US20020129388A1 - Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis - Google Patents

Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis Download PDF

Info

Publication number
US20020129388A1
US20020129388A1 US10/023,529 US2352901A US2002129388A1 US 20020129388 A1 US20020129388 A1 US 20020129388A1 US 2352901 A US2352901 A US 2352901A US 2002129388 A1 US2002129388 A1 US 2002129388A1
Authority
US
United States
Prior art keywords
lbp
glu
seq
polypeptide
ala
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
US10/023,529
Inventor
Ann Lees
Robert Lees
Simon Law
Anibal Arjona
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.)
Boston Heart Foundation Inc
Original Assignee
Boston Heart Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/979,608 external-priority patent/US6355451B1/en
Priority claimed from US09/517,849 external-priority patent/US6605588B1/en
Application filed by Boston Heart Foundation Inc filed Critical Boston Heart Foundation Inc
Priority to US10/023,529 priority Critical patent/US20020129388A1/en
Publication of US20020129388A1 publication Critical patent/US20020129388A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/044Hyperlipemia or hypolipemia, e.g. dyslipidaemia, obesity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/323Arteriosclerosis, Stenosis

Definitions

  • LBPs low density lipoprotein
  • LDL low density lipoprotein
  • Atherosclerosis is the principal cause of heart attacks and strokes. It has been reported that about 50% of all deaths in the United States, Europe and Japan are due to atherosclerosis. Atherosclerotic lesions in the arterial wall characterize atherosclerosis. Cholesteryl esters (CE) are present in these atherosclerotic lesions. Low density lipoprotein (LDL) has been shown to be the major carrier of plasma CE, and has been implicated as the agent by which CE enter the atherosclerotic lesions.
  • LDL Low density lipoprotein
  • Scattered groups of lipid-filled macrophages are the first visible signs of atherosclerosis and are described as type I lesions. These macrophages are reported to contain CE derived from LDL. The macrophages recognize oxidized LDL, but not native LDL, and become foam cells by phagocytosing oxidized LDL. Larger, more organized collections of foam cells, fatty streaks, represent type II lesions. These lesions further develop into complex lesions called plaques, which can result in impeding the flow of blood in the artery.
  • LDL LDL-induced endothelial cells
  • LDL both native LDL and methylated LDL
  • LDL accumulates focally and irreversibly only at the edges of regenerating endothelial islands in aortic lesions, where functionally modified endothelial cells are present, but not in the centers of these islands where endothelial regeneration is completed.
  • LDL accumulates in human atherosclerotic lesions. The mechanism by which the LDL accumulates focally and irreversibly in arterial lesions has not heretofore been understood.
  • Still another object of the invention is to utilize an LBP (low density lipoprotein binding protein) gene and/or polypeptide, or fragments, analogs and variants thereof, to aid in the treatment, diagnosis and/or identification of therapeutic agents for atherosclerosis.
  • LBP low density lipoprotein binding protein
  • the invention features an isolated polynucleotide comprising a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8; SEQ ID NO: 9; SEQ ID NO: 43; SEQ ID NO: 44; SEQ ID NO: 47 or a polynucleotide capable of hybridizing to and which is at least about 95% identical to any of the above polynucleotides and wherein the encoded polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polynucleotides wherein the encoded polypeptide is capable of binding to LDL.
  • the polynucleotide comprises the nucleic acid sequence as set forth in SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17; SEQ ID NO: 18; SEQ ID NO: 45; SEQ ID NO: 46; SEQ ID NO: 48.
  • Another aspect of the invention is an isolated polypeptide comprising a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8; SEQ ID NO: 9; SEQ ID NO: 43; SEQ ID NO: 44; SEQ ID NO: 47; or a polypeptide which is at least about 95% identical to any of the above polypeptides and wherein the polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL.
  • Another aspect of the invention is a method for determining if an animal is at risk for atherosclerosis.
  • An animal is provided.
  • An aspect of LBP metabolism or structure is evaluated in the animal.
  • An abnormality in the aspect of LBP metabolism or structure is diagnostic of being at risk for atherosclerosis.
  • Another aspect of the invention is a method for evaluating an agent for use in treating atherosclerosis.
  • a test cell, cell-free system or animal is provided.
  • An agent is provided.
  • the agent is administered to the test cell, cell-free system or animal in a therapeutically effective amount.
  • the effect of the agent on an aspect of LBP metabolism or structure is evaluated.
  • a change in the aspect of LBP metabolism or structure is indicative of the usefulness of the agent in treating atherosclerosis.
  • Another aspect of the invention is a method for evaluating an agent for the ability to alter the binding of LBP polypeptide to a binding molecule, e.g., native LDL, modified LDL, e.g., methylated LDL or oxidized LDL, or an arterial extracellular matrix structural component.
  • An agent is provided.
  • An LBP polypeptide is provided.
  • a binding molecule is provided.
  • the agent, LBP polypeptide and binding molecule are combined.
  • the formation of a complex comprising the LBP polypeptide and binding molecule is detected.
  • An alteration in the formation of the complex in the presence of the agent as compared to in the absence of the agent is indicative of the agent altering the binding of the LBP polypeptide to the binding molecule.
  • Another aspect of the invention is a method for evaluating an agent for the ability to bind to an LBP polypeptide.
  • An agent is provided.
  • An LBP polypeptide is provided.
  • the agent is contacted with the LBP polypeptide.
  • the ability of the agent to bind to the LBP polypeptide is evaluated.
  • Another aspect of the invention is a method for evaluating an agent for the ability to bind to a nucleic acid encoding an LBP regulatory sequence.
  • An agent is provided.
  • a nucleic acid encoding an LBP regulatory sequence is provided.
  • the agent is contacted with the nucleic acid.
  • the ability of the agent to bind to the nucleic acid is evaluated.
  • Another aspect of the invention is a method for treating atherosclerosis in an animal.
  • An animal in need of treatment for atherosclerosis is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the animal in a therapeutically effective amount such that treatment of the atherosclerosis occurs.
  • the agent is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof.
  • the agent is a polypeptide of no more than about 100, 50, 30, 20, 10, 5, 4, 3 or 2 amino acid residues in length.
  • the agent is a polypeptide having an amino acid sequence that includes at least about 20%, 40%, 60%, 80%, 90%, 95% or 98% acidic amino acid residues.
  • Another aspect of the invention is a method for treating an animal at risk for atherosclerosis.
  • An animal at risk for atherosclerosis is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the animal in a therapeutically effective amount such that treatment of the animal occurs.
  • Another aspect of the invention is a method for treating a cell having an abnormality in structure or metabolism of LBP.
  • a cell having an abnormality in structure or metabolism of LBP is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the cell in a therapeutically effective amount such that treatment of the cell occurs.
  • Another aspect of the invention is a pharmaceutical composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier.
  • Another aspect of the invention is a vaccine composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier.
  • Another aspect of the invention is a method for diagnosing atherosclerotic lesions in an animal.
  • An animal is provided.
  • a labeled agent capable of binding to LBP, e.g., LBP-1, LBP-2 or LBP-3, present in atherosclerotic lesions is provided.
  • the labeled agent is administered to the animal under conditions which allow the labeled agent to interact with the LBP so as to result in labeled LBP.
  • the localization or quantification of the labeled LBP is determined by imaging so as to diagnose the presence of atherosclerotic lesions in the animal.
  • Another aspect of the invention is a method for immunizing an animal against an LBP, e.g., LBP-1, LBP-2 or LBP-3, or fragment or analog thereof.
  • An animal having LDL is provided.
  • the LBP or fragment or analog thereof is administered to the animal so as to stimulate antibody production by the animal to the LBP or fragment or analog thereof such that binding of the LBP to the LDL is altered, e.g., decreased or increased.
  • Another aspect of the invention is a method of making a fragment or analog of LBP polypeptide, the fragment or analog having the ability to bind to native LDL and to modified LDL, e.g., methylated LDL, oxidized LDL, acetylated LDL, or cyclohexanedione-treated LDL.
  • An LBP polypeptide is provided. The sequence of the LBP polypeptide is altered. The altered LBP polypeptide is tested for the ability to bind to modified LDL and native LDL.
  • Yet another aspect of the invention is a method for isolating a cDNA encoding an LBP.
  • a cDNA library is provided.
  • the cDNA library is screened for a cDNA encoding a polypeptide which binds to native LDL and modified LDL, e.g., methylated LDL or oxidized LDL.
  • the cDNA which encodes the polypeptide is isolated, the cDNA encoding an LBP.
  • FIG. 1 depicts the amino acid sequence of rabbit LBP-1 (SEQ ID NO: 1). Differences in amino acids between rabbit and human LBP-1 are depicted in bold type.
  • FIG. 2A depicts the nucleotide sequence (SEQ ID NO: 48) and amino acid sequence (SEQ ID NO: 47) of rabbit LBP-2.
  • FIG. 2B depicts a portion of the amino acid sequence of rabbit LBP-2 (SEQ ID NO: 2). Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. Where the sequences depicted in FIG. 2A and FIG. 2B differ, FIG. 2A represents the rabbit LBP-2 sequence.
  • FIG. 3 depicts the amino acid sequence of amino acids 319 to 350 of rabbit LBP-2 (SEQ ID NO: 3).
  • FIG. 4 depicts the amino acid sequence of amino acids 299 to 350 of rabbit LBP-2 (SEQ ID No: 4).
  • FIG. 5 depicts the amino acid sequence of rabbit LBP-3 (SEQ ID NO: 5). Differences in amino acids between rabbit and human LBP-3 are depicted in bold type.
  • FIG. 6 depicts the amino acid sequence of human LBP-1 (SEQ ID NO: 6). Differences in amino acids between rabbit and human LBP-1 are depicted in bold type.
  • FIG. 7A depicts the nucleotide sequence (SEQ ID NO: 45) and amino acid sequence (SEQ ID NO: 43) of human LBP-2.
  • FIG. 7B depicts the amino acid sequence of amino acids 322 to 538 of human LBP-2 (SEQ ID NO: 7). Differences in amino acids between rabbit and human LBP-2 are depicted in bold type.
  • FIG. 8A depicts the nucleotide sequence (SEQ ID NO: 46) and amino acid sequence (SEQ ID NO: 44) of human LBP-3.
  • FIG. 8B depicts the amino acid sequence of amino acids 17 to 546 of human LBP-3 (SEQ ID NO: 8). Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. Where the sequences depicted in FIG. 8A and FIG. 8B differ, FIG. 8A represents the human LBP-3 sequence.
  • FIG. 9 depicts the amino acid sequence of amino acids 14 to 33 of human or rabbit LBP-1, called BHF-1 (SEQ ID NO: 9).
  • FIG. 10 depicts the cDNA sequence encoding rabbit LBP-1 (SEQ ID NO: 10) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-1 are depicted in bold type.
  • FIG. 11 depicts a cDNA sequence encoding a portion of rabbit LBP-2 (SEQ ID NO: 11) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. Where the sequences depicted in FIG. 2A and FIG. 11 differ, FIG. 2A represents the rabbit LBP-2 sequence.
  • FIG. 12 depicts a cDNA sequence of nucleotides 256 to 1617 (SEQ ID NO: 12) of SEQ ID NO: 11 of rabbit LBP-2 and the corresponding amino acid sequence.
  • FIG. 13 depicts a cDNA sequence of nucleotides 196 to 1617 (SEQ ID NO: 13) of SEQ ID NO: 11 of rabbit LBP-2 and the corresponding amino acid sequence.
  • FIG. 14 depicts the cDNA sequence encoding rabbit LBP-3 (SEQ ID NO: 14) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-3 are depicted in bold type.
  • FIG. 15 depicts the cDNA sequence encoding human LBP-1 (SEQ ID NO: 15) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-1 are depicted in bold type.
  • FIG. 16 depicts a cDNA sequence encoding a portion of human LBP-2 (SEQ ID NO: 16) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-2 are depicted in bold type.
  • FIG. 17 depicts a cDNA sequence encoding a portion of human LBP-3 (SEQ ID NO: 17) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. Where the sequences depicted in FIG. 8A and FIG. 17 differ, FIG. 8A represents the human LBP-3 sequence.
  • FIG. 18 depicts the cDNA sequence encoding BHF-1 (SEQ ID NO: 18).
  • FIG. 19 corresponds to the amino acid sequence of rabbit LBP-1 (top sequence) in alignment with the amino acid sequence of human LBP-1 (bottom sequence).
  • FIG. 20 corresponds to the amino acid sequence of a portion of the amino acid sequence of rabbit LBP-2 (top sequence) in alignment with a portion of the amino acid sequence of human LBP-2 (bottom sequence).
  • FIG. 21 corresponds to the amino acid sequence of rabbit LBP-3 (top sequence) in alignment with the amino acid sequence of a portion of human LBP-3 (bottom sequence).
  • FIG. 22 depicts the genomic sequence of human LBP-1.
  • FIG. 23 depicts the genomic sequence of human LBP-2.
  • FIG. 24 depicts the genomic sequence of human LBP-3.
  • novel mature human and rabbit polypeptides LBP-1, LBP-2 and LBP-3, and biologically active analogs and fragments thereof, and there are provided isolated polynucleotides which encode such polypeptides.
  • LBP is an abbreviation for low density lipoprotein (LDL) binding protein.
  • polynucleotide, nucleotide and oligonucleotide are used interchangeably herein, and the terms polypeptides, proteins and peptides are used interchangeably herein.
  • This invention provides for an isolated polynucleotide comprising a polynucleotide encoding the polypeptide having the amino acid sequence of rabbit LBP-1 as set forth in FIG. 1 (SEQ ID NO: 1); rabbit LBP-2 as set forth in FIG. 2A (SEQ ID NO: 47); a portion of rabbit LBP-2 as set forth in FIG. 2B (SEQ ID NO: 2); 319 to 350 of rabbit LBP-2 as set forth in FIG. 3 (SEQ ID NO: 3); 299 to 350 of rabbit LBP-2 as set forth in FIG. 4 (SEQ ID NO: 4); rabbit LBP-3 as set forth in FIG. 5 (SEQ ID NO: 5); human LBP-1 as set forth in FIG.
  • SEQ ID NO: 9 SEQ ID NO: 9
  • This invention also includes an isolated polynucleotide comprising a polynucleotide encoding the polypeptide having amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) of human LBP-2 as set forth in FIG. 7A (SEQ ID NO: 43); amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) of rabbit or human LBP-I as set forth in FIG. 1 (SEQ ID NO: 1) and FIG.
  • polynucleotide capable of hybridizing to and which is at least about 80% identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to any of the above polynucleotides, and wherein the encoded polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polynucleotides wherein the encoded polypeptide is capable of binding to LDL.
  • polynucleotide encoding a polypeptide is meant a polynucleotide which includes only coding sequence for the polypeptide, as well as a polynucleotide which includes additional coding and/or non-coding sequences.
  • 1 - 9 may include only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequences 5′ and/or 3′ of the coding sequence for the mature polypeptide.
  • polynucleotides of the invention are also meant to include polynucleotides in which the coding sequence for the mature polypeptide is fused in the same reading frame to a polynucleotide sequence which aids in expression and/or secretion of a polypeptide from a host cell, e.g., a leader sequence.
  • the polynucleotides are also meant to include polynucleotides in which the coding sequence is fused in frame to a marker sequence which, e.g., allows for purification of the polypeptide.
  • the polynucleotides of the present invention may be in the form of RNA, DNA or PNA, e.g., cRNA, cDNA, genomic DNA, or synthetic DNA, RNA or PNA.
  • the DNA may be double-stranded or single stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
  • the polynucleotide comprises the nucleic acid of rabbit LBP-1 as set forth in FIG. 10 (SEQ ID NO: 10); rabbit LBP-2 as set forth in FIG. 2A (SEQ ID NO: 48) or FIG. 11 (SEQ ID NO: 11); nucleotide 256 to 1617 of SEQ ID NO: 11 of rabbit LBP-2 as set forth in FIG. 12 (SEQ ID NO: 12); nucleotide 196 to 1617 of SEQ ID NO: 11 of rabbit LBP-2 as set forth in FIG. 13 (SEQ ID NO: 13); rabbit LBP-3 as set forth in FIG. 14 (SEQ ID NO: 14); human LBP-1 as set forth in FIG.
  • the polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 30 SEQ ID NO:30 (GAAGAGGAAGAAGATGATGATGAAGATGAAGATGAAGAAGATGAT), SEQ ID NO:31 (GAAGAGGAAGAAGATGATGATGAAGATGAAGATGAAGAAGA TGAT GTGTCAGAGGGCTCTGAAGTGCCCGAGAGTGAC), SEQ ID NO:32 (GTGTCAGAGGGCTCTGAAGTGCCCGAGAGTGAC), SEQ ID NO:33 (GAGGATGATGACCCCGATGGCTTCTTAGGC), SEQ ID NO:34 (GTGGACGTGGATGAATATGACGAGAACAAGTTCGTGGACGAA GAAGATGGGGGCGACGGCCAGGCCGGGCCCGACGAGGGCGAGGTGGAC), SEQ ID NO:35 (GACGAGGGCGAGGTGGAC), SEQ ID NO:36 (GAGGAGGAGGAGGAGGAGGAGGAAGACGACGAGG
  • the coding sequence which encodes the mature polypeptide may be identical to the coding sequences shown in FIGS. 2A, 7A, 8 A and 10 - 18 (SEQ ID NOS: 10-18, 45, 46, and 48) or SEQ ID NOS: 30-40 or 42, or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptides as the DNA of FIGS. 2A, 7A, 8 A and 10 - 18 (SEQ ID NOS: 10-18, 45, 46, and 48) and SEQ ID NOS: 30-40 and 42.
  • This invention also includes recombinant vectors comprising the polynucleotides described above.
  • the vector can be, e.g., a plasmid, a viral particle or a phage.
  • the recombinant vector is an expression vector.
  • the vectors may also include various marker genes which are useful in identifying cells containing such vectors.
  • This invention also includes a cell comprising such a recombinant vector.
  • the recombinant vectors described herein can be introduced into a host cell, e.g., by transformation, transfection or infection.
  • This invention also includes a method for producing an LBP comprising culturing such a cell under conditions that permit expression of the LBP.
  • This invention also includes an isolated polypeptide comprising a polypeptide having the amino acid sequence as set forth in FIG. 1 (SEQ ID NO: 1); FIG. 2A (SEQ ID NO: 47); FIG. 2B (SEQ ID NO: 2); FIG. 3 (SEQ ID NO: 3); FIG. 4 (SEQ ID NO: 4); FIG. 5 (SEQ ID NO: 5); FIG. 6 (SEQ ID NO: 6); FIG. 7A (SEQ ID NO: 43); FIG. 7B (SEQ ID No: 7); FIG. 8A (SEQ ID NO: 44); FIG. 8B (SEQ ID NO: 8); or FIG.
  • SEQ ID NO: 9 SEQ ID NO: 9
  • a polypeptide which is at least about 80% identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to the above polypeptides, and wherein said polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL.
  • Differences in amino acids between the rabbit and human LBP-1, LBP-2 and LBP-3 genes are depicted in bold type in the figures. Differences in the amino acid sequences between rabbit and human LBP-1, LBP-2 and LBP-3 are also specifically shown in FIGS. 19, 20 and 21 , respectively.
  • This invention also includes an isolated polypeptide comprising a polypeptide having amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) as set forth in FIG. 7A (SEQ ID NO: 47); amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) as set forth in FIG. 1 (SEQ ID NO: 1) and FIG.
  • polypeptide 8A (SEQ ID NO: 8); or a polypeptide which is at least about 80%identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to the above polypeptides, and wherein said polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL.
  • polypeptides of the invention are meant to include, e.g., a naturally purified product, a chemically synthesized product, and a recombinantly derived product.
  • the polypeptides can be used, e.g., to bind to LDL, thereby inhibiting formation of atherosclerotic plaques.
