression level of the DGAT2α gene in a biological sample of interest; and the like. [0075]
  • Samples, as used herein, include biological fluids such as blood, cerebrospinal fluid, tears, saliva, lymph, semen, dialysis fluid and the like; organ or tissue culture derived fluids; and fluids extracted from physiological tissues. Also included in the term are derivatives and fractions of such fluids. The cells may be dissociated, in the case of solid tissues, or tissue sections may be analyzed. Alternatively a lysate of the cells may be prepared. [0076]
  • A number of methods are available for determining the expression level of a gene or protein in a particular sample. Diagnosis may be performed by a number of methods to determine the absence or presence or altered amounts of normal or abnormal DGAT2α in a patient sample. For example, detection may utilize staining of cells or histological sections with labeled antibodies, performed in accordance with conventional methods. Cells are permeabilized to stain cytoplasmic molecules. The antibodies of interest are added to the cell sample, and incubated for a period of time sufficient to allow binding to the epitope, usually at least about 10 minutes. The antibody may be labeled with radioisotopes, enzymes, fluorescers, chemiluminescers, or other labels for direct detection. Alternatively, a second stage antibody or reagent is used to amplify the signal. Such reagents are well known in the art. For example, the primary antibody may be conjugated to biotin, with horseradish peroxidase-conjugated avidin added as a second stage reagent. Alternatively, the secondary antibody conjugated to a flourescent compound, e.g. fluorescein, rhodamine, Texas red, etc. Final detection uses a substrate that undergoes a color change in the presence of the peroxidase. The absence or presence of antibody binding may be determined by various methods, including flow cytometry of dissociated cells, microscopy, radiography, scintillation counting, etc. [0077]
  • Alternatively, one may focus on the expression of DGAT2α encoding nucleic acids. Biochemical studies may be performed to determine whether a sequence polymorphism in a DGAT2α coding region or control regions is associated with disease. Disease associated polymorphisms may include deletion or truncation of the gene, mutations that alter expression level, that affect the activity of the protein, etc. [0078]
  • Changes in the promoter or enhancer sequence that may affect expression levels of DGAT2α can be compared to expression levels of the normal allele by various methods known in the art. Methods for determining promoter or enhancer strength include quantitation of the expressed natural protein; insertion of the variant control element into a vector with a reporter gene such as β-galactosidase, luciferase, chloramphenicol acetyltransferase, etc. that provides for convenient quantitation; and the like. [0079]
  • A number of methods are available for analyzing nucleic acids for the presence of a specific sequence, e.g. a disease associated polymorphism. Where large amounts of DNA are available, genomic DNA is used directly. Alternatively, the region of interest is cloned into a suitable vector and grown in sufficient quantity for analysis. Cells that express DGAT2α may be used as a source of mRNA, which may be assayed directly or reverse transcribed into cDNA for analysis. The nucleic acid may be amplified by conventional techniques, such as the polymerase chain reaction (PCR), to provide sufficient amounts for analysis. The use of the polymerase chain reaction is described in Saiki, et al. (1985), [0080] Science 239:487, and a review of techniques may be found in Sambrook, et al. Molecular Cloning: A Laboratory Manual, CSH Press 1989, pp.14.2-14.33. Alternatively, various methods are known in the art that utilize oligonucleotide ligation as a means of detecting polymorphisms, for examples see Riley et al. (1990), Nucl Acids Res. 18:2887-2890; and Delahunty et al. (1996), Am. J. Hum. Genet. 58:1239-1246.
  • A detectable label may be included in an amplification reaction. Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein (6-FAM), 2′,7′-dimethoxy-4′,5′-dichloro-6-carboxyfluorescein (JOE), 6-carboxy-X-rhodamine (ROX), 6-carboxy-2′,4′,7′,4,7-hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N′,N′-tetramethyl-6-carboxyrhodamine (TAMRA), radioactive labels, e.g. [0081] 32P, 35S, 3H; etc. The label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label. The label may be conjugated to one or both of the primers. Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.
  • The sample nucleic acid, e.g. amplified or cloned fragment, is analyzed by one of a number of methods known in the art. The nucleic acid may be sequenced by dideoxy or other methods, and the sequence of bases compared to a wild-type DGAT2α sequence. Hybridization with the variant sequence may also be used to determine its presence, by Southern blots, dot blots, etc. The hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilized on a solid support, as described in U.S. Pat. No. 5,445,934, or in WO 95/35505, may also be used as a means of detecting the presence of variant sequences. Single strand conformational polymorphism (SSCP) analysis, denaturing gradient gel electrophoresis (DGGE), and heteroduplex analysis in gel matrices are used to detect conformational changes created by DNA sequence variation as alterations in electrophoretic mobility. Alternatively, where a polymorphism creates or destroys a recognition site for a restriction endonuclease, the sample is digested with that endonuclease, and the products size fractionated to determine whether the fragment was digested. Fractionation is performed by gel or capillary electrophoresis, particularly acrylamide or agarose gels. [0082]
  • Screening for mutations in DGAT2α may be based on the functional or antigenic characteristics of the protein. Protein truncation assays are useful in detecting deletions that may affect the biological activity of the protein. Various immunoassays designed to detect polymorphisms in DGAT2α proteins may be used in screening. Where many diverse genetic mutations lead to a particular disease phenotype, functional protein assays have proven to be effective screening tools. The activity of the encoded DGAT2α protein may be determined by comparison with the wild-type protein. [0083]
  • Diagnostic methods of the subject invention in which the level of DGAT2α expression is of interest will typically involve comparison of the DGAT2α nucleic acid abundance of a sample of interest with that of a control value to determine any relative differences, where the difference may be measured qualitatively and/or quantitatively, which differences are then related to the presence or absence of an abnormal DGAT2α expression pattern. A variety of different methods for determining the nucleic acid abundance in a sample are known to those of skill in the art, where particular methods of interest include those described in: Pietu et al., Genome Res. (June 1996) 6: 492-503; Zhao et al., Gene (Apr. 24, 1995) 156: 207-213; Soares , Curr. Opin. Biotechnol. (October 1997) 8: 542-546; Raval, J. Pharmacol Toxicol Methods (November 1994) 32: 125-127; Chalifour et al., Anal. Biochem (Feb. 1, 1994) 216: 299-304; Stolz & Tuan, Mol. Biotechnol. (December 19960 6: 225-230; Hong et al., Bioscience Reports (1982) 2: 907; and McGraw, Anal. Biochem. (1984) 143: 298. Also of interest are the methods disclosed in WO 97/27317, the disclosure of which is herein incorporated by reference. [0084]
  • The subject diagnostic or screening methods may be used to identify the presence of, or predisposition to, disease conditions associated with acylglycerol metabolism, particularly those associated with DGAT2α activity. Such disease conditions include: hyperlipidemia (including excess serum triglyceride levels), cardiovascular disease, obesity, diabetes, cancer, neurological disorders, immunological disorders, and the like. [0085]
  • Also provided are methods of regulating, including enhancing and inhibiting, DGAT2α activity in a host. A variety of situations arise where modulation of DGAT2α activity in a host is desired, where such conditions include disease conditions associated with DGAT2α activity and non-disease conditions in which a modulation of DGAT2α activity is desired for a variety of different reasons, e.g. cosmetic weight control. [0086]
  • For the modulation of DGAT2α activity in a host, an effective amount of active agent that modulates the activity, e.g. reduces the activity, of DGAT2α in vivo, is administered to the host. The active agent may be a variety of different compounds, including: the polynucleotide compositions of the subject invention, the polypeptide compositions of the subject invention, a naturally occurring or synthetic small molecule compound, an antibody, fragment or derivative thereof, an antisense composition, and the like. [0087]
  • The nucleic acid compositions of the subject invention find use as therapeutic agents in situations where one wishes to enhance DGAT2α activity in a host, e.g. in a mammalian host in which DGAT2α activty is low resulting in a disease condition, etc. The DGAT2α genes, gene fragments, or the encoded DGAT2α protein or protein fragments are useful in gene therapy to treat disorders associated with DGAT2α defects. Expression vectors may be used to introduce the DGAT2α gene or encoding nucleic acid into a cell. Such vectors generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences. Transcription cassettes may be prepared comprising a transcription initiation region, the target gene or fragment thereof, and a transcriptional termination region. The transcription cassettes may be introduced into a variety of vectors, e.g. plasmid; retrovirus, e.g. lentivirus; adenovirus; and the like, where the vectors are able to transiently or stably be maintained in the cells, usually for a period of at least about one day, more usually for a period of at least about several days to several weeks. [0088]
  • Naturally occurring or synthetic small molecule compounds of interest include numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Of particular interest are those agents identified by the screening assays of the subject invention, as described above. [0089]