  • the polypeptides can also be used, e.g., in gene therapy, by expression of such polypeptides in vivo.
  • the polypeptides can also be used in pharmaceutical or vaccine compositions.
  • the polypeptides can also be used as immunogens to produce antibodies thereto, which in turn, can be used as antagonists to the LBP polypeptides.
  • the LBPs provide the mechanism by which atherosclerosis is promoted through LDL oxidation.
  • the LBPs are believed to be required in order for focal, irreversible LDL binding to occur at the arterial wall, and that such binding is a critical early event in atherosclerosis because it allows the time necessary for LDL to be changed from its native state to a fully oxidized state.
  • LDL Since oxidized, but not native, LDL is a foreign protein, macrophages ingest it, first becoming the foam cells of type I lesions, and subsequently forming the fatty streaks of type II lesions.
  • This invention also includes a method for determining if an animal is at risk for atherosclerosis.
  • An animal is provided.
  • An aspect of LBP metabolism or structure is evaluated in the animal.
  • An abnormality in the aspect of LBP metabolism or structure is diagnostic of being at risk for atherosclerosis.
  • Atherosclerosis is meant a disease or condition which comprises several stages which blend imperceptibly into each other, including irreversible binding of LDL, LDL oxidation, macrophage recruitment, blockage of the artery and tissue death (infarction).
  • animal By animal is meant human as well as non-human animals.
  • Nonhuman animals include, e.g., mammals, birds, reptiles, amphibians, fish. insects and protozoa.
  • the nonhuman animal is a mammal, e.g., a rabbit, a rodent, e.g., a mouse, rat or guinea pig, a primate, e.g., a monkey, or a pig.
  • An animal also includes transgenic non-human animals.
  • transgenic animal is meant to include an animal that has gained new genetic information from the introduction of foreign DNA, i.e., partly or entirely heterologous DNA, into the DNA of its cells; or introduction of a lesion, e.g., an, in vitro induced mutation, e.g., a deletion or other chromosomal rearrangement into the DNA of its cells; or introduction of homologous DNA into the DNA of its cells in such a way as to alter the genome of the cell into which the DNA is inserted, e.g., it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout or replacement of the homologous host gene or results in altered and/or regulatable expression and/or metabolism of the gene.
  • the animal may include a transgene in all of its cells including germ line cells, or in only one or some of its cells.
  • Transgenic animals of the invention can serve as a model for studying atherosclerosis or for evaluating agents to treat atherosclerosis.
  • the determination for being at risk for atherosclerosis is done in a prenatal animal.
  • LBP low density lipoprotein
  • methylated LDL is meant that about 50% to about 90% of the lysine residues of LDL have a methyl group chemically attached. Methylated LDL is not recognized by previously reported cell surface receptors. See, eg., Weisgraber et al., J. Biol. Chem. 253: 9053-9062 (1978).
  • the LBP is also capable of binding oxidized LDL.
  • the binding of LDL to an LBP is irreversible.
  • the LBP does not transport the LDL to any intracellular compartment. Examples of LBPs are LBP-1, LBP-2 and LBP-3 described herein.
  • LBP metabolism is meant any aspect of the production, release, expression, function, action, interaction or regulation of LBP.
  • the metabolism of LBP includes modifications, e.g., covalent or non-covalent modifications, of LBP polypeptide.
  • the metabolism of LBP includes modifications, e.g., covalent or noncovalent modifications, that LBP induces in other substances.
  • the metabolism of LBP also includes changes in the distribution of LBP polypeptide, and changes LBP induces in the distribution of other substances.
  • LBP metabolism examples include the binding activity of LBP polypeptide to a binding molecule, e.g., LDL; the transactivation activity of LBP polypeptide on a target gene; the level of LBP protein; the level of LBP mRNA; the level of LBP modifications, e.g., phosphorylation, glycosylation or acylation; or the effect of LBP expression on transfected mammalian cell binding of LDL.
  • binding molecule is meant any molecule to which LBP can bind, e.g., a nucleic acid, e.g., a DNA regulatory region, a protein, e.g., LDL, a metabolite, a peptide mimetic, a non-peptide mimetic, an antibody, or any other type of ligand.
  • the aspect of LBP metabolism that is evaluated is the ability of LBP to bind to native LDL and/or methylated LDL and/or oxidized LDL. Binding to LDL can be shown, e.g., by antibodies against LDL, affinity chromatography, affinity coelectrophoresis (ACE) assays, or ELISA assays.
  • ACE affinity coelectrophoresis
  • LBP lipoprotein
  • an arterial extracellular matrix structural component that is evaluated.
  • examples of such components include proteoglycans, e.g., chondroitin sulfate proteoglycans and heparin sulfate proteoglycans; elastin; collagen; fibronectin; vitronectin; integrins; and related extracellular matrix molecules. Binding to arterial extracellular matrix structural components can be shown by standard methods known to those skilled in the art, e.g., by ELISA assays. Primary antibodies to the LBP are then added, followed by an enzyme-conjugated secondary antibody to the primary antibody, which produces a stable color in the presence of an appropriate substrate, and color development on the plates is measured in a microtiter plate reader.
  • proteoglycans e.g., chondroitin sulfate proteoglycans and heparin sulfate proteoglycans
  • elastin collagen
  • fibronectin vitr
  • Transactivation of a target gene by LBP can be determined, e.g., in a transient transfection assay in which the promoter of the target gene is linked to a reporter gene, e.g., ⁇ -galactosidase or luciferase, and co-transfected with an LBP expression vector.
  • a reporter gene e.g., ⁇ -galactosidase or luciferase
  • LBP expression vector e.g., ⁇ -galactosidase or luciferase
  • levels of LBP protein, mRNA or phosphorylation can be measured, e.g., in a sample, e.g., a tissue sample, e.g., arterial wall, by standard methods known to those skilled in the art.
  • an aspect of LBP structure is evaluated, e.g., LBP gene structure or LBP protein structure.
  • LBP gene structure or LBP protein structure For example, primary, secondary or tertiary structures can be evaluated.
  • the DNA sequence of the gene is determined and/or the amino acid sequence of the protein is determined. Standard cloning and sequencing methods can be used as are known to those skilled in the art.
  • the binding activity of an antisense nucleic acid with the cellular LBP mRNA and/or genomic DNA is determined using standard methods known to those skilled in the art so as to detect the presence or absence of the target mRNA or DNA sequences to which the antisense nucleic acid would normally specifically bind.
  • the risk for atherosclerosis that is determined can be a reduced risk or an increased risk as compared to a normal animal.
  • an abnormality which would give a reduced risk is an inactive LBP polypeptide.
  • An abnormality which would give an increased risk would be, e.g., an LBP polypeptide that has higher activity, e.g., LDL binding activity, than native LBP polypeptide.
  • the invention also includes a method for evaluating an agent for use in treating atherosclerosis.
  • a test cell, cell-free system or animal is provided.
  • An agent is provided.
  • the agent is administered to the test cell, cell-free system or animal in a therapeutically effective amount.
  • the effect of the agent on an aspect of LBP metabolism or structure is evaluated.
  • a change in the aspect of LBP metabolism or structure is indicative of the usefulness of the agent in treating atherosclerosis.
  • the method employs two phases for evaluating an agent for use in treating atherosclerosis, an initial in vitro phase and then an in vivo phase.
  • the agent is administered to the test cell or cell-free system in vitro, and if a change in an aspect of LBP metabolism occurs, then the agent is further administered to a test animal in a therapeutically effective amount and evaluated in vivo for an effect of the agent on an aspect of LBP metabolism.
  • cell is meant a cell or a group of cells, or a cell that is part of an animal.
  • the cell can be a human or non-human cell.
  • Cell is also meant to include a transgenic cell.
  • the cell can be obtained, e.g., from a culture or from an animal.
  • Animals are meant to include, e.g., natural animals and non-human transgenic animals.
  • the transgenic cell or nonhuman transgenic animal has an LBP transgene, or fragment or analog thereof.
  • the transgenic cell or non-human transgenic animal has a knockout for the LBP gene.
  • the test cell, cell-free system or animal can have a wild type pattern or a non-wild type pattern of LBP metabolism.
  • a non-wild type pattern of LBP metabolism can result, e.g., from under-expression, over-expression, no expression, or a temporal, site or distribution change.
  • Such a non-wild type pattern can result, e.g., from one or more mutations in the LBP gene, in a binding molecule gene, a regulatory gene. or in any other gene which directly or indirectly affects LBP metabolism.
  • a mutation is meant to include, e.g., an alteration, e.g., in gross or fine structure, in a nucleic acid.
  • Examples include single base pair alterations, e.g., missense or nonsense mutations, frameshifts, deletions, insertions and translocations. Mutations can be dominant or recessive. Mutations can be homozygous or heterozygous. Preferably, an aspect of LBP-1, LBP-2 or LBP-3 metabolism is evaluated.
  • An agent is meant to include, e.g., any substance, e.g., an anti-atherosclerosis drug.
  • the agent of this invention preferably can change an aspect of LBP metabolism. Such change can be the result of any of a variety of events, including, e.g., preventing or reducing interaction between LBP and a binding molecule, e.g., LDL or an arterial extracellular matrix structural component; inactivating LBP and/or the binding molecule, e.g., by cleavage or other modification; altering the affinity of LBP and the binding molecule for each other; diluting out LBP and/or the binding molecule; preventing expression of LBP and/or the binding molecule; reducing synthesis of LBP and/or the binding molecule; synthesizing an abnormal LBP and/or binding molecule; synthesizing an alternatively spliced LBP and/or binding molecule; preventing or reducing proper conformational folding of LBP and/or the binding molecule; modulating the binding
  • agents include LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof, a nucleic acid encoding LBP polypeptide or a biologically active fragment or analog thereof; a nucleic acid encoding an LBP regulatory sequence or a biologically active fragment or analog thereof; a binding molecule for LBP polypeptide; a binding molecule for LBP nucleic acid, the LBP nucleic acid being, e.g., a nucleic acid comprising a regulatory region for LBP or a nucleic acid comprising a structural region for LBP or a biologically active fragment of LBP; an antisense nucleic acid; a mimetic of LBP or a binding molecule; an antibody for LBP or a binding molecule; a metabolite; or an inhibitory carbohydrate or glycoprotein.
  • the agent is an antagonist, agonist or super agonist.
  • the agent is a natural ligand for LBP. In certain embodiments, the agent is an artificial ligand for LBP.
  • analog is meant a compound that differs from naturally occurring LBP in amino acid sequence or in ways that do not involve sequence, or both.
  • Analogs of the invention generally exhibit at least about 80% homology, preferably at least about 90% homology, more preferably yet at least about 95% homology, and most preferably at least about 98% homology, with substantially the entire sequence of a naturally occurring LBP sequence, preferably with a segment of about 100 amino acid residues, more preferably with a segment of about 50 amino acid residues, more preferably yet with a segment of about 30 amino acid residues, more preferably yet with a segment of about 20 amino acid residues, more preferably yet with a segment of about 10 amino acid residues, more preferably yet with a segment of about 5 amino acid residues, more preferably yet with a segment of about 4 amino acid residues, more preferably yet with a segment of about 3 amino acid residues, and most preferably with a segment of about 2 amino acid residues.
  • Non-sequence modifications include, e.g., in vivo or in vitro chemical derivatizations of LBP.
  • Non-sequence modifications include, e.g., changes in phosphorylation, acetylation, methylation, carboxylation, or glycosylation. Methods for making such modifications are known to those skilled in the art.
  • phosphorylation can be modified by exposing LBP to phosphorylation-altering enzymes, e.g., kinases or phosphatases.
  • Preferred analogs include LBP or biologically active fragments thereof whose sequences differ from the wild-type sequence by one or more conservative amino acid substitutions or by one or more non-conservative amino acid substitutions, deletions, or insertions which do not abolish LBP biological activity.
  • Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics, e.g., substitutions within the following groups: valine, glycine; glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. Other examples of conservative substitutions are shown in Table 1.
  • Amino acid sequence variants of a protein can be prepared by any of a variety of methods known to those skilled in the art. For example, random mutagenesis of DNA which encodes a protein or a particular domain or region of a protein can be used, e.g., PCR mutagenesis (using, e.g., reduced Taq polymerase fidelity to introduce random mutations into a cloned fragment of DNA; Leung et al., BioTechnique 1: 11-15 (1989)), or saturation mutagenesis (by, e.g., chemical treatment or irradiation of single-stranded DNA in vitro, and synthesis of a complementary DNA strand; Mayers et al., Science 229: 242 (1985)).
  • PCR mutagenesis using, e.g., reduced Taq polymerase fidelity to introduce random mutations into a cloned fragment of DNA; Leung et al., BioTechnique 1: 11-15 (1989)
  • Random mutagenesis can also be accomplished by, e.g., degenerate oligonucleotide generation (using, e.g., an automatic DNA synthesizer to chemically synthesize degenerate sequences; Narang, Tetrahedron 39: 3 (1983); Itakura et al., Recombinant DNA, Proc. 3rd Cleveland Sympos. Macromolecules, ed. A. G. Walton, Amsterdam: Elsevier, pp. 273-289 (1981)).
  • Non-random or directed mutagenesis can be used to provide specific sequences or mutations in specific regions.
  • variants which include, e.g., deletions, insertions, or substitutions, of residues of the known amino acid sequence of a protein.
  • the sites for mutation can be modified individually or in series, e.g., by (i) substituting first with conserved amino acids and then with more radical choices depending upon results achieved, (ii) deleting the target residue, (iii) inserting residues of the same or a different class adjacent to the located site, or (iv) combinations of the above.
  • analogs can be made by in vitro DNA sequence modifications of the sequences of FIGS. 2A, 7A, 8 A, 10 - 18 (SEQ ID NOS: 10-18, 45, 46, and 48).
  • in vitro mutagenesis can be used to convert any of these DNA sequences into a sequence which encodes an analog in which one or more amino acid residues has undergone a replacement, e.g., a conservative replacement as described in Table 1.
  • Methods for identifying desirable mutations include, e.g., alanine scanning mutagenesis (Cunningham and Wells, Science 244: 1081-1085 (1989)), oligonucleotide-mediated mutagenesis (Adelman et al., DNA, 2: 183 (1983)); cassette mutagenesis (Wells et al., Gene 34: 315 (1985)), combinatorial mutagenesis, and phage display libraries (Ladner et al., PCT International Appln. No. WO88/06630).
  • the LBP analogs can be tested, e.g., for their ability to bind to LDL and/or to an arterial extracellular matrix component, as described herein.
  • analogs within the invention include, e.g., those with modifications which increase peptide stability. Such analogs may contain, e.g., one or more non-peptide bonds (which replace the peptide bonds) in the peptide sequence. Also included are, e.g.: analogs that include residues other than naturally occurring L-amino acids, e.g., D-amino acids or nonnaturally occurring or synthetic amino acids, e.g., ⁇ or ⁇ amino acids; and cyclic analogs.
  • Analogs are also meant to include peptides in which structural modifications have been introduced into the peptide backbone so as to make the peptide non-hydrolyzable. Such peptides are particularly useful for oral administration, as they are not digested.
  • Peptide backbone modifications include, e.g., modifications of the amide nitrogen, the ⁇ -carbon, the amide carbonyl, or the amide bond, and modifications involving extensions, deletions or backbone crosslinks.
  • the backbone can be modified by substitution of a sulfoxide for the carbonyl, by reversing the peptide bond, or by substituting a methylene for the carbonyl group. Such modifications can be made by standard procedures known to those skilled in the art.
  • An analog is also meant to include polypeptides in which one or more of the amino acid residues include a substituent group, or polypeptides which are fused with another compound, e.g., a compound to increase the half-life of the polypeptide, e.g., polyethylene glycol.
  • fragment is meant some portion of the naturally occurring LBP polypeptide.
  • the fragment is at least about 100 amino acid residues, more preferably at least about 50 amino acid residues, more preferably yet at least about 30 amino acid residues, more preferably yet at least about 20 amino acid residues, more preferably yet at least about 5 amino acid residues, more preferably yet at least about 4 amino acid residues, more preferably yet at least about 3 amino acid residues, and most preferably at least about 2 amino acid residues in length.
  • Fragments include, e.g., truncated secreted forms, proteolytic fragments, splicing fragments, other fragments, and chimeric constructs between at least a portion of the relevant gene, e.g., LBP-1, LBP-2 or LBP-3, and another molecule. Fragments of LBP can be generated by methods known to those skilled in the art. In certain embodiments, the fragment is biologically active. The ability of a candidate fragment to exhibit a biological activity of LBP can be assessed by methods known to those skilled in the art. For example, LBP fragments can be tested for their ability to bind to LDL and/or to an arterial extracellular matrix structural component, as described herein. Also included are LBP fragments containing residues that are not required for biological activity of the fragment or that result from alternative mRNA splicing or alternative protein processing events.
  • Fragments of a protein can be produced by any of a variety of methods known to those skilled in the art, e.g., recombinantly, by proteolytic digestion, or by chemical synthesis.
  • Internal or terminal fragments of a polypeptide can be generated by removing one or more nucleotides from one end (for a terminal fragment) or both ends (for an internal fragment) of a nucleic acid which encodes the polypeptide.
  • Expression of the mutagenized DNA produces polypeptide fragments. Digestion with “end-nibbling” endonucleases can thus generate DNAs which encode an array of fragments.
  • DNAs which encode fragments of a protein can also be generated; e.g., by random shearing, restriction digestion or a combination of the above-discussed methods.
  • fragments of LBP can be made by expressing LBP DNA which has been manipulated in vitro to encode the desired fragment, e.g., by restriction digestion of any of the DNA sequences of FIGS. 2A, 7A, 8 A, 10 - 18 (SEQ ID NOS: 10-18, 45, 46, and 48).
  • Fragments can also be chemically synthesized using techniques known in the art, e.g., conventional Merrifield solid phase f-Moc or t-Boc chemistry for example, peptides of the present invention can be arbitrarily divided into fragments of desired length with no overlap of the fragments, or divided into overlapping fragments of a desired length.
  • LBP or a biologically active fragment or analog thereof, or a binding molecule or a biologically active fragment or analog thereof can, e.g., compete with its cognate molecule for the binding site on the complementary molecule, and thereby reduce or eliminate binding between LBP and the cellular binding molecule.
  • LBP or a binding molecule can be obtained, e.g., from purification or secretion of naturally occurring LBP or binding molecule, from recombinant LBP or binding molecule, or from synthesized LBP or binding molecule.
  • An agent can also be a nucleic acid used as an antisense molecule.
  • Antisense therapy is meant to include, e.g., administration or in situ generation of oligonucleotides or their derivatives which specifically hybridize, e.g., bind, under cellular conditions, with the cellular mRNA and/or genomic DNA encoding an LBP polypeptide, or mutant thereof, so as to inhibit expression of the encoded protein, e.g., by inhibiting transcription and/or translation.
  • the binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix.
  • the antisense construct binds to a naturally-occurring sequence of an LBP gene which, e.g., is involved in expression of the gene. These sequences include, e.g., promoter, start codons, stop codons, and RNA polymerase binding sites. In other embodiments, the antisense construct binds to a nucleotide sequence which is not present in the wild type gene. For example, the antisense construct can bind to a region of an LBP gene which contains an insertion of an exogenous, non-wild type sequence.
  • the antisense construct can bind to a region of an LBP gene which has undergone a deletion, thereby bringing two regions of the gene together which are not normally positioned together and which, together, create a non-wild type sequence.
  • antisense constructs which bind to non-wild type sequences provide the advantage of inhibiting the expression of a mutant LBP gene, without inhibiting expression of any wild type LBP gene.
  • an antisense construct of the present invention can be delivered, e.g., as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the cellular mRNA which encodes an LBP polypeptide.