  • Also of interest as active agents are antibodies that modulate, e.g. reduce, if not inhibit, DGAT2α activity in the host. Suitable antibodies are obtained by immunizing a host animal with peptides comprising all or a portion of a DGAT protein, such as the DGAT2α polypeptide compositions of the subject invention. Suitable host animals include mouse, rat sheep, goat, hamster, rabbit, etc. The origin of the protein immunogen may be mouse, human, rat, monkey etc. The host animal will generally be a different species than the immunogen, e.g. human DGAT used to immunize mice, etc. [0090]
  • The immunogen may comprise the complete protein, or fragments and derivatives thereof. Preferred immunogens comprise all or a part of DGAT2α, where these residues contain the post-translation modifications, such as glycosylation, found on the native DGAT2α. Immunogens comprising the extracellular domain are produced in a variety of ways known in the art, e.g. expression of cloned genes using conventional recombinant methods, isolation from HEC, etc. [0091]
  • For preparation of polyclonal antibodies, the first step is immunization of the host animal with DGAT2α, where the DGAT2α will preferably be in substantially pure form, comprising less than about 1% contaminant. The immunogen may comprise complete DGAT2α, fragments or derivatives thereof. To increase the immune response of the host animal, the DGAT may be combined with an adjuvant, where suitable adjuvants include alum, dextran, sulfate, large polymeric anions, oil & water emulsions, e.g. Freund's adjuvant, Freund's complete adjuvant, and the like. The DGAT2α may also be conjugated to synthetic carrier proteins or synthetic antigens. A variety of hosts may be immunized to produce the polyclonal antibodies. Such hosts include rabbits, guinea pigs, rodents, e.g. mice, rats, sheep, goats, and the like. The DGAT2α is administered to the host, usually intradermally, with an initial dosage followed by one or more, usually at least two, additional booster dosages. Following immunization, the blood from the host will be collected, followed by separation of the serum from the blood cells. The Ig present in the resultant antiserum may be further fractionated using known methods, such as ammonium salt fractionation, DEAE chromatography, and the like. [0092]
  • Monoclonal antibodies are produced by conventional techniques. Generally, the spleen and/or lymph nodes of an immunized host animal provide a source of plasma cells. The plasma cells are immortalized by fusion with myeloma cells to produce hybridoma cells. Culture supernatant from individual hybridomas is screened using standard techniques to identify those producing antibodies with the desired specificity. Suitable animals for production of monoclonal antibodies to the human protein include mouse, rat, hamster, etc. To raise antibodies against the mouse protein, the animal will generally be a hamster, guinea pig, rabbit, etc. The antibody may be purified from the hybridoma cell supernatants or ascites fluid by conventional techniques, e.g. affinity chromatography using DGAT2α bound to an insoluble support, protein A sepharose, etc. [0093]
  • The antibody may be produced as a single chain, instead of the normal multimeric structure. Single chain antibodies are described in Jost et al. (1994) [0094] J.B.C. 269:26267-73, and others. DNA sequences encoding the variable region of the heavy chain and the variable region of the light chain are ligated to a spacer encoding at least about 4 amino acids of small neutral amino acids, including glycine and/or serine. The protein encoded by this fusion allows assembly of a functional variable region that retains the specificity and affinity of the original antibody.
  • For in vivo use, particularly for injection into humans, it is desirable to decrease the antigenicity of the antibody. An immune response of a recipient against the blocking agent will potentially decrease the period of time that the therapy is effective. Methods of humanizing antibodies are known in the art. The humanized antibody may be the product of an animal having transgenic human immunoglobulin constant region genes (see for example International Patent Applications WO 90/10077 and WO 90/04036). Alternatively, the antibody of interest may be engineered by recombinant DNA techniques to substitute the CH1, CH2, CH3, hinge domains, and/or the framework domain with the corresponding human sequence (see WO 92/02190). [0095]
  • The use of Ig cDNA for construction of chimeric immunoglobulin genes is known in the art (Liu et al. (1987) [0096] P.N.A.S. 84:3439 and (1987) J. Immunol. 139:3521). mRNA is isolated from a hybridoma or other cell producing the antibody and used to produce CDNA. The cDNA of interest may be amplified by the polymerase chain reaction using specific primers (U.S. Pat. Nos. 4,683,195 and 4,683,202). Alternatively, a library is made and screened to isolate the sequence of interest. The DNA sequence encoding the variable region of the antibody is then fused to human constant region sequences. The sequences of human constant regions genes may be found in Kabat et al. (1991) Sequences of Proteins of Immunological Interest, N.I.H. publication no. 91-3242. Human C region genes are readily available from known clones. The choice of isotype will be guided by the desired effector functions, such as complement fixation, or activity in antibody-dependent cellular cytotoxicity. Preferred isotypes are IgG1, IgG3 and IgG4. Either of the human light chain constant regions, kappa or lambda, may be used. The chimeric, humanized antibody is then expressed by conventional methods.
  • Antibody fragments, such as Fv, F(ab′)[0097] 2 and Fab may be prepared by cleavage of the intact protein, e.g. by protease or chemical cleavage. Alternatively, a truncated gene is designed. For example, a chimeric gene encoding a portion of the F(ab′)2 fragment would include DNA sequences encoding the CH1 domain and hinge region of the H chain, followed by a translational stop codon to yield the truncated molecule.
  • Consensus sequences of H and L J regions may be used to design oligonucleotides for use as primers to introduce useful restriction sites into the J region for subsequent linkage of V region segments to human C region segments. C region cDNA can be modified by site directed mutagenesis to place a restriction site at the analogous position in the human sequence. [0098]
  • Expression vectors include plasmids, retroviruses, YACs, EBV derived episomes, and the like. A convenient vector is one that encodes a functionally complete human CH or CL immunoglobulin sequence, with appropriate restriction sites engineered so that any VH or VL sequence can be easily inserted and expressed. In such vectors, splicing usually occurs between the splice donor site in the inserted J region and the splice acceptor site preceding the human C region, and also at the splice regions that occur within the human CH exons. Polyadenylation and transcription termination occur at native chromosomal sites downstream of the coding regions. The resulting chimeric antibody may be joined to any strong promoter, including retroviral LTRs, e.g. SV-40 early promoter, (Okayama et al. (1983) [0099] Mol. Cell. Bio. 3:280), Rous sarcoma virus LTR (Gorman et al. (1982) P.N.A.S. 79:6777), and moloney murine leukemia virus LTR (Grosschedl et al. (1985) Cell 41:885); native Ig promoters, etc.
  • In yet other embodiments of the invention, the active agent is an agent that modulates, and generally decreases or down regulates, the expression of DGAT2α encoding nucleic acids in the host. Antisense molecules can be used to down-regulate expression of these target nucleic aicds in cells. The anti-sense reagent may be antisense oligonucleotides (ODN), particularly synthetic ODN having chemical modifications from native nucleic acids, or nucleic acid constructs that express such anti-sense molecules as RNA. The antisense sequence is complementary to the mRNA of the targeted gene, and inhibits expression of the targeted gene products. Antisense molecules inhibit gene expression through various mechanisms, e.g. by reducing the amount of mRNA available for translation, through activation of RNAse H, or steric hindrance. One or a combination of antisense molecules may be administered, where a combination may comprise multiple different sequences. [0100]
  • Antisense molecules may be produced by expression of all or a part of the target gene sequence in an appropriate vector, where the transcriptional initiation is oriented such that an antisense strand is produced as an RNA molecule. Alternatively, the antisense molecule is a synthetic oligonucleotide. Antisense oligonucleotides will generally be at least about 7, usually at least about 12, more usually at least about 20 nucleotides in length, and not more than about 500, usually not more than about 50, more usually not more than about 35 nucleotides in length, where the length is governed by efficiency of inhibition, specificity, including absence of cross-reactivity, and the like. It has been found that short oligonucleotides, of from 7 to 8 bases in length, can be strong and selective inhibitors of gene expression (see Wagner et al. (1996), [0101] Nature Biotechnol. 14:840-844).
  • A specific region or regions of the endogenous sense strand mRNA sequence is chosen to be complemented by the antisense sequence. Selection of a specific sequence for the oligonucleotide may use an empirical method, where several candidate sequences are assayed for inhibition of expression of the target gene in an in vitro or animal model. A combination of sequences may also be used, where several regions of the mRNA sequence are selected for antisense complementation. [0102]
  • Antisense oligonucleotides may be chemically synthesized by methods known in the art (see Wagner et al. (1993), supra, and Milligan et al., supra.) Preferred oligonucleotides are chemically modified from the native phosphodiester structure, in order to increase their intracellular stability and binding affinity. A number of such modifications have been described in the literature, which alter the chemistry of the backbone, sugars or heterocyclic bases. [0103]
  • Among useful changes in the backbone chemistry are phosphorothioates; phosphorodithioates, where both of the non-bridging oxygens are substituted with sulfur; phosphoroamidites; alkyl phosphotriesters and boranophosphates. Achiral phosphate derivatives include 3′-O′-5′-S-phosphorothioate, 3′-S-5′-O-phosphorothioate, 3′-CH2-5′-O-phosphonate and 3′-NH-5′-O-phosphoroamidate. Peptide nucleic acids replace the entire ribose phosphodiester backbone with a peptide linkage. Sugar modifications are also used to enhance stability and affinity. The α-anomer of deoxyribose may be used, where the base is inverted with respect to the natural β-anomer. The 2′-OH of the ribose sugar may be altered to form 2′-O-methyl or 2′-O-allyl sugars, which provides resistance to degradation without comprising affinity. Modification of the heterocyclic bases must maintain proper base pairing. Some useful substitutions include deoxyuridine for deoxythymidine; 5-methyl-2′-deoxycytidine and 5-bromo-2′-deoxycytidine for deoxycytidine. 5- propynyl-2′-deoxyuridine and 5-propynyl-2′-deoxycytidine have been shown to increase affinity and biological activity when substituted for deoxythymidine and deoxycytidine, respectively. [0104]
  • As an alternative to anti-sense inhibitors, catalytic nucleic acid compounds, e.g. ribozymes, anti-sense conjugates, etc. may be used to inhibit gene expression. Ribozymes may be synthesized in vitro and administered to the patient, or may be encoded on an expression vector, from which the ribozyme is synthesized in the targeted cell (for example, see International patent application WO 9523225, and Beigelman et al. (1995), [0105] Nucl. Acids Res. 23:4434-42). Examples of oligonucleotides with catalytic activity are described in WO 9506764. Conjugates of anti-sense ODN with a metal complex, e.g. terpyridylCu(II), capable of mediating mRNA hydrolysis are described in Bashkin et al. (1995), Appl. Biochem. Biotechnol. 54:43-56.
  • As mentioned above, an effective amount of the active agent is administered to the host, where “effective amount” means a dosage sufficient to produce a desired result, where the desired result in the desired modulation, e.g. enhancement, reduction, of DGAT2α activity. [0106]
  • In the subject methods, the active agent(s) may be administered to the host using any convenient means capable of resulting in the desired effect. Thus, the agent can be incorporated into a variety of formulations for therapeutic administration. More particularly, the agents of the present invention can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols. [0107]
  • As such, administration of the agents can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intracheal,etc., administration. [0108]
  • In pharmaceutical dosage forms, the agents may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting. [0109]
  • For oral preparations, the agents can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents. [0110]
  • The agents can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. [0111]
  • The agents can be utilized in aerosol formulations to be administered via inhalation. The compounds of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like. [0112]
  • Furthermore, the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds of the present invention can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature. [0113]
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more inhibitors. Similarly, unit dosage forms for injection or intravenous administration may comprise the inhibitor(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier. [0114]
  • The term “unit dosage form,” as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for the novel unit dosage forms of the present invention depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host. [0115]
  • The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public. [0116]
  • Where the agent is a polypeptide, polynucleotide, analog or mimetic thereof, e.g. antisense composition, it may be introduced into tissues or host cells by any number of routes, including viral infection, microinjection, or fusion of vesicles. Jet injection may also be used for intramuscular administration, as described by Furth et al. (1992), [0117] Anal Biochem 205:365-368. The DNA may be coated onto gold microparticles, and delivered intradermally by a particle bombardment device, or “gene gun” as described in the literature (see, for example, Tang et al. (1992), Nature 356:152-154), where gold microprojectiles are coated with the DGAT DNA, then bombarded into skin cells.
  • Those of skill will readily appreciate that dose levels can vary as a function of the specific compound, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. [0118]
  • The subject methods find use in the treatment of a variety of different disease conditions involving acylglycerol metabolism, and particularly DGAT2α activity, including both insufficient or hypo- DGAT2α activity and hyper- DGAT2α activity. Representative diseases that may be treated according to the subject methods include: hyperlipidemia (including excess serum triglyceride levels), cardiovascular disease, obesity, diabetes, cancer, neurological disorders, immunological disorders, skin disorders associated with sebaceous gland activity, e.g. acne, and the like. [0119]
  • By treatment is meant at least an amelioration of the symptoms associated with the pathological condition afflicting the host, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the pathological condition being treated, such as serum triglyceride level, weight, total body fat content, etc. As such, treatment also includes situations where the pathological condition, or at least symptoms associated therewith, are completely inhibited, e.g. prevented from happening, or stopped, e.g. terminated, such that the host no longer suffers from the pathological condition, or at least the symptoms that characterize the pathological condition. For example, where the disease condition is marked by the presence of elevated lipid levels, treatment includes at least a reduction in the observed lipid levels, including a restoration of normal lipid levels. As another example, where the disease is obesity, treatment results ill at least a reduction in the overall weight and/or total body fat content of the host. [0120]
  • The subject methods also find use in the modulation of DGAT2α activity in hosts not suffering from a disease condition but in which the modulation of DGAT2α activity is nonetheless desired. For example, sperm production in males has been associated with diglyceride acyltransferase activity. As such, in males where at least reduced sperm production is desired, the subject methods can be used to reduce this target activity in such males, e.g. by administering an agent that reduces DGAT2α activity in such males, where such agents are described above. In other words, the subject methods provide a means of male contraception. Alternatively, where increased sperm count in a given male is desired, e.g. in those conditions where the male has reduced fertility, the subject methods can be used to enhance this target activity in the male and thereby increase sperm count and fertility, e.g. by administering to the male host a DGAT2α enhancing agent, as described above. [0121]
  • A variety of hosts are treatable according to the subject methods. Generally such hosts are “mammals” or “mammalian,” where these terms are used broadly to describe organisms which are within the class mammalia, including the orders carnivore (e.g., dogs and cats), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys). In many embodiments, the hosts will be humans. [0122]
  • Kits with unit doses of the active agent, usually in oral or injectable doses, are provided. In such kits, in addition to the containers containing the unit doses will be an informational package insert describing the use and attendant benefits of the drugs in treating pathological condition of interest. Preferred compounds and unit doses are those described herein above. [0123]
  • METHODS AND COMPOSITIONS FOR PRODUCING TRIGLYCERIDES AND TRIGLYCERIDE COMPOSITIONS
  • Also provided by the subject invention are methods for preparing triglycerides and triglyceride comprising compositions, as well as the compositions produced by these methods. In preparing triglycerides with the subject invention, at least the direct substrates of the desired triacylglyercol, e.g. diacylglycerol and fatty acyl CoA, will be combined in the presence of the polypeptide under conditions sufficient for the acylation of the diacylglycerol to occur. The synthesis may occur in an in vitro system, e.g. in a vessel in which the substrates or precursors thereof and the DGAT2α enzyme, as well as any other requisite enzymes (e.g. as need to convert the substrate precursors to substrates), or an in vivo system, e.g. a host cell that naturally comprises the substrates and into which a DGAT2α gene or nucleic acid according to the subject invention has been inserted in a manner sufficient for expression of the gene and provision of the DGAT2α enzyme, where the resultant triglyceride products may be separated from the host cell using standard separation techniques. [0124]
  • Of interest for use in producing triglyceride compositions are transgenic plants/fungi that have been genetically manipulated using the nucleic acid compositions of the subject invention to produce triglycerides and/or compositions thereof in one or more desirable ways. Transgenic plants/fungi of the subject invention are those plants/fungi that at least: (a) produce more triglyceride or triglyceride composition than wild type, e.g. produce more oil, such as by producing seeds having a higher oil content, as compared to wild-type; (b) produce triglyceride compositions, e.g. oils, that are enriched for triglycerides and/or enriched for one or more particular triglycerides as compared to wild type; and the like. Of interest are transgenic plants that produce commercially valuable triglyceride compositions or oils, such as canola, rapeseed, palm, corn, etc., containing various poly- and mono-unsaturated fatty acids, and the like. Of particular interest are transgenic plants, such as canola, rapeseed, palm, oil, etc., which have been genetically modified to produce seeds having higher oil content than the content found in the corresponding wild type, where the oil content of the seeds produced by such plants is at least 10% higher, usually at least 20% higher, and in many embodiments at least 30% higher than that found in the wild type, where in many embodiments seeds having oil contents that are 50% higher, or even greater, as compared to seeds produced by the corresponding wild-type plant, are produced. The seeds produced by such DGAT2α transgenic plants can be used as sources of oil or as sources of additional DGAT2α transgenic plants. Such transgenic plants and seeds therefore find use in methods of producing oils. In such methods, DGAT2α transgenic plants engineered to produce seeds having a higher oil content than the corresponding wild-type, e.g. seeds in which the DGAT2α gene is overexpressed, are grown, the seeds are harvested and then processed to recover the oil. The subject transgenic plants can also be used to produce novel oils characterized by the presence of triglycerides in different amounts and/or ratios than those observed in naturally occurring oils. The transgenic plants/fungi described above can be readily produced by those of skill in the art armed with the nucleic acid compositions of the subject invention. See the discussion on how to prepare transgenic plants, supra. [0125]