  • the antisense construct is an oligonucleotide which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA (duplexing) and/or genomic sequences (triplexing) of an LBP gene.
  • Such oligonucleotides are preferably modified oligonucleotides which are resistant to endogenous nucleases, e.g.
  • nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate, phosphorodithioates and methylphosphonate analogs of DNA and peptide nucleic acids (PNA).
  • PNA peptide nucleic acids
  • mimetic is meant a molecule which resembles in shape and/or charge distribution LBP or a binding molecule.
  • the mimetic can be a peptide or a non-peptide.
  • Mimetics can act as therapeutic agents because they can, e.g., competitively inhibit binding of LBP to a binding molecule.
  • scanning mutagenesis e.g., alanine scanning mutagenesis, linker scanning mutagenesis or saturation mutagenesis
  • peptide mimetics e.g., diazepine or isoquinoline derivatives
  • Non-hydrolyzable peptide analogs of such residues can be generated using, e.g., benzodiazepine (see, e.g., Freidinger et al., in Peptides: Chemistry and Biology, G. R. Marshall ed., ESCOM Publisher: Leiden, Netherlands (1988)); azepine (see, e.g., Huffman et al., in Peptides: Chemistry and Biology, G. R. Marshall ed., ESCOM Publisher: Leiden, Netherlands (1988)); substituted gamma lactam rings (see, e.g., Garvey et al., in Peptides: Chemistry and Biology, G. R.
  • keto-methylene pseudopeptides see, eg., Ewenson et al., J. Med. Chem. 29: 295 (1986); Ewenson et al., in Peptides: Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co. Rockland, Ill. (1985)); ⁇ -turn dipeptide cores (see, e.g., Nagai et al., Tetrahedron Lett. 26: 647 (1985); Sato et al., J. Chem. Soc. Perkin Trans.
  • Antibodies are meant to include antibodies against any moiety that directly or indirectly affects LBP metabolism.
  • the antibodies can be directed against, e.g., LBP or a binding molecule, or a subunit or fragment thereof.
  • antibodies include anti-LBP-1, LBP-2 or LBP-3 antibodies; and anti-binding molecule antibodies.
  • Antibody fragments are meant to include, e.g., Fab fragments, Fab′ fragments, F(ab′) 2 fragments, F(v) fragments, heavy chain monomers, heavy chain dimers, heavy chain trimers, light chain monomers, light chain dimers, light chain trimers, dimers consisting of one heavy and one light chain, and peptides that mimic the activity of the anti-LBP or anti-binding molecule antibodies.
  • Fab 2 ′ fragments of the inhibitory antibody can be generated through, e.g., enzymatic cleavage.
  • Both polyclonal and monoclonal antibodies can be used in this invention. Preferably, monoclonal antibodies are used.
  • Natural antibodies, recombinant antibodies or chimeric-antibodies, e.g., humanized antibodies, are included in this invention.
  • humanized antibodies are used when the subject is a human.
  • the antibodies have a constant region derived from a human antibody and a variable region derived from an inhibitory mouse monoclonal antibody.
  • Production of polyclonal antibodies to LBP is described in Example 6.
  • Monoclonal and humanized antibodies are generated by standard methods known to those skilled in the art. Monoclonal antibodies can be produced, e.g., by any technique which provides antibodies produced by continuous cell lines cultures.
  • humanized antibodies are raised through conventional production and harvesting techniques (Berkower, I., Curr. Opin. Biotechnol. 7:622-628 (1996); Ramharayan and Skaletsky, Am. Biotechnol.
  • the antibodies are raised against the LBP, preferably the LDL-binding site, and the Fab fragments produced. These antibodies, or fragments derived therefrom, can be used, e.g., to block the LDL-binding sites on the LBP molecules.
  • Agents also include inhibitors of a molecule that are required for synthesis, post-translational modification, or functioning of LBP. and/or a binding molecule, or activators of a molecule that inhibits the synthesis or functioning of LBP and/or the binding molecule.
  • Agents include, e.g., cytokines, chemokines, growth factors, hormones, signaling components, kinases, phosphatases, homeobox proteins, transcription factors, editing factors, translation factors and post-translation factors or enzymes.
  • Agents are also meant to include ionizing radiation, non-ionizing radiation, ultrasound and toxic agents which can, e.g., at least partially inactivate or destroy LBP and/or the binding molecule.
  • An agent is also meant to include an agent which is not entirely LBP specific.
  • an agent may alter other genes or proteins related to arterial plaque formation. Such overlapping specificity may provide additional therapeutic advantage.
  • the invention also includes the agent so identified as being useful in treating atherosclerosis.
  • the invention also includes a method for evaluating an agent for the ability to alter the binding of LBP polypeptide to a binding molecule.
  • An agent is provided.
  • An LBP polypeptide is provided.
  • a binding molecule is provided.
  • the agent, LBP polypeptide and binding molecule are combined.
  • the formation of a complex comprising the LBP polypeptide and binding molecule is detected.
  • An alteration in the formation of the complex in the presence of the agent as compared to in the absence of the agent is indicative of the agent altering the binding of the LBP polypeptide to the binding molecule.
  • the LBP polypeptide is LBP-1, LBP-2 or LBP-3.
  • a binding molecule include native LDL, modified LDL, e.g., methylated LDL or oxidized LDL, and arterial extracellular matrix structural components.
  • Altering the binding includes, e.g., inhibiting or promoting the binding.
  • the efficacy of the agent can be assessed, e.g., by generating dose response curves from data obtained using various concentrations of the agent. Methods for determining formation of a complex are standard and are known to those skilled in the art, e.g., affinity coelectrophoresis (ACE) assays or ELISA assays as described herein.
  • ACE affinity coelectrophoresis
  • the invention also includes the agent so identified as being able to alter the binding of an LBP polypeptide to a binding molecule.
  • the invention also includes a method for evaluating an agent for the ability to bind to an LBP polypeptide.
  • An agent is provided.
  • An LBP polypeptide is provided.
  • the agent is contacted with the LBP polypeptide.
  • the ability of the agent to bind to the LBP polypeptide is evaluated.
  • the LBP polypeptide is LBP-1, LBP-2 or LBP-3. Binding can be determined, e.g., by measuring formation of a complex by standard methods known to those skilled in the art, e.g., affinity coelectrophoresis (ACE) assays or ELISA assays as described herein.
  • ACE affinity coelectrophoresis
  • the invention also includes the agent so identified as being able to bind to LBP polypeptide.
  • the invention also includes a method for evaluating an agent for the ability to bind to a nucleic acid encoding an LBP regulatory sequence.
  • An agent is provided.
  • a nucleic acid encoding an LBP regulatory sequence is provided.
  • the agent is contacted with the nucleic acid.
  • the ability of the agent to bind to the nucleic acid is evaluated.
  • the LBP regulatory sequence is an LBP-1, LBP-2 or LBP-3 regulatory sequence.
  • Binding can be determined, e.g., by measuring formation of a complex by standard methods known to those skilled in the art, e.g., DNA mobility shift assays, DNase I footprint analysis Molecular Biology, The invention being able to bind sequence. (Ausubel et al., ed., Current Protocols in John Wiley & Sons, New York, N.Y., (1989)).
  • the invention also includes the agent so identified as to a nucleic acid encoding an LBP regulatory sequence.
  • the invention also includes a method for treating atherosclerosis in an animal.
  • An animal in need of treatment for atherosclerosis is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the animal in a therapeutically effective amount such that treatment of the atherosclerosis occurs.
  • the agent is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof.
  • the agent can be, e.g., the polypeptide as set forth in SEQ ID NOS: 1-9, 43, 44, and 47.
  • the agent is a polypeptide of no more than about 100 amino acid residues in length, more preferably of no more than about 50 amino acid residues, more preferably yet of no more than about 30 amino acid residues, more preferably yet of no more than about 20 amino acid residues, more preferably yet of no more than about 10 amino acid residues, more preferably yet of no more than about 5 amino acid residues, more preferably yet of no more than about 4 amino acid residues, more preferably yet of no more than about 3 amino acid residues, and most preferably of no more than about 2 amino acid residues.
  • the polypeptide includes at least about 20% acidic amino acid residues, more preferably yet at least about 40% acidic amino acid residues, more preferably yet at least about 60% acidic amino acid residues, more preferably yet at least about 80% acidic amino acid residues, more preferably yet at least about 90% acidic amino acid residues, more preferably yet at least about 95% acidic amino acid residues, and most preferably at least about 98% acidic amino acid residues.
  • Acidic amino acid residues include aspartic acid and glutamic acid.
  • An example of such an LBP poly-peptide is BHF-1, which is a 20 amino acid length fragment of human or rabbit LBP-1 which contains amino acid residues 14 through 33. See FIG. 9 (SEQ ID NO: 9). 45% of the amino acid residues of BHF-1 are acidic.
  • the invention also includes biologically active fragments and analogs of BHF-1.
  • Other preferred acidic regions from the LBPs are amino acid residues 329 through 343 (SEQ ID NO: 19), 329 through 354 (SEQ ID NO: 20), 344 through 354 (SEQ ID NO: 21), and 529 through 538 (SEQ ID NO: 22) of human LBP-2 as depicted in FIG. 7A (SEQ. ID NO: 43); amino acid residues 14 through 43 (SEQ ID NO: 23)and 38 through 43 (SEQ ID NO: 24) of rabbit or human LBP-1 as depicted in FIG. 1 (SEQ ID NO: 1) and FIG.
  • agents include homopolymers and heteropolymers of any amino acid or amino acid analog.
  • the agent is a homopolymer of an acidic amino acid or analog thereof.
  • the agent is a heteropolymer of one or more acidic amino acids and one or more other amino acids, or analogs thereof.
  • agents include poly(glu), poly(asp), poly(glu asp), poly(glu N), poly (asp N) and poly(glu asp N).
  • N is meant any amino acid, or analog thereof, other than glu or asp.
  • poly(glu asp) is meant all permutations of glu and asp for a given length peptide.
  • a preferred peptide is poly(glu) of no more than about 10 amino acids in length, preferably about 7 amino acids in length.
  • the agent is an LBP nucleic acid or a biologically active fragment or analog thereof, e.g., a nucleic acid encoding LBP-1, LBP-2 or LBP-3 polypeptide, or a biologically active fragment or analog thereof.
  • the agent can be, e.g., a nucleic acid comprising a nucleotide sequence as set forth in SEQ ID NOS: 10-18, 45, 46, and 48.
  • the agent is an antisense molecule, e.g., one which can bind to an LBP gene sequence.
  • Treating is meant to include, e.g., preventing, treating, reducing the symptoms of, or curing the atherosclerosis.
  • Administration of the agent can be accomplished by any method which allows the agent to reach the target area, e.g., a target cell or the extracellular matrix. These methods include, e.g., injection, deposition, implantation, suppositories, oral ingestion, inhalation, topical administration, or any other method of administration where access to the target area by the agent is obtained. Injections can be, e.g., intravenous, intradermal, subcutaneous, intramuscular or intraperitoneal.
  • Implantation includes inserting implantable drug delivery systems, e.g., microspheres, hydrogels, polymeric reservoirs, cholesterol matrices, polymeric systems, e.g., matrix erosion and/or diffusion systems and non-polymeric systems, e.g., compressed, fused or partially fused pellets.
  • Suppositories include glycerin suppositories.
  • Oral ingestion doses can be enterically coated.
  • Inhalation includes administering the agent with an aerosol in an inhalator, either alone or attached to a carrier that can be absorbed.
  • Administration of the agent can be alone or in combination with other therapeutic agents.
  • the agent can be combined with a suitable carrier, incorporated into a liposome, or incorporated into a polymer release system.
  • the administration can be designed so as to result in sequential exposures to the agent over some time period, e.g., hours, days, weeks, months or years. This can be accomplished by repeated administrations of the agent by one of the methods described above, or alternatively, by a controlled release delivery system in which the agent is delivered to the animal over a prolonged period without repeated administrations.
  • a controlled release delivery system is meant that total release of the agent does not occur immediately upon administration, but rather is delayed for some time. Release can occur in bursts or it can occur gradually and continuously.
  • Administration of such a system can be, e.g., by long acting oral dosage forms, bolus injections, transdermal patches or subcutaneous implants.
  • Examples of systems in which release occurs in bursts include, e.g., systems in which the agent is entrapped in liposomes which are encapsulated in a polymer matrix, the liposomes being sensitive to a specific stimulus, e.g., temperature, pH, light, magnetic field, or a degrading enzyme, and systems in which the agent is encapsulated by an ionically-coated microcapsule with a microcapsule core-degrading enzyme.
  • Examples of systems in which release of the agent is gradual and continuous include, e.g., erosional systems in which the agent is contained in a form within a matrix, and diffusional systems in which the agent permeates at a controlled rate, e.g., through a polymer.
  • Such sustained release systems can be, e.g., in the form of pellets or capsules.
  • the agent can be suspended in a liquid, e.g., in dissolved form or colloidal form.
  • the liquid can be a solvent, partial solvent or non-solvent. In many cases water or an organic liquid can be used.
  • the agent can be administered prior to or subsequent to the appearance of atherosclerosis symptoms.
  • the agent is administered to patients with familial histories of atherosclerosis, or who have phenotypes that may indicate a predisposition to atherosclerosis, or who have been diagnosed as having a genotype which predisposes the patient to atherosclerosis, or who have other risk factors, e.g., hypercholesterolemia, hypertension or smoking.
  • the agent is administered to the animal in a therapeutically effective amount.
  • therapeutically effective amount is meant that amount which is capable of at least partially preventing or reversing atherosclerosis.
  • a therapeutically effective amount can be determined on an individual basis and will be based, at least in part, on consideration of the species of animal, the animal's size, the animal's age, the agent used, the type of delivery system used, the time of administration relative to the onset of atherosclerosis symptoms, and whether a single, multiple, or controlled release dose regimen is employed.
  • a therapeutically effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • the concentration of the agent is at a dose of about 0.1 to about 1000 mg/kg body weight/day, more preferably at about 0.1 to about 500 mg/kg/day, more preferably yet at about 0.1 to about 100 mg/kg/day, and most preferably at about 0.1 to about 5 mg/kg/day.
  • the specific concentration partially depends upon the particular agent used, as some are more effective than others.
  • the dosage concentration of the agent that is actually administered is dependent at least in part upon the final concentration that is desired at the site of action, the method of administration, the efficacy of the particular agent, the longevity of the particular agent, and the timing of administration relative to the onset of the atherosclerosis symptoms.
  • the dosage form is such that it does not substantially deleteriously affect the animal.
  • the dosage can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • various gene constructs can be used as part of a gene therapy protocol to deliver nucleic acids encoding an agent, e.g., either an agonistic or antagonistic form of an LBP polypeptide.
  • agent e.g., either an agonistic or antagonistic form of an LBP polypeptide.
  • expression vectors can be used for in vivo transfection and expression of an LBP polypeptide in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of, LBP polypeptide in a cell in which non-wild type LBP is expressed.
  • Expression constructs of the LBP polypeptide, and mutants thereof may be administered in any biologically effective carrier, e.g., any formulation or composition capable of effectively delivering the LBP gene to cells in vivo.
  • Approaches include, e.g., insertion of the subject gene in viral vectors including, e.g., recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids.
  • viral vectors including, e.g., recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids.
  • Viral vectors infect or transduce cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectinTM (Life Technologies, Inc., Gaithersburg, Md.) or derivatized (e.g.
  • the agent is administered to arterial wall cells of the animal.
  • a genetically engineered LBP gene is administered to arterial wall cells.
  • administration is done in a prenatal animal or embryonic cell. It will be recognized that the particular gene construct provided for in vivo transduction of LBP expression is also useful for in vitro transduction of cells, such as for use in the diagnostic assays described herein.
  • therapy of atherosclerosis is performed with antisense nucleotide analogs of the genes which code for the LBPs.
  • the antisense nucleotides have non-hydrolyzable “backbones,” e.g., phosphorothioates, phosphorodithioates or methylphosphonates.
  • the nucleoside base sequence is complementary to the sequence of a portion of the gene coding for, e.g., LBP-1, 2 or 3. Such a sequence might be, e.g., ATTGGC if the gene sequence for the LBP is TAACCG.
  • an antisense analog of a portion of one of the LBP genes in a slowrelease medium, e.g., polyvinyl alcohol, which is administered, e.g., by subcutaneous injection, so as to release the antisense nucleotide analog over a period of weeks or months.
  • a slowrelease medium e.g., polyvinyl alcohol
  • the antisense analog is incorporated into a polymeric matrix, e.g., polyvinyl alcohol, such that the gel can be applied locally to an injured arterial wall to inhibit LBP synthesis and prevent LDL accumulation, e.g., after angioplasty or atherectomy.
  • the invention also includes a method for treating an animal at risk for atherosclerosis.
  • An animal at risk for atherosclerosis is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the animal in a therapeutically effective amount such that treatment of the animal occurs.
  • Being at risk for atherosclerosis can result from, e.g., a family history of atherosclerosis, or phenotypic symptoms which predispose to atherosclerosis, e.g., having hypercholesterolemia, hypertension or smoking.
  • the invention also includes a method for treating a cell having an abnormality in structure or metabolism of LBP.
  • a cell having an abnormality in structure or metabolism of LBP is provided.
  • An agent capable of altering an aspect of LBP structure or metabolism is provided.
  • the agent is administered to the cell in a therapeutically effective amount such that treatment of the cell occurs.
  • the cell is obtained from a cell culture or tissue culture or an embryo fibroblast.
  • the cell can be, e.g., part of an animal, e.g., a natural animal or a nonhuman transgenic animal.
  • the LBP is LBP-1, LBP-2 or LBP-3.
  • the invention also includes a pharmaceutical composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include, e.g., saline, liposomes and lipid emulsions.
  • the agent of the pharmaceutical composition is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof.
  • the agent can be, e.g., the polypeptide as set forth in SEQ ID NOS: 1-9, 43, 44, and 47.
  • the agent is a polypeptide of no more than about 100 amino acid residues in length, more preferably of no more than about 50 amino acid residues, more preferably yet of no more than about 30 amino acid residues, more preferably yet of no more than about 20 amino acid residues, more preferably yet of no more than about 10 amino acid residues, more preferably yet of no more than about 5 amino acid residues, more preferably yet of no more than about 4 amino acid residues, more preferably yet of no more than about 3 amino acid residues, and most preferably of no more than about 2 amino acid residues.
  • the polypeptide includes at least about 20% acidic amino acid residues, more preferably yet at least about 40% acidic amino acid residues, more preferably yet at least about 60% acidic amino acid residues, more preferably yet at least about 80% acidic amino acid residues, more preferably yet at least about 90% acidic amino acid residues, more preferably yet at least about 95% acidic amino acid residues, and most preferably at least about 98%acidic amino acid residues.
  • the agent is an LBP nucleic acid, e.g., a nucleic acid encoding LBP-1, LBP-2 or LBP-3 polypeptide, or a biologically active fragment or analog thereof.
  • the agent can be, e.g., a nucleic acid comprising a nucleotide sequence as set forth in SEQ ID NOS: 10-18, 45, 46, and 48.
  • the invention also includes a vaccine composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier.
  • the invention also includes a method for diagnosing atherosclerotic lesions in an animal.
  • An animal is provided.
  • a labeled agent capable of binding to LBP present in atherosclerotic lesions is provided.
  • the labeled agent is administered to the animal under conditions which allow the labeled agent to interact with the LBP so as to result in labeled LBP.
  • the localization or quantification of the labeled LBP is determined by imaging so as to diagnose the presence of atherosclerotic lesions in the animal.
  • the LBP is LBP-1, LBP-2 or LBP-3.
  • the imaging can be performed by standard methods known to those skilled in the art, including, e.g., magnetic resonance imaging, gamma camera imaging, single photon emission computed tomographic (SPECT) imaging, or positron emission tomography (PET).