  • The triglyceride compositions described above find use in a variety of different applications. For example, such compositions or oils find use as food stuffs, being used as ingredients, spreads, cooking materials, etc. Alternatively, such oils find use as industrial feedstocks for use in the production of chemicals, lubricants, surfactants and the like. [0126]
  • Also of interest are transgenic non-human animals suitable for use as sources of food products and/or animal based industrial products. Such trans-genic non-human animals, e.g. transgenic mice, rats, livestock, such as cows, pigs, horses, birds, etc, may be produced using methods known in the art and reviewed supra. Such trans-genic non-human animals can be used for sources of a variety of different food and industrial products in which the triglyceride content is specifically tailored in a desirable manner. For example, such transgenic animals that have been modified in a manner such that DGAT2α activity is reduced as compared to the wild type can be used as sources of food products that are low in triglyceride content, e.g. low fat or lean meat products, low fat milk, low fat eggs, and the like. [0127]
  • The following examples are offered primarily for purposes of illustration. It will be readily apparent to those skilled in the art that the formulations, dosages, methods of administration, and other parameters of this invention may be further modified or substituted in various ways without departing from the spirit and scope of the invention. [0128]
  • EXPERIMENTAL
  • I. Existence of DGAT2α[0129]
  • A. Mice (DGAT1-/-) lacking DGAT, as described in WO 99/67268 are lean and resistant to diet-induced obesity, but are still capable of synthesizing triglycerides (TG) and have normal plasma TG levels. However, DGAT activity is virtually absent in membrane preparations from DGAT1-/- tissues (Smith et al., Nat. Genet. 2000 (25), 87-90). Using pulse assays in living cells, we measured that the residual TG synthesis activity in DGAT 1-/-Mouse Embryonic Fibroblasts (MEF) or adipocytes was about 40% of that in wild-type cells. The results are graphically depicted in FIGS. 1A and 1B. In FIG. 1A the membrane fraction isolated from MEF or adipocytes of wild-type or DGAT1-/- mice was used as the enzyme source in DGAT assays in vitro. In FIH. [0130] 1B living cells were pulse-labeled with [14C]oleic acid for 24 hours and [14C] incorporation in the TG fraction was measured.
  • In further assays, increased DGAT activity was observed in DGAT[0131] -1-membranes assayed without magnesium; and DGAT activity was observed to vary with magnesium concentration in liver and adipose tissue.
  • The above findings indicate the existence of DGAT2α, a second enzyme with diglyceride acyltransferase activity. [0132]
  • II. Mammalian DGAT2α Sequences [0133]
  • A. The human DGAT2α nucleic acid and amino acid sequences were identified using standard procedures, as described above. The human DGAT2α cDNA has the sequence appearing as SEQ ID NO:01, infra, while the protein encoded thereby has the sequence appearing as SEQ ID NO:02, infra. [0134]
  • B. The mouse DGAT2α nucleic acid and amino acid sequences were identified using standard procedures, as described above. The mouse DGAT2α cDNA has the sequence appearing as SEQ ID NO:03, infra, while the protein encoded thereby has the sequence appearing as SEQ ID NO:04, infra. [0135]
  • II. Characterization of DGAT2α[0136]
  • A. Molecular Weight [0137]
  • The mouse DGAT2α cDNA was determined to encode a 43 kD predicted protein based on the amino acid sequence. The mouse DGAT2α cDNA was determined to have no sequence homology to DGAT1, as described in Cases et al., supra. The mouse DGAT2α amino acid sequence was determined to have 2 putative N-linked glycosylation sites. The mouse DGAT2α amino acid sequence was determined to have 6 putative PKC phosphorylation sites.A Hydrophobicity plot assessed by Kyte-Doolittle (K-D) analysis revealed the existence of multiple putative transmembrane domains in the mouse DGAT2α amino acid sequence. FIG. 2 provides a graphical result of this analysis. As such, there are regions of higher hydrophobicity compatible with the existence of one or more transmembrane domain. [0138]
  • III. Further Characterization [0139]
  • A. Expression of DGAT2α in insect cells [0140]
  • Sf9 insect cells were infected with wild-type baculovirus, mouse FLAG-tagged DGAT2α or mouse FLAG-tagged DGAT1 (Cases et al., supra) recombinant baculoviruses, and the membrane fractions were assayed for DGAT activity. The results are graphically provided in FIG. 3A. In FIG. 3A a time course of DGAT2α virus infection is provided. Insect cell membranes were isolated at the indicated times after infection. Expression of the FLAG-tagged DGAT2α protein was detected by immunoblotting with an anti-FLAG antibody (Inset). DGAT activity was measured at low (5 mM) or high (100 mM) magnesium concentration, using [[0141] 14C]oleoyl CoA and cold diacylglycerol. The experiment was repeated three times and a representative experiment is shown. FIG. 3B shows that DGAT2α activity is dependent on the presence of the diacylglycerol substrate. Assays were performed at low magnesium concentration, using [14C]oleoyl CoA with or without exogenous cold diacylglycerol. When no diacylglycerol is added, no significant DGAT activity can be detected over background. Data represent the mean (±SD) of three experiments. To compare the DGAT activity of DGAT1 and DGAT2α, membranes expressing equal levels of DGAT1 or DGAT2α (as assessed by immunoblotting with an anti-FLAG antibody) were assayed at low magnesium concentration using increased amounts of cold oleoyl CoA in the presence of exogenous diacylglycerol. The results are provided in FIG. 3C. Lipids were extracted and separated by TLC and TG accumulation was visualized by charring and quantified by densitometry.
  • B. Analysis of DGAT2α mRNA expression mouse DGAT2α mRNA expression was analyzed. The results are provided in FIG. 4. [0142]
  • C. DGAT2α expression increases during 3T3-L1 adipocyte differentiation [0143]
  • Mouse 3T3-[0144] L 1 adipocyte differentiation was induced and mRNA were isolated at the indicated times shown in FIG. 5. Quantitation of DGAT2α mRNA levels in triplicate samples was perfromed by Phosphorimager analysis and corrected for loading relative to actin expression. The results are shown in FIG. 5.
  • IV. Summary of DGAT2α[0145]
  • mouse DGAT2α has no sequence homology to DGAT1 [0146]
  • mouse DGAT2α diacylglycerol acyltransferase activity inhibited by high magnesium concentrationsm; [0147]
  • mouse DGAT2α RNA expression in many tissues, highest levels found in liver, adipose tissue, and mammary gland [0148]
  • mouse DGAT2α markedly increased mRNA expression during 3T3-L1 adipocyte differentiation. [0149]
  • It is apparent from the above results and discussion that polynucleotides encoding mammalian DGAT2α enzymes, as well as novel polypeptides encoded thereby, are provided. The subject invention is important for both research and therapeutic applications. Using the DGAT2a probes of the subject invention, the role of DGAT2α and its regulation in a number of physiological processes can be studied in vivo. The subject invention also provides for important new ways of treating diseases associated with DGAT2α, such as hypertriglycemia and obesity, as well as in the production of tryglycerides. [0150]
  • All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. [0151]
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims. [0152]
  • 1 16 1 1231 DNA Homo sapiens 1 ttcagccatg aagaccctca tagccgccta ctccggggtc ctgcgcggcg agcgtcaggc 60 cgaggctgac cggagccagc gctctcacgg aggacccgtg tcgcgcgagg ggtctgggag 120 atggggcact ggatccagca tcctctccgc cctccaggac ctcttctctg tcacctggct 180 caataggtcc aaggtggaaa agcagctaca ggtcatctca gtgctccagt gggtcctgtc 240 cttccttgta ctgggagtgg cctgcagtgc catcctcatg tacatattct gcactgattg 300 ctggctcatc gctgtgctct acttcacttg gctggtgttt gactggaaca cacccaagaa 360 aggtggcagg aggtcacagt gggtccgaaa ctgggctgtg tggcgctact ttcgagacta 420 ctttcccatc cagctggtga agacacacaa cctgctgacc accaggaact atatctttgg 480 ataccacccc catggtatca tgggcctggg tgccttctgc aacttcagca cagaggccac 540 agaagtgagc aagaagttcc caggcatacg gccttacctg gctacactgg caggcaactt 600 ccgaatgcct gtgttgaggg agtacctgat gtctggaggt atctgccctg tcagccggga 660 caccatagac tatttgcttt caaagaatgg gagtggcaat gctatcatca tcgtggtcgg 720 gggtgcggct gagtctctga gctccatgcc tggcaagaat gcagtcaccc tgcggaaccg 780 caagggcttt gtgaaactgg ccctgcgtca tggagctgac ctggttccca tctactcctt 840 tggagagaat gaagtgtaca agcaggtgat cttcgaggag ggctcctggg gccgatgggt 900 ccagaagaag ttccagaaat acattggttt cgccccatgc atcttccatg gtcgaggcct 960 cttctcctcc gacacctggg ggctggtgcc ctactccaag cccatcacca ctgttgtggg 1020 agagcccatc accatcccca agctggagca cccaacccag caagacatcg acctgtacca 1080 caccatgtac atggaggccc tggtgaagct cttcgacaag cacaagacca agttcggcct 1140 cccggagact gaggtcctgg aggtgaactg agccagcctt cggggccaat tccctggagg 1200 aaccagctgc aaatcacttt tttgctctgt a 1231 2 388 PRT Homo sapiens 2 Met Lys Thr Leu Ile Ala Ala Tyr Ser Gly Val Leu Arg Gly Glu Arg 1 5 10 15 Gln Ala Glu Ala Asp Arg Ser Gln Arg Ser His Gly Gly Pro Ala Leu 20 25 30 Ser Arg Glu Gly Ser Gly Arg Trp Gly Thr Gly Ser Ser Ile Leu Ser 35 40 45 Ala Leu Gln Asp Leu Phe Ser Val Thr Trp Leu Asn Arg Ser Lys Val 50 55 60 Glu Lys Gln Leu Gln Val Ile Ser Val Leu Gln Trp Val Leu Ser Phe 65 70 75 80 Leu Val Leu Gly Val Ala Cys Ser Ala Ile Leu Met Tyr Ile Phe Cys 85 90 95 Thr Asp Cys Trp Leu Ile Ala Val Leu Tyr Phe Thr Trp Leu Val Phe 100 105 110 Asp Trp Asn Thr Pro Lys Lys Gly Gly Arg Arg Ser Gln Trp Val Arg 115 120 125 Asn Trp Ala Val Trp Arg Tyr Phe Arg Asp Tyr Phe Pro Ile Gln Leu 130 135 140 Val Lys Thr His Asn Leu Leu Thr Thr Arg Asn Tyr Ile Phe Gly Tyr 145 150 155 160 His Pro His Gly Ile Met Gly Leu Gly Ala Phe Cys Asn Phe Ser Thr 165 170 175 Glu Ala Thr Glu Val Ser Lys Lys Phe Pro Gly Ile Arg Pro Tyr Leu 180 185 190 Ala Thr Leu Ala Gly Asn Phe Arg Met Pro Val Leu Arg Glu Tyr Leu 195 200 205 Met Ser Gly Gly Ile Cys Pro Val Ser Arg Asp Thr Ile Asp Tyr Leu 210 215 220 Leu