  • SPECT single photon emission computed tomographic
  • PET positron emission tomography
  • agents that bind tightly to LBPs in atherosclerotic lesions are used for atherosclerotic imaging and diagnosis.
  • the agent is radiolabeled with, e.g., 99m Tc or another isotope suitable for clinical imaging by gamma camera, SPECT, PET scanning or other similar technology. Since LBPs occur in very early lesions, such imaging is more sensitive than angiography or ultrasound for locating very early lesions which do not yet impinge on the arterial lumen to cause a visible bulge or disturbed flow.
  • the imaging agents which bind to LBPs can also be used to follow the progress of atherosclerosis, as a means of evaluating the effectiveness of both dietary and pharmacological treatments.
  • a diagnostic embodiment of the invention is the adaptation of, e.g., a peptide complementary to one of the LBPs, by radiolabeling it and using it as an injectable imaging agent for detection of occult atherosclerosis.
  • the peptide is selected from those known to bind to LBPs, e.g., RRRRRRR or KKLKLXX, or any other polycationic peptide which binds to the highly electronegative domains of the LBPs.
  • technetium-binding ligands e.g., CGC, GGCGC, or GGCGCF
  • MRI magnetic resonance imaging
  • DTPA diethylene triamine penta-acetic acid
  • the LBP-binding peptides are covalently bound, e.g., to magnetic ion oxide particles by standard methods known to those skilled in the art, e.g., conjugating the peptides with activated polystyrene resin beads containing magnetic ion oxide.
  • the invention also includes a method for immunizing an animal against an LBP, e.g., LBP-1, LBP-2 or LBP-3, or fragment or analog thereof.
  • An animal having LDL is provided.
  • An LBP or fragment or analog thereof is provided.
  • the LBP or fragment or analog thereof is administered to the animal so as to stimulate antibody production by the animal to the LBP or fragment or analog thereof such that binding of the LBP to the LDL is altered, e.g., decreased or increased.
  • the invention also includes a method of making a fragment or analog of LBP polypeptide, the fragment or analog having the ability to bind to modified LDL and native LDL.
  • An LBP polypeptide is provided.
  • the sequence of the LBP polypeptide is altered.
  • the altered LBP polypeptide is tested for the ability to bind to modified LDL, e.g., methylated LDL, oxidized LDL, acetylated LDL, cyclohexanedione-treated LDL (CHD-LDL), and to native LDL.
  • the fragments or analogs can be generated and tested for their ability to bind to these modified LDLs and to native LDL, by methods known to those skilled in the art, e.g., as described herein. Preferably, they are tested for their ability to bind to methylated LDL and native LDL.
  • the binding activity of the fragment or analog can be greater or less than the binding activity of the native LBP. Preferably, it is greater.
  • the LBP is LBP-1, LBP-2 or LBP-3.
  • the invention also includes a method for isolating a cDNA encoding an LBP.
  • a cDNA library is provided.
  • the cDNA library is screened for a cDNA encoding a polypeptide which binds to native LDL and modified LDL, e.g., methylated LDL or oxidized LDL.
  • the cDNA which encodes this polypeptide is isolated, the cDNA encoding an LBP.
  • Atherosclerosis in a hyperlipidemic subject can be reduced following the generation of an immune response in the subject by immunization with LBPs.
  • Numerous immunotherapeutic products can be used to generate antibodies that will block the binding between LDL and LBPs.
  • LBP immunogens that can be used in the invention include human LBPs, non-human LBPs, recombinant LBPs, and proteins structurally related to the LBPs described herein, e.g. non-naturally occurring proteins that differ from a naturally occurring LBP at one or more amino acid residues.
  • injecting one or more peptides that include an LBP domain can generate an effective immune response.
  • a peptide comprising an LBP domain having LDL-binding activity can cause an organism to make antibodies to the LBP binding sites for LDL.
  • peptide immunogens can include sequences derived from human LBPs, non-human LBPs, recombinant LBPs, and proteins structurally related to the LBPs described herein.
  • Modifications can be made to a protein or peptide immunogen of the invention to increase its immunogenicity.
  • the immunogen can be conjugated or coupled with a carrier, e.g. a Cholera toxin B chain or monoclonal antibodies.
  • the immunogen can be precipitated with aluminum salts or cross-linked with formaldehyde or other aldehydes.
  • the protein may be mixed with a physiologically acceptable diluent such as water, phosphate buffered saline, or saline.
  • the composition may further include an adjuvant.
  • adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide are all well known in the art.
  • Adjustments in the adjuvant of the invention can be made to affect the immunogenicity of the peptide or protein.
  • modifications include using: aluminum salts; cytokines; MF59 (microfluidized emulsion of oil and surfactants); SAF-1 (oil-based emulsion); saponin derivatives; polymers (such as polyphosphazene); and bacterial toxins. Additional descriptions of antigenic protein-adjuvant combinations are described in WO 99/54452 (herein incorporated by reference) and WO 99/49890 (herein incorporated by reference).
  • the LBP immunogen can be delivered either directly as a protein antigen or alternatively as a nucleic acid that encodes the protein antigen.
  • the immunotherapeutic products of the invention can be delivered by numerous delivery routes. These include injection, deposition, implantation, suppositories, oral ingestion, inhalation (e.g., delivery via a nasal spray), and topical administration (e.g., delivery via a skin patch).
  • a nucleic acid encoding an immunogen of the invention can be directly administered, for example by injection, to tissues and expressed as a protein.
  • the DNA or RNA can be either associated with a delivery vehicle (e.g., viruses, bacteria, liposomes, and gold beads) or naked (free from association with transfection-facilitating proteins, viral particles, liposomal formulations, charged lipids and calcium phosphate precipitating).
  • the nucleic acid can optionally include a promoter, e.g. a viral promoter.
  • the immunogen encoded by the nucleic acid is produced in the host, resulting in the generation of an immune response.
  • nucleic acid sequences encoding therapeutic proteins and peptides are described in detail by Felgner et al. (U.S. Pat. No. 25 5,580,859; herein incorporated by reference) and Barbet et al. (U.S. Pat. No. 6,025,338:; herein incorporated by reference).
  • Vaccine compositions of viral liposomes comprising a nucleic acid, e.g. an RNA, encoding a protein antigen are described in WO 99/52503 (herein incorporated by reference).
  • Proteins and nucleic acids encoding peptides can also be delivered to an individual by their encapsulation in liposomes, microparticles, and ISCOMS, all of which are well known in the art (see, e.g., U.S. Pat. No. 6,013,258, herein incorporated by reference).
  • a nucleic acid encoding an immunogen of the invention can also be included in the genome of a plant, so as to result in the production of the immunogen by plant tissues.
  • the genetically modified plant may then consumed by an individual, resulting in the ingestion of the immunogen and the generation of an anti-LBP immune response.
  • Methodology for the generation and usage of edible plant vaccines is described in WO 99/54452 (herein incorporated by reference).
  • the edible vaccine can include a plant cell transformed with a nucleic acid construct comprising a promoter and a sequence encoding an LBP.
  • the sequence may optionally encode a chimeric protein, comprising a cholera toxin subunit B peptide fused to the LBP peptide.
  • Preferred plant promoters of the invention include CaMV 35S, patatin, mas, and granule-bound starch synthase promoters. Additional useful promoters and enhancers are described in WO 99/54452.
  • the edible vaccine of the invention can be administered to a mammal suffering from or at risk of atherosclerosis.
  • an edible vaccine is administered orally, e.g. consuming a transgenic plant of the invention.
  • the transgenic plant can be in the form of a plant part, extract, juice, liquid, powder, or tablet.
  • the edible vaccine can also be administered via an intranasal route.
  • Microorganisms e.g., attenuated viruses or bacteria, can be used in the invention by including a nucleic acid encoding an LBP immunogen in the genome of the microorganism.
  • This modified vector can then be delivered to a host, resulting in the in vivo production of the immunogen.
  • the immune response generated by these vectors is expected to result in anti-atherosclerotic immunity.
  • Nucleic acid molecules are inserted into microorganism genomes by standard methods known in the art (U.S. Pat. No. 5,866,136 and U.S. Pat. No. 6,025,164, both of which are herein incorporated by reference)
  • the anti-atherosclerotic methods of the invention are directed to treating a subject, e.g., a human, primate, horse, dog, cat, or goat, at risk for atherosclerosis by stimulating an anti-LBP response in the subject by immunotherapy.
  • a subject e.g., a human, primate, horse, dog, cat, or goat
  • the LBP proteins and peptides of the invention may be delivered to the subject by the numerous delivery systems described herein.
  • the immunotherapy may comprise an initial immunization followed by additional, e.g. one, two, or three, boosters.
  • the invention also includes a method of treating a subject at risk for atherosclerosis by (1) providing a subject at risk for atherosclerosis and (2) administering to the subject one or more of the following: (a) an LBP protein or fragment or analog thereof and an adjuvant; (b) a nucleic acid encoding an LBP protein; (c) a virus or bacteria comprising a nucleic acid encoding an LBP protein; and (d) an edible plant comprising a nucleic acid encoding an LBP protein.
  • the LBP protein used in this method can be any LBP described herein, e.g., LBP-1, LBP-2, or LBP-3.
  • a combination of more than one nucleic acid or LBP protein or fragment or analog thereof can be administered to the subject.
  • combinations of LBP proteins, or nucleic acids encoding LBP proteins include: (1) LBP-I and LBP-2; (2) LBP-I and LBP-3; (3)LBP-2 and LBP-3; and (4) LBP-1, LBP-2, and LBP-3.
  • This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis.
  • Also provided by the invention is a method of treating a subject at risk for atherosclerosis whereby a non-autologous LBP protein or a nucleic acid encoding a non-autologous LBP protein is delivered to the subject to generate an immune response to an autologous LBP.
  • this method entails identifying one or more autologous LBP proteins, e.g., LBP-1, LBP-2, or LBP-3, produced by the subject.
  • the identification can by, e.g., DNA sequence analysis, protein sequence analysis, antibody reactivity, hybridization analysis, or nucleic acid amplification.
  • a non-autologous LBP protein e.g., allogeneic, xenogeneic, or a genetically modified, non-naturally occurring protein that differs at one or more amino acid residues from the one or more LBP proteins
  • a nucleic acid encoding a non-autologous LBP protein is administered to the subject.
  • the anti-atherosclerotic effectiveness of this immunotherapeutic product is determined by its ability to induce an immune response against one or more autologous LBP proteins when administered to the subject. It is therefore expected that extensive differences between a non-autologous and autologous LBP protein will not result in cross immunoreactivity.
  • This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis.
  • Another method of the invention is a method of treating a subject at risk for atherosclerosis by increasing the levels of one or more LBP proteins circulating in the plasma.
  • LBP proteins include, e.g., allogeneic LBP, xenogeneic LBP, and genetically modified LBP.
  • the plasma levels of one or more LBP proteins can be increased by the delivery of a nucleic acid encoding an LBP protein. Because LBP generally does not normally occur as a circulating protein, the endogenous molecule is expected to be susceptible to immune recognition when delivered in a soluble form.
  • This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis.
  • compositions containing one or more LBP proteins, e.g., LBP-1, LBP-2, or LBP-3, mixed with an adjuvant, suitable for use in humans.
  • the pharmaceutical composition can contain a combination of more than one LBP protein.
  • compositions can include any of the following: (1) LBP-1 and LBP-2; (2) LBP-I and LBP-3; (3) LBP-2 and LBP-3; and (4) LBP-1, LBP-2, and LBP-3.
  • a cell therapy system whereby a cell expressing an LBP is delivered to a subject at risk for atherosclerosis.
  • This cell can be engineered to express either an autologous or non-autologous LBP protein or peptide of the invention. Delivery of this engineered cell to a subject results in the in vivo production of an LBP protein and the associated immunotherapy produced when either the protein or a nucleic acid encoding the protein is provided to an individual.
  • Cell therapy methods are described in U.S. Pat. No. 5,955,095 (herein incorporated by reference).
  • This example illustrates the construction of a rabbit cDNA library using mRNA from balloon-deendothelialized healing rabbit abdominal aorta.
  • Balloon-catheter deendothelialized rabbit aorta has been shown to be a valid model for atherosclerosis (Minick et al., Am. J. Pathol. 95:131-158 (1979).
  • RNA was obtained four weeks after ballooning to maximize focal LDL binding in the ballooned rabbit aorta.
  • First strand cDNA synthesis was carried out in a 50 ⁇ l reaction mixture containing 4 ⁇ g mRNA; 2 ⁇ g oligo d(T)primer; methylation dNTP mix (10 mM each); 10 mM DTT; 800 units superscript II RT (Life Technologies, Gaithersburg, Md.); 1 ⁇ first strand cDNA synthesis buffer (50 mM Tris-HCl, pH 8.3; 75 mM KCl; 5 mM MgCl 2 ), which was incubated for I hr at 37° C.
  • the reaction mixture was then adjusted to 250 ⁇ l through the addition of 1 ⁇ second strand buffer (30 mM Tris-HCl, pH 7.5; 105 mM KCl; 5.2 mM MgCl 2 ); 0.1 mM DTT; methylation dNTP mix (10 mM each); 50 units E. coli DNA polymerase I, 3 units RNase H; 15 units E. coli DNA ligase (all enzymes from Life Technologies), which was incubated for an additional 2.5 hr at 15° C. The resulting double-stranded cDNAs (dscDNA) were then treated with 1.5 units T4 DNA polymerase (Novagen Inc., Madison, Wis.) for 20 min at 11° C.
  • T4 DNA polymerase Novagen Inc., Madison, Wis.
  • This example illustrates a method of functionally screening a rabbit cDNA library so as to identify cDNAs encoding LBPs which bind to both native LDL and methyl LDL.
  • Methyl LDL is not recognized by previously reported cell surface receptors. See, e.g., Weisgraber et al., J. Biol. Chem. 253:9053-9062 (1978).
  • a fresh overnight culture of E. coli ER1 647 cells (Novagen Inc.) was infected with the cDNA phage obtained from Example 1, and plated at a density of 2 ⁇ 10 4 plaque-forming units (pfu) in 150 mm diameter plates containing 2 ⁇ YT agar. A total of 50 plates, equivalent to 1 ⁇ 10 6 phage, were plated and incubated at 37° C. until the plaques reached 1 mm in diameter (5-6 hr). A dry nitrocellulose membrane, which had previously been saturated with 10 mM IPTG solution, was layered on top of each plate to induce the production of recombinant protein, as well as to immobilize the proteins on the membranes. The plates were incubated at 37° C. for an additional 3-4 hr, and then overnight at 4° C.
  • the membranes were then blocked for 30 min at room temperature in a solution of TBSEN with 5% non-fat dry milk, followed by 10 min in TBSEN with 1% non-fat dry milk. Following blocking, the membranes were incubated with native human LDL (obtained as described in Example 11 or methylated human LDL (meLDL) (see Weisgraber et al., J. Biol. Chem. 253:9053-9062 (1978)), at a concentration of 4 ⁇ g/ml, in a solution containing 1 ⁇ TBSEN, 1% non-fat dry milk, 1 mM PMSF, 0.5 ⁇ protease inhibitor solution (1 mM ⁇ -amino caproic acid/1 mM benzamidine). Incubation was for 4 hr at room temperature in a glass Petri dish with gentle stirring on a stirring table, followed by overnight at 4° C. with no stirring.
  • native human LDL obtained as described in Example 11 or methylated human LDL (meLDL) (see Weisgraber
  • E. coli plys E cells were grown to log phase, spun down and resuspended in PBS containing 1 mM PMSF, 2 mM ⁇ -amino caproic acid, and 1 mM benzamidine. The cell suspension then underwent 8 freeze-thaw cycles via immersion in liquid nitrogen and cold running tap water, respectively.
  • the anti LDL antibodies/cell extract solution were incubated with gentle stirring for 1 hr at 4° C. (1 ml of antibody solution/3 mg crude cell extract). Following incubation, the mixture was centrifuged (10,000 ⁇ g; 10 min; 4° C.) and the supernatant was stored at 4° C. in the presence of 0.02% NaN 3 , until use.
  • the membranes were processed for immunoscreening as follows: (i) three 5-min washes at room temperature in TBSEN containing 1% gelatin; (ii) 30 min incubation in PBS, pH 7.4 with 1% gelatin; (iii) two-hr room temperature incubation with gentle stirring in fresh PBS/gelatin solution containing adsorbed sheep anti-human LDL antibodies (Boehringer Manheim, Indianapolis, Ind.) (1:1000 dilution); (iv) three brief washes in TBS, pH 7.4; (v) one-hr room temperature incubation with gentle stirring in PBS/gelatin solution containing donkey anti-sheep alkaline phosphatase-conjugated antibodies (Sigma, St.
  • the phage from Example 1 containing the LBP cDNAs were plaque-purified and converted into plasmid subclones by following a protocol called “Autosubcloning by Cremediated Plasmid Excision” provided by Novagen Inc.
  • DNA sequences were obtained by the dideoxynucleotide chain-termination method (Sanger et al., Proc. Natl. Acad. Sci., USA 74: 5463-5467 (1977), and analyzed by an Applied Biosystems automated sequencer.
  • the open reading frame (ORF) of each cDNA was determined from consensus sequences obtained from both the sense and antisense strands of the cDNAs. Sequencing confirmed that three previously unknown genes had been isolated.
  • LBPs LDL binding proteins
  • the sizes of the recombinant proteins were determined to be 16.2 kDa for LBP-1, 40 kDa for LBP-2, and 62.7 kDa for LBP-3.
  • RNA was isolated from different rabbit tissues: adrenals, thoracic aorta, abdominal aorta, ballooned and reendothelialized abdominal aorta, heart, kidney, smooth muscle cells, lung and liver, by Trizol reagent (Life Technologies) and concentrated by ethanol precipitation.
  • Gel electrophoresis of RNA was carried out in 1.2% agarose gel containing 1 ⁇ MOPS buffer (0.2 M MOPS, pH 7.0; 50 mM sodium acetate; 5 mM EDTA, pH 8.0) and 0.37 M formaldehyde.
  • RNAs were blotted onto supported nitrocellulose membranes (Schleicher & Schuell, Keene, N.H.) and immobilized by baking at 80° C. for 2 hr.
  • Hybridization to radiolabeled LBP-1, LBP-2 and LBP-3 cDNA or cRNA probes was carried out by standard procedures known to those skilled in the art (see, e.g., Ausubel et al., Current Protocols in Molecular Biology; John Wiley & Sons (1989)); signals were detected by autoradiography.
  • the results were as follows: the sizes of the mRNAs were about 1.3 kb for LBP-1, about 2.3-2.5 kb for LBP-2, and about 4.7 kb for LBP-3. LBP-1, LBP-2 and LBP-3 mRNA were found in all tissues tested, but the highest amount was in ballooned abdominal aorta.
  • This example illustrates isolation of human LBP cDNAs.
  • Human LBP cDNA clones were isolated from three cDNA libraries.
  • a human fetal brain cDNA library was obtained from Stratagene, LaJolla, Calif.
  • a human liver and a human aorta cDNA library were obtained from Clontech, Palo Alto, Calif., and screened with a radiolabeled cDNA probe derived from rabbit LBP-1, LBP-2 or LBP-3, according to the method described in Law et al., Gene Expression 4:77-84 (1994).
  • Several strongly hybridizing clones were identified and plaque-purified.
  • Clones were confirmed to be human LBP-1, LBP-2 and LBP-3, by DNA sequencing using the dideoxynucleotide chain-termination method and analysis by an Applied Biosystems automated sequencer.
  • the cDNA sequences and the corresponding proteins for human LBP-1, LBP-2 and LBP-3 are set forth in SEQ ID NOS: 15, 16 and 17, respectively.