Ser Lys Asn Gly Ser Gly Asn Ala Ile Ile Ile Val Val Gly Gly 225 230 235 240 Ala Ala Glu Ser Leu Ser Ser Met Pro Gly Lys Asn Ala Val Thr Leu 245 250 255 Arg Asn Arg Lys Gly Phe Val Lys Leu Ala Leu Arg His Gly Ala Asp 260 265 270 Leu Val Pro Ile Tyr Ser Phe Gly Glu Asn Glu Val Tyr Lys Gln Val 275 280 285 Ile Phe Glu Glu Gly Ser Trp Gly Arg Trp Val Gln Lys Lys Phe Gln 290 295 300 Lys Tyr Ile Gly Phe Ala Pro Cys Ile Phe His Gly Arg Gly Leu Phe 305 310 315 320 Ser Ser Asp Thr Trp Gly Leu Val Pro Tyr Ser Lys Pro Ile Thr Thr 325 330 335 Val Val Gly Glu Pro Ile Thr Ile Pro Lys Leu Glu His Pro Thr Gln 340 345 350 Gln Asp Ile Asp Leu Tyr His Thr Met Tyr Met Glu Ala Leu Val Lys 355 360 365 Leu Phe Asp Lys His Lys Thr Lys Phe Gly Leu Pro Glu Thr Glu Val 370 375 380 Leu Glu Val Asn 385 3 1167 DNA Mus musculus misc_feature 1137 n = A,T,C or G 3 atgaagaccc tcatcgccgc ctactccggg gtcctgcggg gtgagcgtcg ggcggaagct 60 gcccgcagcg aaaacaagaa taaaggatct gccctgtcac gcgaggggtc tgggcgatgg 120 ggcactggct ccagcatcct ctcagccctc caagacatct tctctgtcac ctggctcaac 180 agatcyaagg tggaaaaaca gctgcaggtc atctcagtac tacaatgggt cctatccttc 240 ctggtgctag gagtggcctg cagtgtcatc ctcatgtaca ccttctgcac agactgctgg 300 ctgatagctg tgctctactt cacctggctg gcatttgact ggaacacgcc caagaaaggt 360 ggcaggagat cgcagtgggt gcgaaactgg gccgtgtggc gctacttccg agactacttt 420 cccatccagc tggtgaagac acacaacctg ctgaccacca ggaactatat ctttggatac 480 cacccccatg gcatcatggg cctgggtgcc ttctgtaact tcagcacaga ggctactgaa 540 gtcagcaaga agtttcctgg cataaggccc tatttggcta cgttggcygg taacttccgg 600 atgcctgtgc ttcgcgagta cctgatgtct ggaggcatct gccctgtcaa ccgagacacc 660 atagactact tgctctccaa gaatgggagt ggcaatgcta tcatcatcgt ggtgggaggt 720 gcagctgagt ccctgagctc catgcctggc aagaacgcag tcaccctgaa gaaccgcaaa 780 ggctttgtga agctggccct gcgccatgga gctgatctgg ttcccactta ttcctttgga 840 gagaatgagg tatacaagca ggtgatcttt gaggagggtt cctggggccg atgggtccag 900 aagaagttcc agaagtatat tggtttcgcc ccctgcatct tccatggccg aggcctcttc 960 tcctctgaca cctgggggct ggtgccctac tccaagccca tcaccaccgt cgtgggggag 1020 cccatcactg tccccaagct ggagcacccg acccagaaag acatcgacct gtaccatgcc 1080 atgtacatgg aggccctggt gaagctcttt gacaatcaca agaccaaatt tggcctncca 1140 gagactgagg tgctggaggt gaactga 1167 4 387 PRT Mus musculus 4 Met Lys Thr Leu Ile Ala Ala Tyr Ser Gly Val Leu Arg Gly Glu Arg 1 5 10 15 Arg Ala Glu Leu Pro Ala Ala Lys Asn Lys Asn Lys Gly Ser Ala Leu 20 25 30 Ser Arg Glu Gly Ser Gly Arg Trp Gly Thr Gly Ser Ser Ile Leu Ser 35 40 45 Ala Leu Gln Asp Ile Phe Ser Val Thr Trp Leu Asn Arg Ser Lys Val 50 55 60 Glu Lys Gln Leu Gln Val Ile Ser Val Leu Gln Trp Val Leu Ser Phe 65 70 75 80 Leu Val Leu Gly Val Ala Cys Ser Val Ile Leu Met Tyr Thr Phe Cys 85 90 95 Thr Asp Cys Trp Leu Ile Ala Val Leu Tyr Phe Thr Trp Leu Ala Phe 100 105 110 Asp Trp Asn Thr Pro Lys Lys Gly Gly Arg Arg Ser Gln Trp Val Arg 115 120 125 Asn Trp Ala Val Trp Arg Tyr Phe Arg Asp Tyr Phe Pro Ile Gln Leu 130 135 140 Val Lys Thr His Asn Leu Leu Thr Thr Arg Asn Tyr Ile Phe Gly Tyr 145 150 155 160 His Pro His Gly Ile Met Gly Leu Gly Ala Phe Cys Asn Phe Ser Thr 165 170 175 Glu Ala Thr Glu Val Ser Lys Lys Phe Pro Gly Ile Arg Pro Tyr Leu 180 185 190 Ala Thr Leu Ala Gly Asn Phe Arg Met Pro Val Leu Arg Glu Tyr Leu 195 200 205 Met Ser Gly Gly Ile Cys Leu Val Asn Arg Asp Thr Ile Asp Tyr Leu 210 215 220 Leu Ser Lys Asn Gly Ser Gly Asn Ala Ile Ile Ile Val Val Gly Gly 225 230 235 240 Ala Ala Glu Ser Leu Ser Ser Met Pro Gly Lys Asn Ala Val Thr Leu 245 250 255 Lys Asn Arg Lys Gly Phe Val Lys Leu Ala Leu Arg His Gly Ala Asp 260 265 270 Leu Val Pro Thr Tyr Ser Phe Gly Glu Asn Glu Val Tyr Lys Gln Val 275 280 285 Ile Phe Glu Glu Gly Ser Trp Gly Arg Trp Val Lys Lys Phe Gln Lys 290 295 300 Tyr Ile Gly Phe Ala Pro Cys Ile Phe His Gly Arg Gly Leu Phe Ser 305 310 315 320 Ser Asp Thr Trp Gly Leu Val Pro Tyr Ser Lys Pro Ile Thr Thr Val 325 330 335 Val Gly Glu Pro Ile Thr Val Pro Lys Leu Glu His Pro Thr Gln Lys 340 345 350 Asp Ile Asp Leu Tyr His Ala Met Tyr Met Glu Ala Leu Val Lys Leu 355 360 365 Phe Asp Asn His Lys Thr Lys Phe Gly Leu Pro Glu Thr Glu Val Leu 370 375 380 Glu Val Asn 385 5 1008 DNA Mus musculus 5 atgatggtcg agttcgcgcc actcaacacc ccgctggcac ggtgcctaca gaccgctgcg 60 gtgctgcagt gggtcctgtc cttcctcctg ctcgtgcagg tgtgcattgg aattatggtg 120 atgctggtcc tgtacaacta ttggttcctt tacatcccat atctggtctg gttttactat 180 gactggagaa ccccagagca aggaggcaga agatggaact gggtccaaag ctggcctgtg 240 tggaagtatt ttaaggagta ttttccaatc tgtcttgtca aaacgcagga tttggatccg 300 ggtcacaatt atatatttgg gtttcaccct catggaatat tcgtgcctgg agcctttgga 360 aatttttgta caaaatactc ggacttcaag aagctatttc ctggctttac atcgtatctc 420 cacgtggcca agatctggtt ctgtttcccg ttgttccgag aatatctgat gagtaacggg 480 ccggtttcag tgtctaagga gagtttgtct catgtgctga gcaaggatgg aggtggcaat 540 gtctcaatca ttgtcctcgg aggtgcaaag gaggcgctgg aggctcaccc aggaacattc 600 accctgtgca tccgccagcg caaagggttt gttaagatgg ccttgaccca tggtgccagt 660 ttggttccag tattttcttt tggtgaaaat gatctatata agcaaattaa caaccccaaa 720 ggctcctggc tacgaactat acaagacgca atgtatgatt caatgggagt agccttgcca 780 ctgatatatg ccagaggaat tttccagcac tactttggca taatgcccta tcggaagctg 840 atctacactg ttgttggccg ccctatccct gttcagcaga ttctgaaccc gacctcagag 900 cagattgaag agctgcatca gacataccta gaggagctaa agaaactatt caatgaacac 960 aaagggaaat atgggattcc ggagcacgaa actctggtat ttaaataa 1008 6 335 PRT Mus musculus 6 Met Met Val Glu Phe Ala Pro Leu Asn Thr Pro Leu Ala Arg Cys Leu 1 5 10 15 Gln Thr Ala Ala Val Leu Gln Trp Val Leu Ser Phe Leu Leu Leu Val 20 25 30 Gln Val Cys Ile Gly Ile Met Val Met Leu Val Leu Tyr Asn Tyr Trp 35 40 45 Phe Leu Tyr Ile Pro Tyr Leu Val Trp Phe Tyr Tyr Asp Trp Arg Thr 50 55 60 Pro Glu Gln Gly Gly Arg Arg Trp Asn Trp Val Gln Ser Trp Pro Val 65 70 75 80 Trp Lys Tyr Phe Lys Glu Tyr Phe Pro Ile Cys Leu Val Lys Thr Gln 85 90 95 Asp Leu Asp Pro Gly His Asn Tyr Ile Phe Gly Phe His Pro His Gly 100 105 110 Ile Phe Val Pro Gly Ala Phe Gly Asn Phe Cys Thr Lys Tyr Ser Asp 115 120 125 Phe Lys Lys Leu Phe Pro Gly Phe Thr Ser Tyr Leu His Val Ala Lys 130 135 140 Ile Trp Phe Cys Phe Pro Leu Phe Arg Glu Tyr Leu Met Ser Asn Gly 145 150 155 160 Pro Val Ser Val Ser Lys Glu Ser Leu Ser His Val Leu Ser Lys Asp 165 170 175 Gly Gly Gly Asn Val Ser Ile Ile Val Leu Gly Gly Ala Lys Glu Ala 180 185 190 Leu Glu Ala His Pro Gly Thr Phe Thr Leu Cys Ile Arg Gln Arg Lys 195 200 205 Gly Phe Val Lys Met Ala Leu Thr His Gly Ala Ser Leu Val Pro Val 210 215 220 Phe Ser Phe Gly Glu Asn Asp Leu Tyr Lys Gln Ile Asn Asn Pro Lys 225 230 235 240 Gly Ser Trp Leu Arg Thr Ile Gln Asp Ala Met Tyr Asp Ser Met Gly 245 250 255 Val Ala Leu Pro Leu Ile Tyr Ala Arg Gly Ile Phe Gln His Tyr Phe 260 265 270 Gly Ile Met Pro Tyr Arg Lys Leu Ile Tyr Thr Val Val Gly Arg Pro 275 280 285 Ile Pro Val Gln Gln Ile Leu Asn Pro Thr Ser Glu Gln Ile Glu Glu 290 295 300 Leu His Gln Thr Tyr Leu Glu Glu Leu Lys Lys Leu Phe Asn Glu His 305 310 315 320 Lys Gly Lys Tyr Gly Ile Pro Glu His Glu Thr Leu Val Phe Lys 325 330 335 7 1129 DNA Homo sapiens 7 cgtgggtgca ggctgcagtg gctggcgccg tcctcgcccg gccaggccat gaaggtagag 60 tttgcaccgc tcaacatcca gctggcgcgg cggctgcaga cggtggccgt gctgcagtgg 120 gtcctttctt ttcttacagg gccgatgtcc attggaatca ctgtgatgct gatcatacac 180 aactatttgt tcctttacat cccttatttg atgtggcttt actttgactg gcatacccca 240 gagcgaggag gcaggagatc cagctggatc aaaaattgga ctctttggaa acactttaag 300 gactattttc caattcatct tatcaaaact caagatttgg atccaagtca caactatata 360 tttgggtttc acccccatgg aataatggca gttggagcct ttgggaattt ttctgtaaat 420 tattctgact tcaaggacct gtttcctggc tttacttcat atcttcacgt gctgccactt 480 tggttctggt gtcctgtctt tcgagaatat gtgatgagtg ttgggctggt ttcagtttcc 540 aagaaaagtg tgtcctacat ggtaagcaag gagggaggtg gaaacatctc tgtcattgtc 600 cttgggggtg caaaagaatc actggatgct catcctggaa agttcactct gttcatccgc 660 cagcggaaag gatttgttaa aattgctttg acccatggcg cctctctggt cccagtggtt 720 tcttttggtg aaaatgaact gtttaaacaa actgacaacc ctgaaggatc atggattaga 780 actgttcaga ataaactgca gaagatcatg gggtttgctt tgcccctgtt tcatgccagg 840 ggagtttttc agtacaattt tggcctaatg acctatagga aagccatcca cactgttgtt 900 ggccgcccga tccctgttcg tcagactctg aacccgaccc aggagcagat tgaggagtta 960 catcagacct atatggagga acttaggaaa ttgtttgagg aacacaaagg aaagtatggc 1020 attccagagc acgagactct tgttttaaaa tgacttgact ataaaaaaaa attaaaaaat 1080 aaaaataaat gacttggctg taataaggca taaagaagga taagagacc 1129 8 334 PRT Homo sapiens 8 Met Lys Val Glu Phe Ala Pro Leu Asn Ile Gln Leu Ala Arg Arg Leu 1 5 10 15 Gln Thr Val Ala Val Leu Gln Trp Val Leu Ser Phe Leu Thr Gly Pro 20 25 30 Met Ser Ile Gly Ile Thr Val Met Leu Ile Ile His Asn Tyr Leu Phe 35 40 45 Leu Tyr Ile Pro Tyr Leu Met Trp Leu Tyr Phe Asp Trp His Thr Pro 50 55 60 Glu Arg Gly Gly Arg Arg Ser Ser Trp Ile Lys Asn Trp Thr Leu Trp 65 70 75 80 Lys His Phe Lys Asp Tyr Phe Pro Ile His Leu Ile Lys Thr Gln Asp 85 90 95 Leu Asp Pro Ser His Asn Tyr Ile Phe Gly Phe His Pro His Gly Ile 100 105 110 Met Ala Val Gly Ala Phe Gly Asn Phe Ser Val Asn Tyr Ser Asp Phe 115 120 125 Lys Asp Leu Phe Pro Gly Phe Thr Ser Tyr Leu His Val Leu Pro Leu 130 135 140 Trp Phe Trp Cys Pro Val Phe Arg Glu Tyr Val Met Ser Val Gly Leu 145 150 155 160 Val Ser Val Ser Lys Lys Ser Val Ser Tyr Met Val Ser Lys Glu Gly 165 170 175 Gly Gly Asn Ile Ser Val Ile Val Leu Gly Gly Ala Lys Glu Ser Leu 180 185 190 Asp Ala His Pro Gly Lys Phe Thr Leu Phe Ile Arg Gln Arg Lys Gly 195 200 205 Phe Val Lys Ile Ala Leu Thr His Gly Ala Ser Leu Val Pro Val Val 210 215 220 Ser Phe Gly Glu Asn Glu Leu Phe Lys Gln Thr Asp Asn Pro Glu Gly 225 230 235 240 Ser Trp Ile Arg Thr Val Gln Asn Lys Leu Gln Lys Ile Met Gly Phe 245 250 255 Ala Leu Pro Leu Phe His Ala Arg Gly Val Phe Gln Tyr Asn Phe Gly 260 265 270 Leu Met Thr Tyr Arg Lys Ala Ile His Thr Val Val Gly Arg Pro Ile 275 280 285 Pro Val Arg Gln Thr Leu Asn Pro Thr Gln Glu Gln Ile Glu Glu Leu 290 295 300 His Gln Thr Tyr Met Glu Glu Leu Arg Lys Leu Phe Glu Glu His Lys 305 310 315 320 Gly Lys Tyr Gly Ile Pro Glu His Glu Thr Leu Val Leu Lys 325 330 9 435 DNA Mus musculus misc_feature 341 n = A,T,C or G 9 ttacctccct cagggtcctg ggcatcatgt cttgctctat gaagactgaa cacttacaga 60 gtctgagcct tctgcagtgg cccttgagct acgttgccat gttttggatt gtgcagccat 120 tgttaatttg cctattgttc acacccttgt ggccgctacc aacagtttac tttgtctggt 180 tacttctcga ctggaagact ccagataaag gtggcaggcg ttcagactgg gtacggaact 240 ggaatgtctg gaaccacatc agggactatt tccccattac aatcctgaag actaaggacc 300 tgtcaccttc agagaactac atcatggggg tccaccccat nggtctcctg accttcggtg 360 ccttctgcaa cttctgcact gaggccacag gcttctcgaa gaccttccca ggcatcactc 420 ctcacttggc cacac 435 10 229 PRT Mus musculus 10 Met Lys Thr Glu His Leu Gln Ser Leu Ser Leu Leu Gln Trp Pro Leu 1 5 10 15 Ser Tyr Val Ala Met Phe Trp Ile Val Gln Pro Leu Leu Ile Cys Leu 20 25 30 Leu Phe Thr Pro Leu Trp Pro Leu Pro Thr Val Tyr Phe Val Trp Leu 35 40 45 Leu Leu Asp Trp Lys Thr Pro Asp Lys Gly Gly Arg Arg Ser Asp Trp 50 55 60 Val Arg Asn Trp Asn Val Trp Asn His Ile Arg Asp Tyr Phe Pro Ile 65 70 75 80 Thr Ile Leu Lys Thr Lys Asp Leu Ser Pro Ser Glu Asn Tyr Ile Met 85 90 95 Gly Val His Pro His Gly Leu Leu Thr Phe Gly Ala Phe Cys Asn Phe 100 105 110 Cys Thr Glu Ala Thr Gly Phe Ser Lys Thr Phe Pro Gly Ile Thr Pro 115 120 125 His Leu Ala Thr Leu Ser Trp Phe Phe Lys Ile Pro Ile Ile Arg Asp 130 135 140 Tyr Ile Met Ala Lys Gly Leu Cys Ser Val Ser Gln Ala Ser Ile Asp 145 150 155 160 Tyr Leu Leu Ser His Gly Thr Gly Asn Leu Val Gly Ile Pro Ile Ile 165 170 175 Thr Val Val Gly Glu Ala Leu Pro Leu Pro Gln Val Lys Asn Pro Ser 180 185 190 Pro Glu Ile Val Asp Lys Tyr His Ala Leu Tyr Met Asp Ala Leu Tyr 195 200 205 Lys Leu Phe Glu Gln His Lys Val Gln Tyr Gly Cys Ser Asn Thr Gln 210 215 220 Lys Leu Ile Phe Leu 225 11 1240 DNA Homo sapiens 11 atcaactcag cttaagaagt tttggccttc tggttaggct tcttgccaca acagaacagc 60 accataacca tggctttctt ctcccgactg aatctccagg agggcctcca aaccttcttt 120 gttttgcaat ggatcccagt ctatatattt ttagtttgga tcttgcagcc attgttcgtc 180 tacctgctgt ttacatcctt gtggccgcta ccagtgcttt actttgcctg gttgttcctg 240 gactggaaga ccccagagcg aggtggcagg cgttcggcct gggtaaggaa ctggtgtgtc 300 tggacccaca tcagggacta tttccccatt acgatcctga agacaaagga cctatcacct 360 gagcacaact acctcatggg ggttcacccc catggcctcc tgacctttgg cgccttctgc 420 aacttctgca ctgaggccac aggcttctcg aagaccttcc caggcatcac tcctcacttg 480 gccacgctgt cctggttctt caagatcccc tttgttaggg agtacctcat ggccaaaggt 540 gtgtgctctg tgagccagcc agccatcaac tatctgctga gccatggcac tggcaacctc 600 gtgggcattg tagtgggagg tgtgggtgag gccctgcaaa gtgtgcccaa caccaccacc 660 ctcatcctcc agaagcgcaa ggggttcgtg cgcacagccc tccagcatgg ggcatacctt 720 gtcccttcat attcctttgg tgagaacgaa gttttcaatc aggagacctt ccctgagggc 780 acgtggttaa ggttgttcca aaaaaccttc caggacacat tcaaaaaaat cctgggacta 840 aatttctgta ccttccatgg ccggggcttc actcgcggat cctggggctt cctgcctttc 900 aatcggccca ttaccactgt tgttggggaa ccccttccaa ttcccaggat taagaggcca 960 aaccagaaga cagtagacaa gtatcacgca ctctacatca gtgccctgcg caagctcttt 1020 gaccaacaca aagttgaata tggcctccct gagacccaag agctgacaat tacataacag 1080 gagccacatt ccccattgat caacccccaa agccatgagg gatccaagta gagccacaga 1140 aaaagaagaa ttccaggaga gggaaagatc gtaaggatga gagaggagac catccaagcc 1200 agaaattatt taataaatca gagttctagc aatagagtcc 1240 12 335 PRT Homo sapiens 12 Met Ala Phe Phe Ser Arg Leu Asn Leu Gln Glu Gly Leu Gln Thr Phe 1 5 10 15 Phe Val Leu Gln Trp Ile Pro Val Tyr Ile Phe Leu Val Trp Ile Leu 20 25 30 Gln Pro Leu Phe Val Tyr Leu Leu Phe Thr Ser Leu Trp Pro Leu Pro 35 40 45 Val Leu Tyr Phe Ala Trp Leu Phe Leu Asp Trp Lys Thr Pro Glu Arg 50 55 60 Gly Gly Arg Arg Ser Ala Trp Val Arg Asn Trp Cys Val Trp Thr His 65 70 75 80 Ile Arg Asp Tyr Phe Pro Ile Thr Ile Leu Lys Thr Lys Asp Leu Ser 85 90 95 Pro Glu His Asn Tyr Leu Met Gly Val His Pro His Gly Leu Leu Thr 100 105 110 Phe Gly Ala Phe Cys Asn Phe Cys Thr Glu Ala Thr Gly Phe Ser Lys 115 120 125 Thr Phe Pro Gly Ile Thr Pro His Leu Ala Thr Leu Ser Trp Phe Phe 130 135 140 Lys Ile Pro Phe Val Arg Glu Tyr Leu Met Ala Lys Gly Val Cys Ser 145 150 155 160 Val Ser Gln Pro Ala Ile Asn Tyr Leu Leu Ser His Gly Thr Gly Asn 165 170 175 Leu Val Gly Ile Val Val Gly Gly Val Gly Glu Ala Leu Gln Ser Val 180 185 190 Pro Asn Thr Thr Thr Leu Ile Leu Gln Lys Arg Lys Gly Phe Val Arg 195 200 205 Thr Ala Leu Gln His Gly Ala Tyr Leu Val Pro Ser Tyr Ser Phe Gly 210 215 220 Glu Asn Glu Val Phe Asn Gln Glu Thr Phe Pro Glu Gly Thr Trp Leu 225 230 235 240 Arg Leu Phe Gln Lys Thr Phe Gln Asp Thr Phe Lys Lys Ile Leu Gly 245 250 255 Leu Asn Phe Cys Thr Phe His Gly Arg Gly Phe Thr Arg Gly Ser Trp 260 265 270 Gly Phe Leu Pro Phe Asn Arg Pro Ile Thr Thr Val Val Gly Glu Pro 275 280 285 Leu Pro Ile Pro Arg Ile Lys Arg Pro Asn Gln Lys Thr Val Asp Lys 290 295 300 Tyr His Ala Leu Tyr Ile Ser Ala Leu Arg Lys Leu Phe Asp Gln His 305 310 315 320 Lys Val Glu Tyr Gly Leu Pro Glu Thr Gln Glu Leu Thr Ile Thr 325 330 335 13 1872 DNA Homo sapiens 13 aattcggctt actcactata gggctcgagc ggcccccggg caggtgccga cttcatttcc 60 aagtctgcac acaatgcagg cagtagccat gcctgacagc cacatgacag atactacacc 120 gctgaatgtg ctctaaccct ggacttggca ttgcccctac tgttgaggaa gcagtgcgtt 180 tttctccagt ctttcaggtc ccttcaccag ggaaccatta acttgtgcat cagaacaagg 240 acatttcctt acattcctgc aaacacagtc ctttcagttt actctttttt tgaggggggg 300 gcgcggggaa cggagtctcg ctctgtcgcc caggctggag tgcaatggtg caatctcagc 360 tcactgcaac ctctgcctcc caggtccaag cgattctcct gcctcagcct cccgggtagc 420 cgggactaca ggcgcctgcc accacgcccg gctaattttt gtatttttag tagagacgag 480 gtttcgccgt gttggcaggc tggtcttgga actcctgacc tcaggtgatt tactcgcctc 540 ggcctcccaa agtgctggga ttacaggcat gagccactgt gcccagtcac aagtttttat 600 tttagccatt ttgataagtg tgaagttccc tgatggctaa tgatgttcct ttttccatgt 660 gctcatttgt catctatgcc agagaagatt tggagaggag gacgtgaatt ggaggaaaac 720 tgttccagga ttccccacct ctggtggccc accgctggct cactgccatt gaccacactg 780 caggcagagc ctagtgcagt gctggagcag ggcccagaga ggagagggct tacagtgtga 840 attcagctca gctggggaag aagacacctt cccttctaga cctgaatcgg gttcccaagc 900 aaccactgtg attgctgtca acctctacct ggtggtgttc acaccatact ggcctgtcac 960 tgtgcttatt cttacctggc tggcttttga ctggaagacc cctcagcgag gcggccgccg 1020 gtttacctgt gtgaggcact ggcgcctgtg gaaacactac agcgattatt tccctctcaa 1080 gcttctgaag actcatgaca tctgccccag ccgcaactac atcctcgtct gccaccctca 1140 tgggctcttt gcccatggat ggtttggcca ctttgccaca gaggcctcag gcttctccaa 1200 gatatttcct ggcatcaccc cttacatact cacactggga gcctttttct ggatgccttt 1260 cctcagagaa tatgtaatgt ctacaggggc ctgctctgtg agtcgatcct ccattgactt 1320 tctgctgact cataaaggca caggcaacat ggtcattgtg gtgattggtg gactggctga 1380 gtgcagatac agcctgccag gttcttctac cctggtgttg aagaaccggt ctggctttgt 1440 gcgcatggcc cttcagcatg gggtgcctct aatacctgcc tatgcctttg gggagacgga 1500 cctctatgat cagcacattt tcactcctgg tggctttgtc aaccgcttcc agaagtggtt 1560 ccagagcatg gtacacatct acccttgtgc tttctatgga cgtggcttca ccaagaactc 1620 ctggggcctt ctgccctata gtcggcctgt aaccaccatc gtcggggagc ctctaccaat 1680 gcccaagatt gagaatccaa gccaggagat cgtggctaaa tatcacacac tctatattga 1740 tgccctacgt aaactgtttg accagcataa gaccaagttt ggtatctcag agacccagga 1800 gctggagata atttgacaga catccccagt agccttcacc ctggctggaa ggtatggatg 1860 gacccagtga ga 1872 14 333 PRT Homo sapiens 14 Met Leu Leu Pro Ser Lys Lys Asp Leu Lys Thr Ala Leu Asp Val Phe 1 5 10 15 Ala Val Phe Gln Trp Ser Phe Ser Ala Leu Leu Ile Thr Thr Thr Val 20 25 30 Ile Ala Val Asn Leu Tyr Leu Val Val Phe Thr Pro Tyr Trp Pro Val 35 40 45 Thr Val Leu Ile Leu Thr Trp Leu Ala Phe Asp Trp Lys Thr Pro Gln 50 55 60 Arg Gly Gly Arg Arg Phe Thr Cys Val Arg His Trp Arg Leu Trp Lys 65 70 75 80 His Tyr Ser Asp Tyr Phe Pro Leu Lys Leu Leu Lys Thr His Asp Ile 85 90 95 Cys Pro Ser Arg Asn Tyr Ile Leu Val Cys His Pro His Gly Leu Phe 100 105 110 Ala His Gly Trp Phe Gly His Phe Ala Thr Glu Ala Ser Gly Phe Ser 115 120 125 Lys Ile Phe Pro Gly Ile Thr Pro Tyr Ile Leu Thr Leu Gly Ala Phe 130 135 140 Phe Trp Met Pro Phe Leu Arg Glu Tyr Val Met Ser Thr Gly Ala Cys 145 150 155 160 Ser Val Ser Arg Ser Ser Ile Asp Phe Leu Leu Thr His Lys Gly Thr 165 170 175 Gly Asn Met Val Ile Val Val Ile Gly Gly Leu Ala Glu Cys Arg Tyr 180 185 190 Ser Leu Pro Gly Ser Ser Thr Leu Val Leu Lys Asn Arg Ser Gly Phe 195 200 205 Val Arg Met Ala Leu Gln His Gly Val Pro Leu Ile Pro Ala Tyr Ala 210 215 220 Phe Gly Glu Thr Asp Leu Tyr Asp Gln His Ile Phe Thr Pro Gly Gly 225 230 235 240 Phe Val Asn Arg Phe Gln Lys Trp Phe Gln Ser Met Val His Ile Tyr 245 250 255 Pro Cys Ala Phe Tyr Gly Arg Gly Phe Thr Lys Asn Ser Trp Gly Leu 260 265 270 Leu Pro Tyr Ser Arg Pro Val Thr Thr Ile Val Gly Glu Pro Leu Pro 275 280 285 Met Pro Lys Ile Glu Asn Pro Ser Gln Glu Ile Val Ala Lys Tyr His 290 295 300 Thr Leu Tyr Ile Asp Ala Leu Arg Lys Leu Phe Asp Gln His Lys Thr 305 310 315 320 Lys Phe Gly Ile Ser Glu Thr Gln Glu Leu Glu Ile Ile 325 330 15 1050 DNA Homo sapiens 15 ccacagcaga gctcacagaa cctgcgggag ccaggctgac ccgccagcat ggtagagttc 60 gcgcccttgt ttgtgccgtg ggagcgcagg ctgcagacac ttgctgtcct acagtttgtc 120 ttctccttct tggcactggg taagatctgc actgtgggct tcatagccct cctgtttaca 180 agattctggc tcctcactgt cctgtatgcg gcctggtggt atctggaccg agacaagcca 240 cggcaggggg gccggcacat ccaggccatc aggtgctgga ctatatggaa gtacatgaag 300 gactatttcc ccatccagct ggtcaagact gctgagctgg acccctctcg gaactacatt 360 gcgggcttcc acccccatgg agtcctggca gtcggagcct ttgccaacct gtgcactgag 420 agcacaggct tctcttcgat cttccccggt atccgccccc atctgatgat gctgaccttg 480 tggttccggg cccccttctt cagagattac atcatgtctg cagggttggt cacatcagaa 540 aaggagagtg ctgctcacat tctgaacagg aagggtggcg gaaacttgct gggcatcatt 600 gtagggggtg cccaggaggc cctggatgcc aggcctggat ccttcacgct gttactgcgg 660 aaccgaaagg gcttcgtcag gctcgccctg acacacgggg cacccctggt gccaatcttc 720 tccttcgggg agaatgacct atttgaccag attcccaact cttctggctc ctggttacgc 780 tatatccaga atcggttgca gaagatcatg ggcatctccc tcccactctt tcatggccgt 840 ggtgtcttcc agtacagctt tggtttaata ccctaccgcc ggcccatcac cactgtgggg 900 aagcccatcg aggtacagaa gacgctgcat ccctcggagg aggaggtgaa ccagctgcac 960 cagcattata tcaaagagct gtgcaacctc ttcgaggccc acaaacttaa gttcaacatc 1020 cctgctgacc agcacttgga gttctgctga 1050 16 333 PRT Homo sapiens 16 Met Val Glu Phe Ala Pro Leu Phe Val Pro Trp Glu Arg Arg Leu Gln 1 5 10 15 Thr Leu Ala Val Leu Gln Phe Val Phe Ser Phe Leu Ala Leu Gly Lys 20 25 30 Ile Cys Thr Val Gly Phe Ile Ala Leu Leu Phe Thr Arg Phe Trp Leu 35 40 45 Leu Thr Val Leu Tyr Ala Ala Trp Trp Tyr Leu Asp Arg Asp Lys Pro 50 55 60 Arg Gln Gly Gly Arg His Ile Gln Ala Ile Arg Cys Trp Thr Ile Trp 65 70 75 80 Lys Tyr Met Lys Asp Tyr Phe Pro Ile Gln Leu Val Lys Thr Ala Glu 85 90 95 Leu Asp Pro Ser Arg Asn Tyr Ile Ala Gly Phe His Pro His Gly Val 100 105 110 Leu Ala Val Gly Ala Phe Ala Asn Leu Cys Thr Glu Ser Thr Gly Phe 115 120 125 Ser Ser Ile Phe Pro Gly Ile Arg Pro His Leu Met Met Leu Thr Leu 130 135 140 Trp Phe Arg Ala Pro Phe Phe Arg Asp Tyr Ile Met Ser Ala Gly Leu 145 150 155 160 Val Thr Ser Glu Lys Glu Ser Ala Ala His Ile Leu Asn Arg Lys Gly 165 170 175 Gly Gly Asn Leu Leu Gly Ile Ile Val Gly Gly Ala Gln Glu Ala Leu 180 185 190 Asp Ala Arg Pro Gly Ser Phe Thr Leu Leu Leu Arg Asn Arg Lys Gly 195 200 205 Phe Val Arg Leu Ala Leu Thr His Gly Ala Pro Leu Val Pro Ile Phe 210 215 220 Ser Phe Gly Glu Asn Asp Leu Phe Asp Gln Ile Pro Asn Ser Ser Gly 225 230 235 240 Ser Trp Leu Arg Tyr Ile Gln Asn Arg Leu Gln Lys Ile Met Gly Ile 245 250 255 Ser Leu Pro Leu Phe His Gly Arg Gly Val Phe Gln Tyr Ser Phe Gly 260 265 270 Leu Ile Pro Tyr Arg Arg Pro Ile Thr Thr Val Gly Lys Pro Ile Glu 275 280 285 Val Gln Lys Thr Leu His Pro Ser Glu Glu Glu Val Asn Gln Leu His 290 295 300 Gln His Tyr Ile Lys Glu Leu Cys Asn Leu Phe Glu Ala His Lys Leu 305 310 315 320 Lys Phe Asn Ile Pro Ala Asp Gln His Leu Glu Phe Cys 325 330