  • a human genomic library was screened with each of the LBP-1, LBP-2, and LBP-3 clones obtained from the cDNA library screening. Clones hybridizing to each of the three cDNAs were isolated and sequenced.
  • the genomic sequence for LBP-1, LBP-2, and LBP-3 are set forth in FIGS. 22 - 24 , respectively.
  • the LBP-1 open reading frame spans four exons of the LBP-1 gene (FIG. 22; SEQ ID NO: 49).
  • the LBP-1 protein predicted by the genomic sequence is identical to that predicted by the cDNA clone described above.
  • the LBP-2 open reading frame spans five exons of the LBP-2 gene (FIG. 23; SEQ ID NO: 50).
  • the LBP-2 protein predicted by the genomic sequence differs from that predicted by the cDNA clone in that it contains an additional 321 amino acids at its amino terminus (the LBP-2 cDNA is a 5′ truncation).
  • the LBP-3 open reading frame spans ten exons of the LBP-3 gene (FIG. 24; SEQ ID NO: 51).
  • the LBP-3 protein predicted by the genomic sequence differs from that predicted by the cDNA clone in that it contains an additional 16 amino acids at its amino terminus (the LBP-3 cDNA is a 5′ truncation) and an Asn at amino acid position 130 (the cDNA predicts a Tyr at this position).
  • a comparison between the corresponding LBP-1, LBP-2 and LBP-3 protein sequences for rabbit and human are shown in FIGS. 19, 20 and 21 .
  • LBP cDNA was isolated from the original pEXlox plasmids obtained as described in Examples 1 and 2, and subcloned into the pPRoEX-HT vector (Life Technologies) for recombinant protein expression. Induction of the recombinant protein by IPTG addition to transformed E. coli DH10B cultures resulted in the expression of recombinant protein containing a 6-histidine tag (N-terminal). This tagged protein was then purified from whole cell proteins by binding to Ni-NTA (nickel nitrilo-triacetic acid) as described in the protocol provided by the manufacturer (Qiagen, Inc., Santa Clara, Calif.). The preparation obtained after the chromatography step was approximately 90% pure; preparative SDS-PAGE was performed as the final purification step.
  • Ni-NTA nickel nitrilo-triacetic acid
  • iodination of LBPs was carried out using Iodobeads (Pierce, Rockford, Ill.).
  • the Iodobeads were incubated with 500 ⁇ Ci of Na 125 I solution (17 Ci/mg) (New England Nuclear, Boston, Mass.) in a capped microfuge tube for 5 min at room temperature.
  • the protein solution was added to the Iodobeads-Na 125 I microfuge tube and incubated for 15 min at room temperature. At the end of this incubation, aliquots were removed for the determination of total soluble and TCA precipitable counts.
  • radiolabeled protein was then precipitated with cold acetone (2.5 vol; ⁇ 20° C.; 2.5 hr). Following this incubation, precipitated protein was collected by centrifugation (14,000 g; 1 hr; room temperature) and resuspended in sample buffer (6 M urea/50 mM Tris, pH 8.0/2 mM EDTA). Integrity of the protein preparation was assessed by SDS-PAGE.
  • This example illustrates the production of polyclonal antibodies to LBP-1, LBP-2 and LBP-3.
  • a mixture of purified recombinant LBP protein (0.5 ml; 200 ⁇ g) and RIBI adjuvant (RIB1 ImmunoChem. Research, Inc., Hamilton, Mont.) was injected subcutaneously into male guinea pigs (Dunkin Hartley; Hazelton Research Products, Inc., Denver, Pa.) at 3-5 sites along the dorsal thoracic and abdominal regions of the guinea pig. Blood was collected by venipuncture on days 1 (pre-immune bleeding), 28, 49 and 70.
  • Booster injections were administered on days 21 (100 ⁇ g; SC), 42 (50 ⁇ g; SC), and 63 (25 ⁇ g; SC).
  • the titer of the guinea pig antiserum was evaluated by serial dilution “dot blotting.” Preimmune antiserum was evaluated at the same time.
  • the titer against the recombinant protein reached a maximal level with a detectable calorimetric response on a dot blot assay of 156 pg.
  • This example illustrates the presence of LBPs in or on endothelial cells covering plaques, in or on adjacent smooth muscle cells, and in the extracellular matrix.
  • co-localization of LDL and LBPs was demonstrated.
  • Ballooned deendothelialized aorta was obtained from rabbits which had received a bolus injection of human LDL (3 mg; i.v.) 24 hr prior to tissue collection.
  • Human aortas containing atherosclerotic plaques were obtained from routine autopsy specimens. Tissues were fixed in 10% buffered formalin ( ⁇ 24 hr) and imbedded in paraffin using an automated tissue-imbedding machine. Tissue sections were cut (5-7 ⁇ ) and mounted onto glass slides by incubating for 1 hr at 60° C. Sections were deparaffinized.
  • Controls included preimmune serum as well as specific antisera to LBP-1, LBP-2, or LBP-3 in which the primary antibody was completely adsorbed and removed by incubation with recombinant LBP-1, LBP-2 or LBP-3 followed by centrifugation prior to incubation with the tissue sections.
  • An affinity purified rabbit polyclonal antibody against human apolipoprotein B (Polysciences Inc.; Warrington, Pa.) was used at a dilution of 1:100 (in 5% normal rabbit serum/PBS). Sections were incubated for 2 hr at room temperature in a humidified chamber.
  • Sections were then rinsed with PBS and antigen-antibody signal amplified using avidin/biotin HRP conjugate (Vectastain ABC kit; Vector Laboratories, Burlingame, Calif.). Sections were developed using DAB substrate (4-6 min; room temperature) and counterstained with hematoxylin.
  • LBP-1, LBP-2 and LBP-3 were located in or on functionally modified endothelial cells at the edges of regenerating endothelial islands, the same location in which irreversible LDL binding has been demonstrated (Chang et al., Arteriosclerosis and Thrombosis 12:1088-1098 (1992)).
  • LBP-1, LBP-2 and LBP-3 were also found in or on intimal smooth muscle cells underneath the functionally modified endothelial cells, and to a lesser extent, in extracellular matrix.
  • ACE Affinity Coelectrophoresis
  • This example illustrates that binding occurs between LBP-1, LBP-2 or LBP-3 and LDL, and that this binding is specific, as illustrated by the fact that binding does not occur between LBP-1, LBP-2 or LBP-3 and HDL (high density lipoprotein).
  • Analysis of the affinity and specificity of recombinant rabbit LBP-1, LBP-2 or LBP-3 binding to LDL was carried out using the principle of affinity electrophoresis (Lee and Lander, Proc. Natl. Acad. Sci. USA 88:2768-2772 (1991)).
  • Melted agarose (1%; 65° C.) was prepared in 50 mM sodium MOPS, pH 7.0; 125 mM sodium acetate, 0.5% CHAPS.
  • a teflon comb consisting of nine parallel bars (45 ⁇ 4 ⁇ 4 mm/3 mm spacing between bars) was placed onto GelBond film (FMC Bioproducts, Rockland, Me.) fitted to a plexiglass casting tray with the long axis of the bars parallel to the long axis of the casting tray.
  • a teflon strip (66 ⁇ 1 ⁇ 1 mm) was placed on edge with the long axis parallel to the short axis of the casting tray, at a distance of 4 mm from the edge of the teflon comb.
  • Melted agarose (>65° C.) was then poured to achieve a height of approximately 4 mm.
  • LDL or HDL samples were prepared in gel buffer (50 mM sodium MOPS, pH 7.0, 125 mM sodium acetate) at twice the desired concentration. Samples were then mixed with an equal volume of melted agarose (in 50 mM MOPS, pH 7.0; 125 mM sodium acetate; 50° C.), pipetted into the appropriate rectangular wells and allowed to gel. The binding affinity and specificity of LBP-1 and LBP-3 was tested using several concentrations of LDL (540 to 14 nM) and HDL (2840177 nM).
  • LDL retarded LBP-1, LBP-2 and LBP-3 migration through the gel in a concentration-dependent, saturable manner, indicating that LBP-1, LBP-2 and LBP-3 binding to LDL was highly specific. This conclusion is supported by the fact that HDL did not retard LBP-1, LBP-2 or LBP-3.
  • Cold LBP-1 inhibited binding of radiolabeled LBP-1 to LDL in a concentration-dependent manner
  • cold LBP-2 inhibited binding of radiolabeled LBP-2 to LDL in a concentration-dependent manner
  • cold LBP-3 inhibited binding of radiolabeled LBP-3 to LDL in a concentration-dependent manner.
  • Rabbit and human LBP-2 contain a long stretch of acidic amino acids at the amino terminal (rabbit LBP-2 amino acid residues 338 through 365 and human LBP-2 amino acid residues 329 through 354).
  • the possibility that this segment of LBP-2 was the LDL binding domain was tested by subcloning two rabbit LBP-2 clones which differ from each other by the presence or absence of this acidic region (clone 26 and clone 45, respectively) into expression vectors, by standard methods known to those skilled in the art.
  • ACE assays were then conducted in order to assess the affinity and specificity of the binding of these two clones to LDL.
  • LDL retarded clone 26 derived radiolabeled LBP-2 migration through the gel in a concentration-dependent, saturable, manner while clone 45 derived radiolabeled LBP-2 migration was not retarded.
  • ACE Affinity Coelectrophoreses
  • This example illustrates that binding between LBP-1 or LBP-2 and LDL is inhibited by polyglutamic acid or BHF-1.
  • the ability of a third compound to inhibit binding between two proteins previously shown to interact was tested by a modification of the ACE assays described in Example 8. The third compound was added to the top or wells together with the radiolabeled protein. If the third compound inhibited binding, the radiolabeled protein would run through the gel. If the third compound did not inhibit binding, migration of the radiolabeled protein was retarded by the protein cast into the gel.
  • ACE Affinity Coelectrophoreses
  • This example illustrates a method for identifying fragments, analogs or mimetics of LBPs which bind to LDL, and which thus can be used as inhibitors of LDL binding to LBP in the arterial walls, by occupying binding sites on LDL molecules, thereby rendering these sites unavailable for binding to LBP in the arterial wall.
  • Fragments of LBPs are generated by chemical cleavage or synthesized from the known amino acid sequences. Samples of these fragments are individually added (cold) to radiolabeled LBP as described in Example 8, to assess the inhibitory potency of the various fragments. By iterative application of this procedure on progressively smaller portions of fragments identified as inhibitory, the smallest active polypeptide fragment or fragments are identified. In a similar manner, analogs of the LBPs are tested to identify analogs which can act as inhibitors by binding to LDL.
  • LBP molecules which resemble the conformation and/or charge distributions of the LDL-binding sites on LBP molecules
  • mimetics of LBP are tested in a similar fashion to identify molecules exhibiting affinities for the LDL-binding sites on LBP.
  • the affinities of the inhibitors so identified are at least as strong as the affinity of LDL itself for the LDL-binding sites on LBP.
  • the inhibitors bind at least competitively, and some irreversibly and preferentially as well, to the LDL-binding sites, thereby rendering such sites unavailable for binding to humoral LDL.
  • This example illustrates the use of ELISA plate assays for the quantification of a test compound's capacity to inhibit the binding of LDL to a specific LBP.
  • the ELISA assay was carried out as follows: LDL was diluted in 50 mM Na 2 HCO 3 , pH 9.6/0.02% NaN 3 and added to the wells of a 96-well plate (Immuno Ware 96-Well Reacti-Bind EIA Polystyrene Plates; Pierce (Rockford, Ill.)) to achieve a final concentration ranging from 0.1 to 1 ⁇ g/well. The plates were incubated for 6 hr at room temperature.
  • the wells were washed 3 times with Tris-buffered saline, pH 7.4 (TBS), and blocked overnight with 200 ⁇ l of 1% bovine serum albumin (BSA) in TBS/0.02% NaN 3 (Sigma; St. Louis Mo.) at room temperature.
  • BSA bovine serum albumin
  • the wells were then incubated with 200 ⁇ l of LBP protein (5-10 ⁇ g/well) in TBS and varying concentrations of the test compound. Plates were incubated for 1 hr at room temperature.
  • the wells were then washed three times with TBS and blocked for 2 hr with 200 ⁇ l of 1% BSA in TBS/0.02% NaN 3 at room temperature.
  • the wells were washed 3 times with TBS and a 1:1000 dilution (in TBS/0.05% Tween 20) of the appropriate guinea pig anti-LBP protein polyclonal antibody was added to the wells and incubated for 1 hr at room temperature. The wells were then washed 3 times with TBS/0.05% Tween 20; a 1:30,000 dilution of goat anti-guinea pig IgG alkaline phophatase conjugate (Sigma) was added to each well. Plates were incubated for 1 hr at room temperature.
  • the wells were washed 3 times with TBS/0.05% Tween 20 and a calorimetric reaction was carried out by adding 200 ml of p-nitrophenyl phosphate substrate (Sigma; St. Louis Mo.) to the wells. The reaction was allowed to proceed for 30 min at room temperature and stopped with 50 ⁇ l of 3 N NaOH. The absorbance was determined at 405 nm using an ELISA plate reader. The test compound's effectiveness in blocking the binding of LDL to the recombinant protein was assessed by comparing the absorbance values of control and treated groups.
  • the ELISA assay was carried out as follows: LDL was diluted in Tris-buffered saline, pH 7.4 (TBS) and added to the wells of a 96-well plate (Immuno Ware 96-Well Reacti-Bind EIA Polystyrene Plates; Pierce (Rockford, Ill.)) to give a plate-saturating concentration of 0.2 ⁇ g/well. The plate was incubated for 1 hr at room temperature, after which the wells were washed three times with TBS, before being blocked for 1 hr at room temperature with 1% bovine serum albumin (BSA in TBS).
  • BSA bovine serum albumin
  • the wells were then washed twice with TBS before LBP-1 or LBP-2 (0.025 ⁇ g/well), or LBP-3 (0.01 ⁇ g/well) were added, without and with varying concentrations of the test inhibitor compound. Each condition was set up in quadruplicate. The plate was incubated for 1 hr at room temperature, then washed three times with TBS/0.02% Tween (TBS/Tween). An appropriate dilution of guinea pig anti-LBP polyclonal antibody (1:750 to 1:1500, depending on the antibody) was added to three wells for each condition and incubated for 1 hr. Anti-LBP antibody was replaced by buffer for the fourth well of each condition, as a negative control.
  • the plate was again washed three times with TBS/Tween before a 1:10,000 dilution (in TBS/Tween) of goat anti-guinea pig IgG alkaline phosphatase-conjugated antibody (Sigma) was added to each well.
  • the plate was incubated for 1 hr at room temperature, then washed three times with TBS/Tween.
  • a fresh solution of substrate was prepared from an Alkaline Phosphatase Substrate Kit (Bio-Rad, Hercules, Calif.) as follows: Mix 1 ml 5 ⁇ concentrated diethanolamine buffer with 4 ml distilled water.
  • LBPs rather than LDL, were bound to the plate.
  • Recombinant LBP protein binding to LDL and the effect of varying concentration of the inhibitor on LBP-LDL binding was determined through the use of antibodies against LDL. This interaction was visualized through the use of a secondary antibody conjugated to a reporter enzyme (e.g. alkaline phosphatase).
  • a reporter enzyme e.g. alkaline phosphatase
  • ELISA plate assays were used to screen agents which can affect the binding of LBP proteins to LDL.
  • peptides derived from LBP-1 and human LBP-3 protein sequences BHF-1 and BHF-2, respectively
  • BHF-1 and BHF-2 were synthesized and have been shown to reduce the binding of LDL to recombinant LBP-1 and LBP-2 in this format.
  • This example illustrates administration to patients of humanized antibodies against LBP-1, LBP-2 or LBP-3 so as to block LDL-binding sites on arterial LBP molecules.
  • Mouse monoclonal antibodies are humanized by recombinant DNA techniques and produced by standard procedures known to those skilled in the art (Berkower, I., Curr. Opin. Biotechnol. 7:622-628 (1996); Ramharayan and Skaletsky, Am. Biotechnol. Lab 13: 26-28 (1995)) against LBPs and/or the LDL-binding sites on the LBPs.
  • the corresponding Fab fragments are also produced, as described in Goding, J.
  • LDL Low density lipoproteins
  • the density of the infranate was raised to 1.050 gm/ml with KBr solution and centrifuged for 22-24 hr, 100,000 ⁇ g, 12° C.
  • LDL was removed from the tops of the centrifuge tubes with a drawn out Pasteur pipette tip. Purity of the LDL preparation was checked by Ouchterlony double immunodiffusion against antibodies to human LDL, human HDL, human immunoglobulins, and human albumin.
  • KBr was removed from the LDL solution by dialysis (1L, ⁇ 2, approximately 16 hr) against 0.9% saline, pH 9.0, containing 1 mM EDTA and 10 ⁇ M butylated hydroxytoluene (BHT), the latter to prevent oxidation of LDL.
  • BHT butylated hydroxytoluene
  • LDL protein was measured by the method of Lowry (Lowry et al., J. Biol. Chem. 193:265-275 (1951)), and the LDL was stored at 4° C. until use. LDL preparations were kept for no more than 4-6 weeks.
  • HDL was prepared from plasma of normolipemic donors. 100 ml of whole blood was placed into tubes containing 100 mM disodium EDTA and plasma was collected by centrifugation (2000 g; 30 min; 4° C.). Apolipoprotein B containing lipoproteins present in plasma were then precipitated by the sequential addition of sodium heparin (5,000 units/ml) and MnCl 2 (1 M) to achieve a final concentration of 200 units/ml and 0.46 M, respectively (Warnick and Albers, J. Lipid Res. 19:65-76 (1978)). Samples were then centrifuged, (2000 g; 1 hr; 4° C.).
  • HDL was separated by ultracentrifugation (100,000 g; >46 hr; 12° C.). Purity of the HDL preparation was assessed via Ouchterlony double immunodiffusion test using antibodies against human HDL, human LDL, human immunoglobulins, and human albumin. HDL samples were dialyzed against saline pH 9.0/1 mM EDTA/10 ⁇ M BHT (4L; 24 hr/4° C.) and total protein was determined by the Lowry protein assay (Lowry et al., J. Biol. Chem. 193:265-275 (1951)). HDL was stored at 4° C. until use. HDL preparations were kept for no longer than 2 weeks.
  • BHF-1 a fragment of human or rabbit LBP-1 which contains amino acid residues 14 through 33.
  • BHF-1 was synthesized using an Applied Biosystems Model 430A peptide synthesizer with standard T-Boc NMP chemistry cycles.
  • the sequence of BHF-1 is as follows:
  • the peptide was cleaved with hydrofluoric acid/anisole (10/1 v/v) for 30 min at ⁇ 10° C. and then incubated for 30 min at 0° C. BHF-1 was then precipitated and washed three times with cold diethyl ether. Amino acid coupling was monitored with the ninhydrin test (>99%).
  • the BHF-1 peptide was purified to homogeneity by high performance liquid chromatography on a reverse phase Vydac C 4 column (2.24 ⁇ 25 cm) using a linear gradient separation (2-98%B in 60 min) with a flow rate of 9 ml/min.
  • Buffer A consisted of 0.1% trifluoroacetic acid (TFA)/Milli Q water and Buffer B consisted of 0.085% TFA/80% acetonitrile.
  • the gradient was run at room temperature and absorbance monitored at 210 and 277 nm.
  • a candidate polypeptide for being an agent is chosen, e.g., LBP-1, LBP-2. LBP-3, BHF-1 or any other polypeptide.
  • the shortest fragment of the polypeptide that inhibits LDL binding to LBPs in vitro is determined.
  • Peptides are synthesized by standard techniques described herein. Inhibition assays are performed using standard ELISA techniques for screening, and affinity coelectrophoresis (ACE) assays to confirm the ELISA results, as described herein. Additional assays that can be used in this screening method include, e.g., fluorescence polarization and pulsed ultra-filtration electrospray mass spectrometry.
  • Short peptides ranging, e.g., from dimers to 20-mers are constructed across sequences of the candidate polypeptide whose chemical characteristics make them likely LDL binding sites, e.g., acidic regions.
  • the ability of shorter and shorter lengths of the peptides to inhibit LDL binding in vitro and to mammalian cells in culture is tested.
  • the effect of the peptide on inhibiting LDL binding in mammalian cells transfected to express an LBP gene is tested.
  • Each of the peptides so identified as an inhibitor is tested with each of LBP-1, LBP-2 and LBP-3, to determine whether a single inhibitor works against all three LBPs.
  • the peptide backbone is modified so as to inhibit proteolysis, as discussed herein. For example, modification is accomplished by substitution of a sulfoxide for the carbonyl, by reversing the peptide bond, by substituting a methylene for the carbonyl group, or other similar standard methodology. See Spatola, A.F., “Peptide Backbone Modifications: A Structure-Activity Analysis of Peptides Containing Amide Bond Surrogates, Conformational Constraints, and Related Backbone Replacements,” in Chemistry and Biochemistry of Amino Acids, Peptides and Proteins, Vol. 7, pp. 267-357, B.
  • This example illustrates cell-based in vitro screening of agents which have been shown by in vitro tests such as ACE assay and ELISA to be potential inhibitors of binding between LDL and LBPs.
  • Mammalian cells such as 293 cells, which are commonly used for expression of recombinant gene constructs, are used to develop cell lines which express LBPs on the cell surface. This is done by subcloning LBP open reading frames (ORFS) into a mammalian expression plasmid vector, pDisplay (Invitrogen, Carlsbad, Calif.), which is designed to express the gene of interest on the cell surface.
  • ORFS LBP open reading frames
  • pDisplay Invitrogen, Carlsbad, Calif.
  • the use of mammalian cells to produce LBPs allows for their expression in a functionally active, native conformation. Therefore, stably transfected mammalian cell lines with surface expression of LBPs individually, or in combination, are particularly suitable for assaying and screening inhibitors that block LDL binding in cell culture, as well as to evaluate the cytotoxicity of these compounds.
  • LBP ORFs are amplified by PCR (Perkin Elmer, Foster City, Calif.) from cDNA templates using Taq polymerase (Perkin Elmer) and appropriate primers.
  • the amplified LBP ORFs are purified by agarose gel electrophoresis and extracted from gel slices with the Bio-Rad DNA Purification kit (Bio-Rad, Hercules, Calif.).
  • the purified DNAs are then cut with the restriction enzymes Bgl II and Sal I (New England Biolabs, Beverly, Mass.) to generate cohesive ends, and purified again by agarose gel electrophoresis and DNA extraction as described above.
  • LBP ORFs are then subcloned into the Bgl II/Sal I sites in the mammalian expression vector, pDisplay (Invitrogen) by ligation.
  • Recombinant plasmids are established by transformation in E. coli strains TOP10 (Invitrogen) or DH5 ⁇ (Life Technologies, Grand Island, N.Y.).
  • Recombinant pDisplay/LBP plasmid DNA is isolated from overnight E. coli cultures with the Bio-Rad Plasmid Miniprep kit, cut with Bgl II/Sal I, and analyzed by agarose gel electrophoresis.
  • LBP ORFs in successfully transformed clones are verified by automated dideoxy DNA sequencing.
  • 1-2 ⁇ g of DNA is mixed with 6 ⁇ l lipofectamine reagent (Life Technologies) and incubated with the cells as described in the Life Technologies protocol.
  • LBP expression in transfected cells is confirmed by Western blot analysis of cell extracts obtained 48 hr after transfection.
  • the antibiotic G418 (Life Technologies) is added to the growth medium at a concentration of 800 ⁇ g/ml. Colonies resistant to G418 are tested for recombinant LBP expression by Western blot, and recombinant clones expressing LBPs are expanded, assayed for LDL binding and used to test compounds for their ability to inhibit LDL binding.
  • Each candidate inhibitor is tested in five to ten ballooned rabbits, while an equal number of rabbits receive a control peptide, or placebo.
  • This example illustrates the effect that induction of immunity against LBP protein has on the accumulation of radiolabeled LDL in the ballooned deendothelialized rabbit aorta model of atherosclerosis.
  • Immunity was induced in male New Zealand White rabbits (Hazelton Research Products, Denver, Pa.) as follows: A mixture of purified human recombinant LBP-2 or BHF-1 peptide (1 ml; 1 mg) and RIBI adjuvant (RIBI ImmunoChem Research, Inc., Hamilton, Mont.) was injected subcutaneously at 2-5 sites along the dorsal thoracic and abdominal regions of the rabbits. Blood was collected by venipuncture on days 1 (preimmune bleeding), 35, 63, and 91. Booster injections were administered on days 28 (500 ⁇ g; SC), 56 (250 ⁇ g; SC), and 84 (125 ⁇ g; SC).
  • the titer of the rabbits was evaluated by serial dilution using an ELISA plate format. Preimmune serum was evaluated at the same time. After the third booster of LBP protein or peptide, the titer reached a maximal level with a detectable calorimetric response on an ELISA plate of 156 pg. Titer is defined as the maximum dilution of antibody which generates an absorbance reading of 0.5 above control in 30 min. Specificity of the polyclonal antibodies was demonstrated using Western blot analysis as described in Example 6.
  • Total TCA precipitable counts were determined by addition of TCA to the serum to a final concentration of 10% followed by incubation for min at 4° C. Serum samples were then centrifuged (2000 rpm; 30 min; 40° C.) and total activity present in the supernate was determined. TCA precipitable counts were calculated by substration: total soluble counts minus counts present in the supernate after TCA precipitation. Blood samples for the determination of antibody titers were collected prior to the injection of the radiolabeled LDL.
  • the rabbits were injected intravenously with 5% Evan's blue dye which was allowed to circulate for 15 min. Areas of the aorta in which the endothelial covering is absent stain blue while those areas covered by endothelium remain unstained. At the end of the incubation period, the rabbits were euthanized and the abdominal and thoracic aorta were dissected out, rinsed, and fixed overnight in 10% TCA at room temperature.
  • the aortas were then rinsed exhaustively with physiological saline, weighed, counted, blotted dry and placed onto X-ray film in order to visualize the pattern of radiolabeled LDL accumulation in the deendothelialized rabbit abdominal aorta.
  • Initial testing includes 20-50 subjects on active drug and 20-50 subjects, matched for age, sex, and atherosclerotic status, on placebo. The number of subjects is pre-determined by the number needed for statistical significance. Endpoints for inhibitor efficacy includes ultrasound measurements of carotid artery thickness in high risk subjects, as well as in subjects with known carotid or coronary disease; atherosclerotic events; atherosclerotic deaths; and all-cause deaths in all subjects.
  • Non-invasive analysis carotid artery thickness by ultrasound
  • Stadler Med. and Biol. 22:25-34 (1996)
  • Atherosclerotic events and deaths, as well as all-cause deaths are tabulated at 3 years.
  • Oral dosage of drug in FDA Phase I trials ranges from 0.01 to 10 gm/day, and is determined by results of animal studies, extrapolated on a per kg basis. Based on data obtained from Phase I studies, the dose range and frequency are narrowed in Phase II and III trials. If parenteral administration of drug is determined by animal studies to be the only effective method, parenteral administration in human subjects is tested by injection, as well as by the transdermal and nasal insufflation routes. Testing of parenteral drug follows the same outline as that for oral administration.
  • This example illustrates a method for treating an individual having atherosclerosis with an LBP fragment, e.g., BHF-1, so as to decrease the levels of arterially bound LDL in the individual.
  • BHF-1 is obtained as described herein.
  • the BHF-1 is administered to the mammal intravenously as a bolus or as an injection at a concentration of 0.5-10 mg/kg body weight. Such administrations are repeated indefinitely in order to prevent the development or progression of symptomatic atherosclerosis, just as is done currently with cholesterol lowering drugs.
  • Stable subjects are examined twice yearly to evaluate the extent of any atherosclerotic disease by physical exam and non-invasive studies, such as carotid artery thickness, ultrasound, and/or gamma camera imaging of the major arteries, to determine if atherosclerotic lesions are present, and, if previously present, have regressed or progressed.
  • Such a regimen results in treatment of the atherosclerosis.
  • mice Four week old apo E knockout mice (Jackson Laboratories, Bar Harbor, Maine) were ear tagged, randomly assigned to different cages and weighed. Body weights were determined weekly. Animals were allowed to habituate for 1 week. Normal rodent chow was provided ad libitum and animals were maintained in a 12:12 light:dark cycle. The following four groups of mice were treated with either recombinant LBP proteins (40 ⁇ g of recombinant protein/mouse) plus RIBI adjuvant or RIBI adjuvant alone (control group).
  • LBP-1 Immunized with rabbit recombinant LBP-1 (6-His tag).
  • LBP-2 Immunized with rabbit recombinant LBP-2 clone 26 (6-His tag).
  • LBP-3 Immunized with rabbit recombinant LBP-3 (6-His tag).
  • Control Received adjuvant.
  • mice Blood samples (pre-immune serum) were collected prior to the initial injection of recombinant protein and RIBI adjuvant (as described in the manufacturer's manual). After 21 days, mice received a booster injection (half-initial dose) and were then bled seven days later. Titer was defined as the maximum dilution of serum that yielded a change in absorbance equivalent to 2 ⁇ that of control serum (60 min; 37° C.). The amount of recombinant protein per well was 100 ng.
  • Booster injections took place at 21 day intervals until an average titer value of 1:10,000 was reached. At this time, mice were switched to western type diet (Harland Teklad, Madison, Wis.) and fed ad libitum. Blood samples were collected at this time (retro-orbital sinus bleeding technique) and monthly thereafter.
  • mice were sacrificed at 26 weeks of age. The mice were anesthetized with methoxyfluorane and exanguinated via cardiac puncture. A midline thoracotomy was performed, a cannula inserted into the right ventricle and perfusate allowed unrestricted flow via an incision into the right atrium. The mice were perfused with saline, followed by 10% phosphate buffered formalin until fasciculations stopped. At this time, the aorta was exposed and adventitial fat removed in situ. The aorta was then removed from the heart down to the iliac bifurcation and placed in 10% phosphate buffered formalin overnight.
  • the aorta was stained as follows: after a brief 70% ethanol rinse, it was immersed in a filtered solution of 0.5% (weight/volume) Sudan IV in 35% ethanol/50% acetone with continuous shaking for 10 minutes at room temperature. Unbound dye was removed by incubating the aorta in an 80% ethanol solution with shaking until the background color was clear. The vessel was then rinsed in distilled water, placed in physiological saline and opened longitudinally from the aortic arch down to the iliac bifurcation. The vessel was pinned out and photographed.
  • Photographs were then digitized using an Astra 1200S scanner (UMAX Technologies Inc., Freemont, Calif.) and a commercially available graphics program (Canvas; Deneba Software, Miami Fla.). Total and lesion areas were determined using the signal processing toolbox of MATLAB (The Mathworks Inc., Natick, Mass.). Percent involvement was calculated by dividing lesion area by total area.
  • a second analysis was done to measure aortic atherosclerosis by a cholesterol extraction method whereby cholesterol is determined as a unit weight of artery. This method may be more accurate in measuring lesion size than attempting to measure the thickness of many sections. Specifically, the weight of an artery was measured, then the cholesterol was extracted. Aortic cholesterol content was then measured by gas-liquid chromatography. The amount of cholesterol per unit weight of artery was then determined.
  • Apo E knockout mice immunized against LBP-1 had moderate to high titers (>1:1000 to ⁇ 1:8000).
  • Apo E knockout mice immunized against LBP-2/26 had moderate titer levels (>1:2000).
  • LBP-3 titers range from moderate to high (>1:1000 to >1:8000) in the Apo knockout mice.
  • mice immunized against LBP-1 had relatively high titers (>1:10,000) while others had moderate to high titers (>1000 to ⁇ 1:10,000).
  • Some of the Apo E knockout mice had moderate ( ⁇ 1:5000) to low ( ⁇ 1:1000) titers.
  • LBP-3 titers ranged from high (>1:5000 to ⁇ 1:10,000) to moderate (>1:1000 to ⁇ 1:5000).
  • LDL receptor (LDLR) knockout mice (B6,129S-Ldlr tmlHer , Jackson Laboratories, Bar Harbor, Me.) were injected with the BHF-1 peptide (see Example 15 for methods of synthesizing the BHF-1 peptide) together with an RIBI adjuvant (RIBI ImmunoChem Research, Inc., Hamilton, Mont.).
  • LDLR knockout mice are hyperlipidemic and thus a model for human atherosclerosis. LDLR knockout mice have high levels of plasma cholesterol and develop naturally-occurring atherosclerotic-like lesions.
  • mice Four week old LDLR knockout mice were ear tagged, randomly assigned to different cages and weighed. Body weights were subsequently determined weekly. Animals were allowed to habituate for one week prior to experimentation. Normal rodent chow was provided ad libitum and animals were maintained in a 12:12 light:dark cycle. Animals were divided into experimental and control groups, as follows: (1) experimental, 16 mice were immunized with the BHF-1.20.L peptide; (2) control, 8 mice were immunized against bovine serum albumin.
  • mice in the experimental group received subcutaneous injections (9.99 ⁇ g/g body weight; 200 ⁇ l final volume) of the BHF-1.20.L peptide daily for 2 weeks, from 5 to 7 weeks of age, prior to the initial injection with the peptide and adjuvant.
  • Blood samples pre-immune serum
  • RIBI adjuvant 50 ⁇ g of peptide/mouse
  • Titer was defined as the maximum dilution of serum that yielded a change in absorbance equivalent to 2 ⁇ that of control serum (60 min; 37° C.). The amount of peptide per well was 100 ng. Booster injections took place at 21 days interval.
  • mice were sacrificed and aorta were removed as described in Example 22.
  • Aortic atherosclerosis was measured by the cholesterol extraction method described in Example 22, whereby cholesterol is determined as a unit weight of artery.
  • mice that were immunized with the BHF-1 peptide had 24% less aortic cholesterol content (P>0.037) as compared to the control, non-immunized mice.
  • the immunization is thought to generate circulating antibodies against the BHF-1 peptide. These antibodies are thought to block LDL binding to the artery wall, thereby reducing aortic cholesterol content.