Claims (23)

What is claimed is:
1. A mammalian polynucleotide present in other than its natural environment encoding a DGAT2α product.
2. The polynucleotide according to claim 2, wherein said polynucleotide comprises a sequence substantially similar or identical to a sequence selected from the group consisting of SEQ ID NO:01 and SEQ ID NO: 03.
3. A fragment of a polynucleotide according to claim 1.
4. A mammalian DGAT2α polypeptide present in other than its naturally occurring environment.
5. The polypeptide according to claim 4, wherein said polypeptide has an amino acid sequence that is substantially the same as or identical to a sequence selected from the group consisting of SEQ ID NO:02 and SEQ ID NO:04.
6. Substantially pure mammalian DGAT2α
7. Isolated mammalian DGAT2α.
8. A fragment of a polypeptide according to claim 4.
9. An expression cassette comprising a transcriptional initiation region functional in an expression host, a polynucleotide having a nucleotide sequence found in the nucleic acid according to claim 1 under the transcriptional regulation of said transcriptional initiation region, and a transcriptional termination region functional in said expression host.
10. A cell comprising an expression cassette according to claim 9 as part of an extrachromosomal element or integrated into the genome of a host cell as a result of introduction of said expression cassette into said host cell.
11. The cellular progeny of the cell according to claim 10.
12. A method of producing a DGAT2α polypeptide, said method comprising:
growing a cell according to claim 10, whereby said polypeptide is expressed; and
isolating said polypeptide substantially free of other proteins.
13. A monoclonal antibody binding specifically to a DGAT2α polypeptide.
14. The monoclonal antibody according to claim 13, wherein said antibody inhibits diglyceride acyltransferase activity of said polypeptide.
15. The monoclonal antibody according to claim 13, wherein said antibody is a humanized antibody.
16. A method for inhibiting the activity of a DGAT2α protein, said method comprising:
contacting said DGAT2α protein with an agent that inhibits the activity of said DGAT2α protein.
17. The method according to claim 16, wherein said agent is a small molecule.
18. The method according to claim 16, wherein said agent is an antibody.
19. The method according to claim 18, wherein said agent is a monoclonal antibody.
20. A method of modulating a symptom in a mammalian host of a disease condition associated with DGAT2α activity, said method comprising:
administering to said host a pharmaceutical composition comprising an effective amount of an active agent that modulates said DGAT2α activity in said host.
21. The method according to claim 20, wherein said symptom is hypertriglycemia.
22. The method according to claim 20, wherein said symptom is obesity
23. A method of producing a triacylglycerol, said method comprising:
contacting a diacylglyercol and fatty acyl CoA with a DGAT2α polypeptide under conditions sufficient to said triacylglycerol to be produced.
US09/794,715 2001-02-23 2001-02-26 Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha) Abandoned US20020119138A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US09/794,715 US20020119138A1 (en) 2001-02-26 2001-02-26 Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha)
US10/046,924 US7045326B2 (en) 2001-02-23 2002-01-14 Mono- and diacylglycerol acyltransferases and methods of use thereof
EP02780736A EP1373483A4 (en) 2001-02-23 2002-02-21 Mono- and diacylglycerol acyltransferases and methods of use thereof
PCT/US2002/005474 WO2002068595A2 (en) 2001-02-23 2002-02-21 Mono-and diacylglycerol acyltransferases and methods of use thereof
AU2002306573A AU2002306573A1 (en) 2001-02-23 2002-02-21 Mono-and diacylglycerol acyltransferases and methods of use thereof
US10/286,581 US20030161831A1 (en) 2001-02-23 2002-10-31 Mono-and diacylglycerol acyltransferases and methods of use thereof
US10/446,441 US7862819B2 (en) 2001-02-23 2003-05-27 Diacylglycerol O-acyltransferase 2α (DGAT2α)
US10/945,738 US20050106697A1 (en) 2001-02-23 2004-09-20 Mono-and diacylglycerol acyltransferases and methods of use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/794,715 US20020119138A1 (en) 2001-02-26 2001-02-26 Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha)