Abstract

Isolated polynucleotides encoding novel polypeptides which are capable of binding to native and methylated LDL (low density lipoprotein), the isolated polypeptides, called LBPs (LDL binding proteins), and biologically active fragments and analogs thereof, are described. Also described are methods for determining if an animal is at risk for atherosclerosis, methods for evaluating an agent for use in treating atherosclerosis, methods for treating atherosclerosis, and methods for treating a cell having an abnormality in structure or metabolism of LBP. Pharmaceutical compositions and vaccine compositions are also provided.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. Ser. No. 09/517,849, filed Mar. 2, 2000, which was a continuation-in part of U.S. Ser. No. 08/979,608, filed Nov. 26, 1997, which claimed priority from U.S. Ser. No. 60/031,930, filed Nov. 27, 1996, and U.S. Ser. No. 60/048,547, filed Jun. 3, 1997.[0001]
  • FIELD OF THE INVENTION
  • This invention relates to novel polypeptides (LBPs) which bind to low density lipoprotein (LDL), polynucleotides which encode these polypeptides, and treatments, diagnoses and therapeutic agents for atherosclerosis. [0002]
  • BACKGROUND OF THE INVENTION
  • Atherosclerosis is the principal cause of heart attacks and strokes. It has been reported that about 50% of all deaths in the United States, Europe and Japan are due to atherosclerosis. Atherosclerotic lesions in the arterial wall characterize atherosclerosis. Cholesteryl esters (CE) are present in these atherosclerotic lesions. Low density lipoprotein (LDL) has been shown to be the major carrier of plasma CE, and has been implicated as the agent by which CE enter the atherosclerotic lesions. [0003]
  • Scattered groups of lipid-filled macrophages, called foam cells, are the first visible signs of atherosclerosis and are described as type I lesions. These macrophages are reported to contain CE derived from LDL. The macrophages recognize oxidized LDL, but not native LDL, and become foam cells by phagocytosing oxidized LDL. Larger, more organized collections of foam cells, fatty streaks, represent type II lesions. These lesions further develop into complex lesions called plaques, which can result in impeding the flow of blood in the artery. [0004]
  • It is widely believed that accumulation of LDL in the artery depends on the presence of functionally modified endothelial cells in the arterial wall. It has been reported in animal models of atherosclerosis that LDL, both native LDL and methylated LDL, accumulates focally and irreversibly only at the edges of regenerating endothelial islands in aortic lesions, where functionally modified endothelial cells are present, but not in the centers of these islands where endothelial regeneration is completed. Similarly, LDL accumulates in human atherosclerotic lesions. The mechanism by which the LDL accumulates focally and irreversibly in arterial lesions has not heretofore been understood. [0005]
  • SUMMARY OF THE INVENTION
  • It is an object of the invention to provide polypeptides which bind to LDL. [0006]
  • It is yet another object of the invention to provide a method for determining if an animal is at risk for atherosclerosis. [0007]
  • It is yet another object of the invention to provide a method for evaluating an agent for use in treating atherosclerosis. It is yet another object of the invention to provide a method for treating atherosclerosis. [0008]
  • Still another object of the invention is to utilize an LBP (low density lipoprotein binding protein) gene and/or polypeptide, or fragments, analogs and variants thereof, to aid in the treatment, diagnosis and/or identification of therapeutic agents for atherosclerosis. [0009]
  • In one aspect, the invention features an isolated polynucleotide comprising a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8; SEQ ID NO: 9; SEQ ID NO: 43; SEQ ID NO: 44; SEQ ID NO: 47 or a polynucleotide capable of hybridizing to and which is at least about 95% identical to any of the above polynucleotides and wherein the encoded polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polynucleotides wherein the encoded polypeptide is capable of binding to LDL. [0010]
  • In certain embodiments, the polynucleotide comprises the nucleic acid sequence as set forth in SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17; SEQ ID NO: 18; SEQ ID NO: 45; SEQ ID NO: 46; SEQ ID NO: 48. [0011]
  • Another aspect of the invention is an isolated polypeptide comprising a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8; SEQ ID NO: 9; SEQ ID NO: 43; SEQ ID NO: 44; SEQ ID NO: 47; or a polypeptide which is at least about 95% identical to any of the above polypeptides and wherein the polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL. [0012]
  • Another aspect of the invention is a method for determining if an animal is at risk for atherosclerosis. An animal is provided. An aspect of LBP metabolism or structure is evaluated in the animal. An abnormality in the aspect of LBP metabolism or structure is diagnostic of being at risk for atherosclerosis. Another aspect of the invention is a method for evaluating an agent for use in treating atherosclerosis. A test cell, cell-free system or animal is provided. An agent is provided. The agent is administered to the test cell, cell-free system or animal in a therapeutically effective amount. The effect of the agent on an aspect of LBP metabolism or structure is evaluated. A change in the aspect of LBP metabolism or structure is indicative of the usefulness of the agent in treating atherosclerosis. [0013]
  • Another aspect of the invention is a method for evaluating an agent for the ability to alter the binding of LBP polypeptide to a binding molecule, e.g., native LDL, modified LDL, e.g., methylated LDL or oxidized LDL, or an arterial extracellular matrix structural component. An agent is provided. An LBP polypeptide is provided. A binding molecule is provided. The agent, LBP polypeptide and binding molecule are combined. The formation of a complex comprising the LBP polypeptide and binding molecule is detected. An alteration in the formation of the complex in the presence of the agent as compared to in the absence of the agent is indicative of the agent altering the binding of the LBP polypeptide to the binding molecule. [0014]
  • Another aspect of the invention is a method for evaluating an agent for the ability to bind to an LBP polypeptide. An agent is provided. An LBP polypeptide is provided. The agent is contacted with the LBP polypeptide. The ability of the agent to bind to the LBP polypeptide is evaluated. [0015]
  • Another aspect of the invention is a method for evaluating an agent for the ability to bind to a nucleic acid encoding an LBP regulatory sequence. An agent is provided. A nucleic acid encoding an LBP regulatory sequence is provided. The agent is contacted with the nucleic acid. The ability of the agent to bind to the nucleic acid is evaluated. [0016]
  • Another aspect of the invention is a method for treating atherosclerosis in an animal. An animal in need of treatment for atherosclerosis is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the animal in a therapeutically effective amount such that treatment of the atherosclerosis occurs. In certain embodiments, the agent is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof. In certain embodiments, the agent is a polypeptide of no more than about 100, 50, 30, 20, 10, 5, 4, 3 or 2 amino acid residues in length. In certain embodiments, the agent is a polypeptide having an amino acid sequence that includes at least about 20%, 40%, 60%, 80%, 90%, 95% or 98% acidic amino acid residues. [0017]
  • Another aspect of the invention is a method for treating an animal at risk for atherosclerosis. An animal at risk for atherosclerosis is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the animal in a therapeutically effective amount such that treatment of the animal occurs. [0018]
  • Another aspect of the invention is a method for treating a cell having an abnormality in structure or metabolism of LBP. A cell having an abnormality in structure or metabolism of LBP is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the cell in a therapeutically effective amount such that treatment of the cell occurs. [0019]
  • Another aspect of the invention is a pharmaceutical composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier. [0020]
  • Another aspect of the invention is a vaccine composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier. [0021]
  • Another aspect of the invention is a method for diagnosing atherosclerotic lesions in an animal. An animal is provided. A labeled agent capable of binding to LBP, e.g., LBP-1, LBP-2 or LBP-3, present in atherosclerotic lesions is provided. The labeled agent is administered to the animal under conditions which allow the labeled agent to interact with the LBP so as to result in labeled LBP. The localization or quantification of the labeled LBP is determined by imaging so as to diagnose the presence of atherosclerotic lesions in the animal. [0022]
  • Another aspect of the invention is a method for immunizing an animal against an LBP, e.g., LBP-1, LBP-2 or LBP-3, or fragment or analog thereof. An animal having LDL is provided. The LBP or fragment or analog thereof is administered to the animal so as to stimulate antibody production by the animal to the LBP or fragment or analog thereof such that binding of the LBP to the LDL is altered, e.g., decreased or increased. [0023]
  • Another aspect of the invention is a method of making a fragment or analog of LBP polypeptide, the fragment or analog having the ability to bind to native LDL and to modified LDL, e.g., methylated LDL, oxidized LDL, acetylated LDL, or cyclohexanedione-treated LDL. An LBP polypeptide is provided. The sequence of the LBP polypeptide is altered. The altered LBP polypeptide is tested for the ability to bind to modified LDL and native LDL. [0024]
  • Yet another aspect of the invention is a method for isolating a cDNA encoding an LBP. A cDNA library is provided. The cDNA library is screened for a cDNA encoding a polypeptide which binds to native LDL and modified LDL, e.g., methylated LDL or oxidized LDL. The cDNA which encodes the polypeptide is isolated, the cDNA encoding an LBP. [0025]
  • The above and other features, objects and advantages of the present invention will be better understood by a reading of the following specification in conjunction with the drawings.[0026]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the amino acid sequence of rabbit LBP-1 (SEQ ID NO: 1). Differences in amino acids between rabbit and human LBP-1 are depicted in bold type. [0027]
  • FIG. 2A depicts the nucleotide sequence (SEQ ID NO: 48) and amino acid sequence (SEQ ID NO: 47) of rabbit LBP-2. [0028]
  • FIG. 2B depicts a portion of the amino acid sequence of rabbit LBP-2 (SEQ ID NO: 2). Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. Where the sequences depicted in FIG. 2A and FIG. 2B differ, FIG. 2A represents the rabbit LBP-2 sequence. [0029]
  • FIG. 3 depicts the amino acid sequence of amino acids 319 to 350 of rabbit LBP-2 (SEQ ID NO: 3). [0030]
  • FIG. 4 depicts the amino acid sequence of amino acids 299 to 350 of rabbit LBP-2 (SEQ ID No: 4). [0031]
  • FIG. 5 depicts the amino acid sequence of rabbit LBP-3 (SEQ ID NO: 5). Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. [0032]
  • FIG. 6 depicts the amino acid sequence of human LBP-1 (SEQ ID NO: 6). Differences in amino acids between rabbit and human LBP-1 are depicted in bold type. [0033]
  • FIG. 7A depicts the nucleotide sequence (SEQ ID NO: 45) and amino acid sequence (SEQ ID NO: 43) of human LBP-2. [0034]
  • FIG. 7B depicts the amino acid sequence of amino acids 322 to 538 of human LBP-2 (SEQ ID NO: 7). Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. [0035]
  • FIG. 8A depicts the nucleotide sequence (SEQ ID NO: 46) and amino acid sequence (SEQ ID NO: 44) of human LBP-3. [0036]
  • FIG. 8B depicts the amino acid sequence of amino acids 17 to 546 of human LBP-3 (SEQ ID NO: 8). Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. Where the sequences depicted in FIG. 8A and FIG. 8B differ, FIG. 8A represents the human LBP-3 sequence. [0037]
  • FIG. 9 depicts the amino acid sequence of amino acids 14 to 33 of human or rabbit LBP-1, called BHF-1 (SEQ ID NO: 9). [0038]
  • FIG. 10 depicts the cDNA sequence encoding rabbit LBP-1 (SEQ ID NO: 10) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-1 are depicted in bold type. [0039]
  • FIG. 11 depicts a cDNA sequence encoding a portion of rabbit LBP-2 (SEQ ID NO: 11) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. Where the sequences depicted in FIG. 2A and FIG. 11 differ, FIG. 2A represents the rabbit LBP-2 sequence. [0040]
  • FIG. 12 depicts a cDNA sequence of [0041] nucleotides 256 to 1617 (SEQ ID NO: 12) of SEQ ID NO: 11 of rabbit LBP-2 and the corresponding amino acid sequence.
  • FIG. 13 depicts a cDNA sequence of [0042] nucleotides 196 to 1617 (SEQ ID NO: 13) of SEQ ID NO: 11 of rabbit LBP-2 and the corresponding amino acid sequence.
  • FIG. 14 depicts the cDNA sequence encoding rabbit LBP-3 (SEQ ID NO: 14) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. [0043]
  • FIG. 15 depicts the cDNA sequence encoding human LBP-1 (SEQ ID NO: 15) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-1 are depicted in bold type. [0044]
  • FIG. 16 depicts a cDNA sequence encoding a portion of human LBP-2 (SEQ ID NO: 16) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-2 are depicted in bold type. [0045]
  • FIG. 17 depicts a cDNA sequence encoding a portion of human LBP-3 (SEQ ID NO: 17) and the corresponding amino acid sequence. Differences in amino acids between rabbit and human LBP-3 are depicted in bold type. Where the sequences depicted in FIG. 8A and FIG. 17 differ, FIG. 8A represents the human LBP-3 sequence. [0046]
  • FIG. 18 depicts the cDNA sequence encoding BHF-1 (SEQ ID NO: 18). [0047]
  • FIG. 19 corresponds to the amino acid sequence of rabbit LBP-1 (top sequence) in alignment with the amino acid sequence of human LBP-1 (bottom sequence). [0048]
  • FIG. 20 corresponds to the amino acid sequence of a portion of the amino acid sequence of rabbit LBP-2 (top sequence) in alignment with a portion of the amino acid sequence of human LBP-2 (bottom sequence). [0049]
  • FIG. 21 corresponds to the amino acid sequence of rabbit LBP-3 (top sequence) in alignment with the amino acid sequence of a portion of human LBP-3 (bottom sequence). [0050]
  • FIG. 22 depicts the genomic sequence of human LBP-1. [0051]
  • FIG. 23 depicts the genomic sequence of human LBP-2. [0052]
  • FIG. 24 depicts the genomic sequence of human LBP-3.[0053]
  • DETAILED DESCRIPTION
  • In accordance with aspects of the present invention, there are provided novel mature human and rabbit polypeptides, LBP-1, LBP-2 and LBP-3, and biologically active analogs and fragments thereof, and there are provided isolated polynucleotides which encode such polypeptides. LBP is an abbreviation for low density lipoprotein (LDL) binding protein. The terms polynucleotide, nucleotide and oligonucleotide are used interchangeably herein, and the terms polypeptides, proteins and peptides are used interchangeably herein. [0054]
  • This invention provides for an isolated polynucleotide comprising a polynucleotide encoding the polypeptide having the amino acid sequence of rabbit LBP-1 as set forth in FIG. 1 (SEQ ID NO: 1); rabbit LBP-2 as set forth in FIG. 2A (SEQ ID NO: 47); a portion of rabbit LBP-2 as set forth in FIG. 2B (SEQ ID NO: 2); 319 to 350 of rabbit LBP-2 as set forth in FIG. 3 (SEQ ID NO: 3); 299 to 350 of rabbit LBP-2 as set forth in FIG. 4 (SEQ ID NO: 4); rabbit LBP-3 as set forth in FIG. 5 (SEQ ID NO: 5); human LBP-1 as set forth in FIG. 6 (SEQ ID NO: 6); human LBP-2 as set forth in FIG. 7A (SEQ ID NO: 43); 322 to 538 of human LBP-2 as set forth in FIG. 7B (SEQ ID NO: 7); human LBP-3 as set forth in FIG. 8A (SEQ ID NO: 44); 17-546 of human LBP-3 as set forth in FIG. 8B (SEQ ID NO: 8); 14 to 33 of human or rabbit LBP-1, called BHF-1, as set forth in FIG. 9 (SEQ ID NO: 9); a polynucleotide capable of hybridizing to and which is at least about 80% identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to any of the above polynucleotides, and wherein the encoded polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polynucleotides wherein the encoded polypeptide is capable of binding to LDL. [0055]
  • This invention also includes an isolated polynucleotide comprising a polynucleotide encoding the polypeptide having amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) of human LBP-2 as set forth in FIG. 7A (SEQ ID NO: 43); amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) of rabbit or human LBP-I as set forth in FIG. 1 (SEQ ID NO: 1) and FIG. 6 (SEQ ID NO: 6); amino acid residues 338-353 (SEQ ID NO: 25), 338-365 (SEQ ID NO: 26), 354-365 (SEQ ID NO: 27) or 444-453 (SEQ ID NO: 28) of rabbit LBP-2 as set forth in FIG. 2A (SEQ ID NO: 47); amino acid residues 96-110 (SEQ ID NO: 29) of rabbit LBP-3 as set forth in FIG. 5 (SEQ ID NO: 5); amino acid residues 69-75 (SEQ ID NO: 41) of human LBP-3 as set forth in FIG. 8A (SEQ ID NO: 44); a polynucleotide capable of hybridizing to and which is at least about 80% identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to any of the above polynucleotides, and wherein the encoded polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polynucleotides wherein the encoded polypeptide is capable of binding to LDL. [0056]
  • By a polynucleotide encoding a polypeptide is meant a polynucleotide which includes only coding sequence for the polypeptide, as well as a polynucleotide which includes additional coding and/or non-coding sequences. Thus, e.g., the polynucleotides which encode for the mature polypeptides of FIGS. [0057] 1-9 (SEQ ID NOS: 1-9, 43, 44 and 47) may include only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence; the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequences 5′ and/or 3′ of the coding sequence for the mature polypeptide. The polynucleotides of the invention are also meant to include polynucleotides in which the coding sequence for the mature polypeptide is fused in the same reading frame to a polynucleotide sequence which aids in expression and/or secretion of a polypeptide from a host cell, e.g., a leader sequence. The polynucleotides are also meant to include polynucleotides in which the coding sequence is fused in frame to a marker sequence which, e.g., allows for purification of the polypeptide.
  • The polynucleotides of the present invention may be in the form of RNA, DNA or PNA, e.g., cRNA, cDNA, genomic DNA, or synthetic DNA, RNA or PNA. The DNA may be double-stranded or single stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand. [0058]
  • In preferred embodiments, the polynucleotide comprises the nucleic acid of rabbit LBP-1 as set forth in FIG. 10 (SEQ ID NO: 10); rabbit LBP-2 as set forth in FIG. 2A (SEQ ID NO: 48) or FIG. 11 (SEQ ID NO: 11); [0059] nucleotide 256 to 1617 of SEQ ID NO: 11 of rabbit LBP-2 as set forth in FIG. 12 (SEQ ID NO: 12); nucleotide 196 to 1617 of SEQ ID NO: 11 of rabbit LBP-2 as set forth in FIG. 13 (SEQ ID NO: 13); rabbit LBP-3 as set forth in FIG. 14 (SEQ ID NO: 14); human LBP-1 as set forth in FIG. 15 (SEQ ID NO: 15); human LBP-2 as set forth in FIG. 7A (SEQ ID NO: 45) or FIG. 16 (SEQ ID NO: 16); human LBP-3 as set forth in FIG. 8A (SEQ ID NO: 46) or FIG. 17 (SEQ ID NO: 17); or nucleotide 97 to 156 of rabbit LBP-1 or nucleotide 157 to 216 of human LBP-1, (BHF-1), as set forth in FIG. 18 (SEQ ID NO: 18).
  • In other preferred embodiments, the polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 30 [0060]
    SEQ ID NO:30 (GAAGAGGAAGAAGATGATGATGAAGATGAAGATGAAGAAGATGAT),
    SEQ ID NO:31 (GAAGAGGAAGAAGATGATGATGAAGATGAAGATGAAGAAGA
    TGAT GTGTCAGAGGGCTCTGAAGTGCCCGAGAGTGAC),
    SEQ ID NO:32 (GTGTCAGAGGGCTCTGAAGTGCCCGAGAGTGAC),
    SEQ ID NO:33 (GAGGATGATGACCCCGATGGCTTCTTAGGC),
    SEQ ID NO:34 (GTGGACGTGGATGAATATGACGAGAACAAGTTCGTGGACGAA
    GAAGATGGGGGCGACGGCCAGGCCGGGCCCGACGAGGGCGAGGTGGAC),
    SEQ ID NO:35 (GACGAGGGCGAGGTGGAC),
    SEQ ID NO:36 (GAGGAGGAGGAGGAGGAGGAGGAAGACGACGAGGACGACG
    ACGACGAC),
    SEQ ID NO:37 (GAGGAGGAGGAGGAGGAGGAGGAAGACGACGAGGACGACG
    ACGACGACGTCGTGTCCGAGGGCTCGGAGGTGCCCGAGAGCGAT),
    SEQ ID NO:38 (GTCGTGTCCGAGGGCTCGGAGGTGCCCGAGAGCGAT),
    SEQ ID NO:39 (CCCCCCGGGAAGCCAGCCCTCCCAGGAGCC),
    SEQ ID NO:40 (GAGGATGGGGTCCAGGGTGAGCCCCCTGAACCTGAAGATGCA
    GAG), or
    SEQ ID NO:42 (CGTGATGTCTCTGAGGAGCTG).
  • The coding sequence which encodes the mature polypeptide may be identical to the coding sequences shown in FIGS. 2A, 7A, [0061] 8A and 10-18 (SEQ ID NOS: 10-18, 45, 46, and 48) or SEQ ID NOS: 30-40 or 42, or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptides as the DNA of FIGS. 2A, 7A, 8A and 10-18 (SEQ ID NOS: 10-18, 45, 46, and 48) and SEQ ID NOS: 30-40 and 42.
  • This invention also includes recombinant vectors comprising the polynucleotides described above. The vector can be, e.g., a plasmid, a viral particle or a phage. In certain embodiments, the recombinant vector is an expression vector. The vectors may also include various marker genes which are useful in identifying cells containing such vectors. [0062]
  • This invention also includes a cell comprising such a recombinant vector. The recombinant vectors described herein can be introduced into a host cell, e.g., by transformation, transfection or infection. [0063]
  • This invention also includes a method for producing an LBP comprising culturing such a cell under conditions that permit expression of the LBP. [0064]
  • This invention also includes an isolated polypeptide comprising a polypeptide having the amino acid sequence as set forth in FIG. 1 (SEQ ID NO: 1); FIG. 2A (SEQ ID NO: 47); FIG. 2B (SEQ ID NO: 2); FIG. 3 (SEQ ID NO: 3); FIG. 4 (SEQ ID NO: 4); FIG. 5 (SEQ ID NO: 5); FIG. 6 (SEQ ID NO: 6); FIG. 7A (SEQ ID NO: 43); FIG. 7B (SEQ ID No: 7); FIG. 8A (SEQ ID NO: 44); FIG. 8B (SEQ ID NO: 8); or FIG. 9 (SEQ ID NO: 9); or a polypeptide which is at least about 80% identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to the above polypeptides, and wherein said polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL. Differences in amino acids between the rabbit and human LBP-1, LBP-2 and LBP-3 genes are depicted in bold type in the figures. Differences in the amino acid sequences between rabbit and human LBP-1, LBP-2 and LBP-3 are also specifically shown in FIGS. 19, 20 and [0065] 21, respectively.