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US10/046,924 Continuation-In-Part US7045326B2 (en) 2001-02-23 2002-01-14 Mono- and diacylglycerol acyltransferases and methods of use thereof
US10/286,581 Continuation-In-Part US20030161831A1 (en) 2001-02-23 2002-10-31 Mono-and diacylglycerol acyltransferases and methods of use thereof
US10/446,441 Continuation US7862819B2 (en) 2001-02-23 2003-05-27 Diacylglycerol O-acyltransferase 2α (DGAT2α)
US10/446,441 Division US7862819B2 (en) 2001-02-23 2003-05-27 Diacylglycerol O-acyltransferase 2α (DGAT2α)

Publications (1)

Publication Number Publication Date
US20020119138A1 true US20020119138A1 (en) 2002-08-29

Family

ID=25163440

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/794,715 Abandoned US20020119138A1 (en) 2001-02-23 2001-02-26 Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha)
US10/446,441 Expired - Fee Related US7862819B2 (en) 2001-02-23 2003-05-27 Diacylglycerol O-acyltransferase 2α (DGAT2α)

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/446,441 Expired - Fee Related US7862819B2 (en) 2001-02-23 2003-05-27 Diacylglycerol O-acyltransferase 2α (DGAT2α)

Country Status (1)

Country Link
US (2) US20020119138A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003053363A2 (en) * 2001-12-19 2003-07-03 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (dgat2) family members and uses therefor
US20040107459A1 (en) * 2002-07-31 2004-06-03 Lardizabal Kathryn D. Diacylglycerol acyltransferase nucleic acid sequences and associated products
US20040209838A1 (en) * 2003-03-21 2004-10-21 Monia Brett P. Modulation of diacylglycerol acyltransferase 1 expression
US20050043524A1 (en) * 2003-08-18 2005-02-24 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20090054359A1 (en) * 2006-08-04 2009-02-26 Xing-Xian Yu Compositions and their uses directed to diacylglycerol acyltransferase 1
US11312962B2 (en) 2015-07-10 2022-04-26 Ionis Pharmaceuticals, Inc. Modulators of diacyglycerol acyltransferase 2 (DGAT2)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6100077A (en) * 1998-10-01 2000-08-08 The Trustees Of Columbia University In The City Of New York Isolation of a gene encoding diacylglycerol acyltransferase

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1003882A1 (en) 1997-06-05 2000-05-31 Calgene LLC Diacylglycerol acyl transferase proteins
DE19813835A1 (en) * 1998-03-20 1999-09-23 Metagen Gesellschaft Fuer Genomforschung Mbh Human nucleic acid sequences and protein products from normal breast tissue, useful for breast cancer therapy
EP1090026B1 (en) 1998-06-24 2008-08-27 The J. David Gladstone Institutes Diacylglycerol o-acyltransferase
US6344548B1 (en) 1998-06-24 2002-02-05 The Regents Of The University Of California Diacylglycerol o-acyltransferase
EP1098962B1 (en) 1998-07-02 2009-09-09 Calgene LLC Diacylglycerol acyl transferase proteins
JP2002522018A (en) 1998-07-24 2002-07-23 財団法人相模中央化学研究所 Human protein having hydrophobic domain and DNA encoding the same
NZ531664A (en) 1998-09-01 2005-07-29 Genentech Inc Pro1317 polypeptides and sequences thereof with homology to the semaphorin B glycoprotein family
AU1837600A (en) 1998-12-02 2000-06-19 E.I. Du Pont De Nemours And Company Plant diacylglycerol acyltransferases
AU2166400A (en) 1998-12-04 2000-06-19 Calgene Llc Diacylglycerol acyl transferase proteins
CN1352690A (en) 1998-12-17 2002-06-05 加拿大国家研究局 Diacylglycerol acyltransferase gene from plants
EP1165803B1 (en) 1999-04-01 2007-03-07 BASF Plant Science GmbH Enzymes of the biosynthetic pathway for the production of triacylglycerol and recombinant dna molecules encoding these enzymes
CA2372632C (en) 1999-05-04 2011-02-15 Basil S. Shorrosh Plant acyltransferases
US20030170691A1 (en) 2001-12-19 2003-09-11 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6100077A (en) * 1998-10-01 2000-08-08 The Trustees Of Columbia University In The City Of New York Isolation of a gene encoding diacylglycerol acyltransferase

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060183210A1 (en) * 2001-12-19 2006-08-17 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
WO2003053363A3 (en) * 2001-12-19 2004-04-29 Millennium Pharm Inc Human diacylglycerol acyltransferase 2 (dgat2) family members and uses therefor
US8334111B2 (en) 2001-12-19 2012-12-18 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US7910346B2 (en) 2001-12-19 2011-03-22 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US20100035339A1 (en) * 2001-12-19 2010-02-11 Gimeno Ruth E Human diacylglycerol acyltransferase 2 (dgat2) family members and uses therefor
US20090269332A1 (en) * 2001-12-19 2009-10-29 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
US7527962B2 (en) 2001-12-19 2009-05-05 Millenium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (DGAT2) family members and uses therefor
WO2003053363A2 (en) * 2001-12-19 2003-07-03 Millennium Pharmaceuticals, Inc. Human diacylglycerol acyltransferase 2 (dgat2) family members and uses therefor
EP1543130A4 (en) * 2002-07-31 2006-07-12 Monsanto Technology Llc Diacylglycerol acyltransferase nucleic acid sequences and associated products
US7935863B2 (en) 2002-07-31 2011-05-03 Monsanto Technology Llc Diacylglycerol acyltransferase nucleic acid sequences and associated products
US7417176B2 (en) 2002-07-31 2008-08-26 Monsanto Technology Llc Diacylglycerol acyltransferase nucleic acid sequences and associated products
US20090011113A1 (en) * 2002-07-31 2009-01-08 Lardizabal Kathryn D Diacylglycerol acyltransferase nucleic acid sequences and associated products
US20040107459A1 (en) * 2002-07-31 2004-06-03 Lardizabal Kathryn D. Diacylglycerol acyltransferase nucleic acid sequences and associated products
US7939714B2 (en) 2002-07-31 2011-05-10 Monsanto Technology Llc Diacylglycerol acyltransferase nucleic acid sequences and associated products
EP1543130A2 (en) * 2002-07-31 2005-06-22 Monsanto Technology, LLC Diacylglycerol acyltransferase nucleic acid sequences and associated products
US8158597B2 (en) 2003-03-21 2012-04-17 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 1 expression
US20040209838A1 (en) * 2003-03-21 2004-10-21 Monia Brett P. Modulation of diacylglycerol acyltransferase 1 expression
US7414033B2 (en) 2003-03-21 2008-08-19 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 1 expression
US20050272680A1 (en) * 2003-08-18 2005-12-08 Sanjay Bhanot Modulation of diacylglycerol acyltransferase 2 expression
EP2284267A2 (en) 2003-08-18 2011-02-16 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20100311810A1 (en) * 2003-08-18 2010-12-09 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20050043524A1 (en) * 2003-08-18 2005-02-24 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US7732590B2 (en) 2003-08-18 2010-06-08 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US8258289B2 (en) 2003-08-18 2012-09-04 Isis Pharmaceuticals, Inc Modulation of diacylglycerol acyltransferase 2 expression
US8883997B2 (en) 2003-08-18 2014-11-11 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US8003620B2 (en) 2006-08-04 2011-08-23 Isis Pharmaceuticals, Inc. Compositions and their uses directed to diacylglycerol acyltransferase 1
US20090054359A1 (en) * 2006-08-04 2009-02-26 Xing-Xian Yu Compositions and their uses directed to diacylglycerol acyltransferase 1
US8455456B2 (en) 2006-08-04 2013-06-04 Isis Pharmaceuticals, Inc. Compositions and their uses directed to diacylglycerol acyltransferase 1
US11312962B2 (en) 2015-07-10 2022-04-26 Ionis Pharmaceuticals, Inc. Modulators of diacyglycerol acyltransferase 2 (DGAT2)

Also Published As

Publication number Publication date
US20030202968A1 (en) 2003-10-30
US7862819B2 (en) 2011-01-04

Similar Documents

Publication Publication Date Title
US20030124126A1 (en) Mono-and diacylglycerol acyltransferases and methods of use thereof
CA2330549C (en) Diacylglycerol o-acyltransferase
US20030161831A1 (en) Mono-and diacylglycerol acyltransferases and methods of use thereof
EP1090026B1 (en) Diacylglycerol o-acyltransferase
US20030072757A1 (en) Diacylglycerol O-Acyltransferase
US20020119138A1 (en) Diacylglycerol o-acyltransferase 2alpha (DGAT2alpha)
US6869937B1 (en) Acyl CoA:cholesterol acyltransferase (ACAT-2)
US6380371B1 (en) Endoglycan: a novel protein having selectin ligand and chemokine presentation activity
US6649377B1 (en) Human aggrecanase and nucleic acid compositions encoding the same
AU741170B2 (en) Human intestinal Npt2B
WO2000027861A1 (en) Novel phosphodiesterase interacting proteins
US6852518B1 (en) Glycosyl sulfotransferases GST-4α, GST-4β, and GST-6
WO2000052030A1 (en) Rflat-1: a transcription factor that activates rantes gene expression

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENT OF THE UNIVERSITY OF CALIFORNIA, THE, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CASES, SYLVAINE;FARESE, ROBERT V.;STONE, SCOT;AND OTHERS;REEL/FRAME:012013/0954;SIGNING DATES FROM 20010626 TO 20010627

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:J. DAVID GLADSTONE INSTITUTES;REEL/FRAME:023638/0361

Effective date: 20091204