  • This invention also includes an isolated polypeptide comprising a polypeptide having amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) as set forth in FIG. 7A (SEQ ID NO: 47); amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) as set forth in FIG. 1 (SEQ ID NO: 1) and FIG. 6 (SEQ ID NO: 6); amino acid residues 338-353 (SEQ ID NO: 25), 338-365 (SEQ ID NO: 26), 354-365 (SEQ ID NO: 27) or 444-453 (SEQ ID NO: 28) as set forth in FIG. 2A (SEQ ID NO: 47); amino acid residues 96-110 (SEQ ID NO: 29) as set forth in FIG. 5 (SEQ ID NO: 5); and amino acid residues 69-75 (SEQ ID NO: 41) as set forth in FIG. 8A (SEQ ID NO: 8); or a polypeptide which is at least about 80%identical, more preferably at least about 90% identical, more preferably yet at least about 95% identical, and most preferably at least about 98% identical to the above polypeptides, and wherein said polypeptide is capable of binding to LDL; or a biologically active fragment of any of the above polypeptides wherein the fragment is capable of binding to LDL. [0066]
  • The polypeptides of the invention are meant to include, e.g., a naturally purified product, a chemically synthesized product, and a recombinantly derived product. [0067]
  • The polypeptides can be used, e.g., to bind to LDL, thereby inhibiting formation of atherosclerotic plaques. The polypeptides can also be used, e.g., in gene therapy, by expression of such polypeptides in vivo. The polypeptides can also be used in pharmaceutical or vaccine compositions. The polypeptides can also be used as immunogens to produce antibodies thereto, which in turn, can be used as antagonists to the LBP polypeptides. [0068]
  • Without being bound by any theory, it is believed that the LBPs provide the mechanism by which atherosclerosis is promoted through LDL oxidation. The LBPs are believed to be required in order for focal, irreversible LDL binding to occur at the arterial wall, and that such binding is a critical early event in atherosclerosis because it allows the time necessary for LDL to be changed from its native state to a fully oxidized state. [0069]
  • Since oxidized, but not native, LDL is a foreign protein, macrophages ingest it, first becoming the foam cells of type I lesions, and subsequently forming the fatty streaks of type II lesions. [0070]
  • This invention also includes a method for determining if an animal is at risk for atherosclerosis. An animal is provided. An aspect of LBP metabolism or structure is evaluated in the animal. An abnormality in the aspect of LBP metabolism or structure is diagnostic of being at risk for atherosclerosis. [0071]
  • By atherosclerosis is meant a disease or condition which comprises several stages which blend imperceptibly into each other, including irreversible binding of LDL, LDL oxidation, macrophage recruitment, blockage of the artery and tissue death (infarction). [0072]
  • By animal is meant human as well as non-human animals. Nonhuman animals include, e.g., mammals, birds, reptiles, amphibians, fish. insects and protozoa. Preferably, the nonhuman animal is a mammal, e.g., a rabbit, a rodent, e.g., a mouse, rat or guinea pig, a primate, e.g., a monkey, or a pig. An animal also includes transgenic non-human animals. The term transgenic animal is meant to include an animal that has gained new genetic information from the introduction of foreign DNA, i.e., partly or entirely heterologous DNA, into the DNA of its cells; or introduction of a lesion, e.g., an, in vitro induced mutation, e.g., a deletion or other chromosomal rearrangement into the DNA of its cells; or introduction of homologous DNA into the DNA of its cells in such a way as to alter the genome of the cell into which the DNA is inserted, e.g., it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout or replacement of the homologous host gene or results in altered and/or regulatable expression and/or metabolism of the gene. The animal may include a transgene in all of its cells including germ line cells, or in only one or some of its cells. Transgenic animals of the invention can serve as a model for studying atherosclerosis or for evaluating agents to treat atherosclerosis. [0073]
  • In certain embodiments, the determination for being at risk for atherosclerosis is done in a prenatal animal. [0074]
  • By LBP is meant a low density lipoprotein (LDL) binding protein which is capable of binding LDL and methylated LDL. By methylated LDL is meant that about 50% to about 90% of the lysine residues of LDL have a methyl group chemically attached. Methylated LDL is not recognized by previously reported cell surface receptors. See, eg., Weisgraber et al., J. Biol. Chem. 253: 9053-9062 (1978). In certain embodiments, the LBP is also capable of binding oxidized LDL. In certain preferred embodiments, the binding of LDL to an LBP is irreversible. In certain preferred embodiments, the LBP does not transport the LDL to any intracellular compartment. Examples of LBPs are LBP-1, LBP-2 and LBP-3 described herein. [0075]
  • By LBP metabolism is meant any aspect of the production, release, expression, function, action, interaction or regulation of LBP. The metabolism of LBP includes modifications, e.g., covalent or non-covalent modifications, of LBP polypeptide. The metabolism of LBP includes modifications, e.g., covalent or noncovalent modifications, that LBP induces in other substances. The metabolism of LBP also includes changes in the distribution of LBP polypeptide, and changes LBP induces in the distribution of other substances. [0076]
  • Any aspect of LBP metabolism can be evaluated. The methods used are standard techniques known to those skilled in the art and can be found in standard references, e.g., Auaubel et al., ed., Current Protocols in Mol. Biology, New York: John Wiley & Sons, 1990; Kriegler, M., ed., Gene Transfer and Expression, Stockton Press, New York, N.Y., 1989; pDisplay gene expression system (Invitrogen, Carlsbad, Calif.). Preferred examples of LBP metabolism that can be evaluated include the binding activity of LBP polypeptide to a binding molecule, e.g., LDL; the transactivation activity of LBP polypeptide on a target gene; the level of LBP protein; the level of LBP mRNA; the level of LBP modifications, e.g., phosphorylation, glycosylation or acylation; or the effect of LBP expression on transfected mammalian cell binding of LDL. [0077]
  • By binding molecule is meant any molecule to which LBP can bind, e.g., a nucleic acid, e.g., a DNA regulatory region, a protein, e.g., LDL, a metabolite, a peptide mimetic, a non-peptide mimetic, an antibody, or any other type of ligand. In certain preferred embodiments, the aspect of LBP metabolism that is evaluated is the ability of LBP to bind to native LDL and/or methylated LDL and/or oxidized LDL. Binding to LDL can be shown, e.g., by antibodies against LDL, affinity chromatography, affinity coelectrophoresis (ACE) assays, or ELISA assays. See Examples. In other embodiments, it is the ability of LBP to bind to an arterial extracellular matrix structural component that is evaluated. Examples of such components include proteoglycans, e.g., chondroitin sulfate proteoglycans and heparin sulfate proteoglycans; elastin; collagen; fibronectin; vitronectin; integrins; and related extracellular matrix molecules. Binding to arterial extracellular matrix structural components can be shown by standard methods known to those skilled in the art, e.g., by ELISA assays. Primary antibodies to the LBP are then added, followed by an enzyme-conjugated secondary antibody to the primary antibody, which produces a stable color in the presence of an appropriate substrate, and color development on the plates is measured in a microtiter plate reader. [0078]
  • Transactivation of a target gene by LBP can be determined, e.g., in a transient transfection assay in which the promoter of the target gene is linked to a reporter gene, e.g., β-galactosidase or luciferase, and co-transfected with an LBP expression vector. Such evaluations can be done in vitro or in vivo. Levels of LBP protein, mRNA or phosphorylation, can be measured, e.g., in a sample, e.g., a tissue sample, e.g., arterial wall, by standard methods known to those skilled in the art. [0079]
  • In certain embodiments, an aspect of LBP structure is evaluated, e.g., LBP gene structure or LBP protein structure. For example, primary, secondary or tertiary structures can be evaluated. For example, the DNA sequence of the gene is determined and/or the amino acid sequence of the protein is determined. Standard cloning and sequencing methods can be used as are known to those skilled in the art. In certain embodiments, the binding activity of an antisense nucleic acid with the cellular LBP mRNA and/or genomic DNA is determined using standard methods known to those skilled in the art so as to detect the presence or absence of the target mRNA or DNA sequences to which the antisense nucleic acid would normally specifically bind. [0080]
  • The risk for atherosclerosis that is determined can be a reduced risk or an increased risk as compared to a normal animal. For example, an abnormality which would give a reduced risk is an inactive LBP polypeptide. An abnormality which would give an increased risk would be, e.g., an LBP polypeptide that has higher activity, e.g., LDL binding activity, than native LBP polypeptide. [0081]
  • The invention also includes a method for evaluating an agent for use in treating atherosclerosis. A test cell, cell-free system or animal is provided. An agent is provided. The agent is administered to the test cell, cell-free system or animal in a therapeutically effective amount. The effect of the agent on an aspect of LBP metabolism or structure is evaluated. A change in the aspect of LBP metabolism or structure is indicative of the usefulness of the agent in treating atherosclerosis. [0082]
  • In certain embodiments, the method employs two phases for evaluating an agent for use in treating atherosclerosis, an initial in vitro phase and then an in vivo phase. The agent is administered to the test cell or cell-free system in vitro, and if a change in an aspect of LBP metabolism occurs, then the agent is further administered to a test animal in a therapeutically effective amount and evaluated in vivo for an effect of the agent on an aspect of LBP metabolism. [0083]
  • By cell is meant a cell or a group of cells, or a cell that is part of an animal. The cell can be a human or non-human cell. Cell is also meant to include a transgenic cell. The cell can be obtained, e.g., from a culture or from an animal. Animals are meant to include, e.g., natural animals and non-human transgenic animals. In certain embodiments, the transgenic cell or nonhuman transgenic animal has an LBP transgene, or fragment or analog thereof. In certain embodiments, the transgenic cell or non-human transgenic animal has a knockout for the LBP gene. [0084]
  • The test cell, cell-free system or animal can have a wild type pattern or a non-wild type pattern of LBP metabolism. A non-wild type pattern of LBP metabolism can result, e.g., from under-expression, over-expression, no expression, or a temporal, site or distribution change. Such a non-wild type pattern can result, e.g., from one or more mutations in the LBP gene, in a binding molecule gene, a regulatory gene. or in any other gene which directly or indirectly affects LBP metabolism. A mutation is meant to include, e.g., an alteration, e.g., in gross or fine structure, in a nucleic acid. Examples include single base pair alterations, e.g., missense or nonsense mutations, frameshifts, deletions, insertions and translocations. Mutations can be dominant or recessive. Mutations can be homozygous or heterozygous. Preferably, an aspect of LBP-1, LBP-2 or LBP-3 metabolism is evaluated. [0085]
  • An agent is meant to include, e.g., any substance, e.g., an anti-atherosclerosis drug. The agent of this invention preferably can change an aspect of LBP metabolism. Such change can be the result of any of a variety of events, including, e.g., preventing or reducing interaction between LBP and a binding molecule, e.g., LDL or an arterial extracellular matrix structural component; inactivating LBP and/or the binding molecule, e.g., by cleavage or other modification; altering the affinity of LBP and the binding molecule for each other; diluting out LBP and/or the binding molecule; preventing expression of LBP and/or the binding molecule; reducing synthesis of LBP and/or the binding molecule; synthesizing an abnormal LBP and/or binding molecule; synthesizing an alternatively spliced LBP and/or binding molecule; preventing or reducing proper conformational folding of LBP and/or the binding molecule; modulating the binding properties of LBP and/or the binding molecule; interfering with signals that are required to activate or deactivate LBP and/or the binding molecule; activating or deactivating LBP and/or the binding molecule in such a way as to prevent binding; or interfering with other receptors, ligands or other molecules which are required for the normal synthesis or functioning of LBP and/or the binding molecule. For example, the agent can block the binding site on LDL for LBPs expressed focally in the arterial wall extracellular matrix, or it could block the binding site on an LBP for LDL, or it could be bifunctional, i.e., it could block both binding sites. [0086]
  • Examples of agents include LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof, a nucleic acid encoding LBP polypeptide or a biologically active fragment or analog thereof; a nucleic acid encoding an LBP regulatory sequence or a biologically active fragment or analog thereof; a binding molecule for LBP polypeptide; a binding molecule for LBP nucleic acid, the LBP nucleic acid being, e.g., a nucleic acid comprising a regulatory region for LBP or a nucleic acid comprising a structural region for LBP or a biologically active fragment of LBP; an antisense nucleic acid; a mimetic of LBP or a binding molecule; an antibody for LBP or a binding molecule; a metabolite; or an inhibitory carbohydrate or glycoprotein. In certain embodiments, the agent is an antagonist, agonist or super agonist. [0087]
  • Knowledge of the existence of the sequence of the LBPs allows a search for natural or artificial ligands to regulate LDL levels in the treatment of atherosclerosis. In certain embodiments, the agent is a natural ligand for LBP. In certain embodiments, the agent is an artificial ligand for LBP. [0088]
  • By analog is meant a compound that differs from naturally occurring LBP in amino acid sequence or in ways that do not involve sequence, or both. Analogs of the invention generally exhibit at least about 80% homology, preferably at least about 90% homology, more preferably yet at least about 95% homology, and most preferably at least about 98% homology, with substantially the entire sequence of a naturally occurring LBP sequence, preferably with a segment of about 100 amino acid residues, more preferably with a segment of about 50 amino acid residues, more preferably yet with a segment of about 30 amino acid residues, more preferably yet with a segment of about 20 amino acid residues, more preferably yet with a segment of about 10 amino acid residues, more preferably yet with a segment of about 5 amino acid residues, more preferably yet with a segment of about 4 amino acid residues, more preferably yet with a segment of about 3 amino acid residues, and most preferably with a segment of about 2 amino acid residues. Non-sequence modifications include, e.g., in vivo or in vitro chemical derivatizations of LBP. Non-sequence modifications include, e.g., changes in phosphorylation, acetylation, methylation, carboxylation, or glycosylation. Methods for making such modifications are known to those skilled in the art. For example, phosphorylation can be modified by exposing LBP to phosphorylation-altering enzymes, e.g., kinases or phosphatases. Preferred analogs include LBP or biologically active fragments thereof whose sequences differ from the wild-type sequence by one or more conservative amino acid substitutions or by one or more non-conservative amino acid substitutions, deletions, or insertions which do not abolish LBP biological activity. Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics, e.g., substitutions within the following groups: valine, glycine; glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. Other examples of conservative substitutions are shown in Table 1. [0089]
    TABLE 1
    CONSERVATIVE AMINO ACID SUBSTITUTIONS
    For Amino Acid Code Replace with any of
    Alanine A D-Ala, Gly, beta-Ala, L-Cys, D-Cys
    Arginine R D-Arg, Lys, D-Lys, homo-Arg, D-homo-Arg,
    Met, Ile, D-Met, D-Ile, Orn, D-Orn, L-
    NMMA, L-NAME
    Asparagine N D-Asn, Asp, D-Asp, Glu, D-Glu, Gln, D-Gln
    Aspartic Acid D D-Asp, D-Asn, Asn, Glu, D-Glu, Gln, D-Gln
    Cysteine C D-Cys, S-Me-Cys, Met, D-Met, Thr, D-Thr
    Glutamine Q D-Gln, Asn, D-Asn, Glu, D-Glu, Asp, D-Asp
    Glutamic Acid E D-Glu, D-Asp, Asp, Asn, D-Asn, Gln, D-Gln
    Glycine G Ala, D-Ala, Pro, D-Pro, β-Ala Acp
    Histidine H D-His
    Isoleucine I D-Ile, Val, D-Val, Leu, D-Leu, Met, D-Met
    Leucine L D-Leu, Val, D-Val, Leu, D-Leu, Met, D-Met
    Lysine K D-Lys, Arg, D-Arg, homo-Arg, D-homo-Arg,
    Met, D-Met, Ile, D-Ile, Orn, D-Orn
    Methionine M D-Met, S-Me-Cys, Ile, D-Ile, Leu, D-Leu,
    Val, D-Val
    Phenylalanine F D-Phe, Tyr, D-Thr, L-Dopa, His, D-His, Trp,
    D-Trp, Trans-3,4, or 5-phenylproline, cis-3,4,
    or 5-phenylproline
    Proline P D-Pro, L-I-thioazolidine-4-carboxylic acid, D-
    or L-1-oxazolidine-4-carboxylic acid
    Serine S D-Ser, Thr, D-Thr, allo-Thr, Met, D-Met,
    Met(O), D-Met(O), L-Cys, D-Cys
    Threonine T D-Thr, Ser, D-Ser, allo-Thr, Met, D-Met,
    Met(O), D-Met(O), Val, D-Val
    Tryptophan W D-Trp, Phe, D-Phe, Tyr, D-Tyr
    Tyrosine Y D-Tyr, Phe, D-Phe, L-Dopa, His, D-His
    Valine V D-Val, Leu, D-Leu, Ile, D-Ile, Met, D-Met
  • Amino acid sequence variants of a protein can be prepared by any of a variety of methods known to those skilled in the art. For example, random mutagenesis of DNA which encodes a protein or a particular domain or region of a protein can be used, e.g., PCR mutagenesis (using, e.g., reduced Taq polymerase fidelity to introduce random mutations into a cloned fragment of DNA; Leung et al., BioTechnique 1: 11-15 (1989)), or saturation mutagenesis (by, e.g., chemical treatment or irradiation of single-stranded DNA in vitro, and synthesis of a complementary DNA strand; Mayers et al., Science 229: 242 (1985)). Random mutagenesis can also be accomplished by, e.g., degenerate oligonucleotide generation (using, e.g., an automatic DNA synthesizer to chemically synthesize degenerate sequences; Narang, Tetrahedron 39: 3 (1983); Itakura et al., Recombinant DNA, Proc. 3rd Cleveland Sympos. Macromolecules, ed. A. G. Walton, Amsterdam: Elsevier, pp. 273-289 (1981)). Non-random or directed mutagenesis can be used to provide specific sequences or mutations in specific regions. These techniques can be used to create variants which include, e.g., deletions, insertions, or substitutions, of residues of the known amino acid sequence of a protein. The sites for mutation can be modified individually or in series, e.g., by (i) substituting first with conserved amino acids and then with more radical choices depending upon results achieved, (ii) deleting the target residue, (iii) inserting residues of the same or a different class adjacent to the located site, or (iv) combinations of the above. For example, analogs can be made by in vitro DNA sequence modifications of the sequences of FIGS. 2A, 7A, [0090] 8A, 10-18 (SEQ ID NOS: 10-18, 45, 46, and 48). For example, in vitro mutagenesis can be used to convert any of these DNA sequences into a sequence which encodes an analog in which one or more amino acid residues has undergone a replacement, e.g., a conservative replacement as described in Table 1.
  • Methods for identifying desirable mutations include, e.g., alanine scanning mutagenesis (Cunningham and Wells, Science 244: 1081-1085 (1989)), oligonucleotide-mediated mutagenesis (Adelman et al., DNA, 2: 183 (1983)); cassette mutagenesis (Wells et al., Gene 34: 315 (1985)), combinatorial mutagenesis, and phage display libraries (Ladner et al., PCT International Appln. No. WO88/06630). The LBP analogs can be tested, e.g., for their ability to bind to LDL and/or to an arterial extracellular matrix component, as described herein. Other analogs within the invention include, e.g., those with modifications which increase peptide stability. Such analogs may contain, e.g., one or more non-peptide bonds (which replace the peptide bonds) in the peptide sequence. Also included are, e.g.: analogs that include residues other than naturally occurring L-amino acids, e.g., D-amino acids or nonnaturally occurring or synthetic amino acids, e.g., β or γ amino acids; and cyclic analogs. [0091]
  • Analogs are also meant to include peptides in which structural modifications have been introduced into the peptide backbone so as to make the peptide non-hydrolyzable. Such peptides are particularly useful for oral administration, as they are not digested. Peptide backbone modifications include, e.g., modifications of the amide nitrogen, the α-carbon, the amide carbonyl, or the amide bond, and modifications involving extensions, deletions or backbone crosslinks. For example, the backbone can be modified by substitution of a sulfoxide for the carbonyl, by reversing the peptide bond, or by substituting a methylene for the carbonyl group. Such modifications can be made by standard procedures known to those skilled in the art. , e.g. Spatola, A. F., “Peptide Backbone Modifications: A Structure-Activity Analysis of Peptides Containing Amide Bond Surrogates, Conformational Constraints, and Related Backbone Replacements,” in Chemistry and Biochemistry of Amino Acids, Peptides and Proteins, Vol. 7, pp. 267-357, B. Weinstein (ed.), Marcel Dekker, Inc., New York (1983). [0092]
  • An analog is also meant to include polypeptides in which one or more of the amino acid residues include a substituent group, or polypeptides which are fused with another compound, e.g., a compound to increase the half-life of the polypeptide, e.g., polyethylene glycol. [0093]
  • By fragment is meant some portion of the naturally occurring LBP polypeptide. Preferably, the fragment is at least about 100 amino acid residues, more preferably at least about 50 amino acid residues, more preferably yet at least about 30 amino acid residues, more preferably yet at least about 20 amino acid residues, more preferably yet at least about 5 amino acid residues, more preferably yet at least about 4 amino acid residues, more preferably yet at least about 3 amino acid residues, and most preferably at least about 2 amino acid residues in length. Fragments include, e.g., truncated secreted forms, proteolytic fragments, splicing fragments, other fragments, and chimeric constructs between at least a portion of the relevant gene, e.g., LBP-1, LBP-2 or LBP-3, and another molecule. Fragments of LBP can be generated by methods known to those skilled in the art. In certain embodiments, the fragment is biologically active. The ability of a candidate fragment to exhibit a biological activity of LBP can be assessed by methods known to those skilled in the art. For example, LBP fragments can be tested for their ability to bind to LDL and/or to an arterial extracellular matrix structural component, as described herein. Also included are LBP fragments containing residues that are not required for biological activity of the fragment or that result from alternative mRNA splicing or alternative protein processing events. [0094]
  • Fragments of a protein can be produced by any of a variety of methods known to those skilled in the art, e.g., recombinantly, by proteolytic digestion, or by chemical synthesis. Internal or terminal fragments of a polypeptide can be generated by removing one or more nucleotides from one end (for a terminal fragment) or both ends (for an internal fragment) of a nucleic acid which encodes the polypeptide. Expression of the mutagenized DNA produces polypeptide fragments. Digestion with “end-nibbling” endonucleases can thus generate DNAs which encode an array of fragments. DNAs which encode fragments of a protein can also be generated; e.g., by random shearing, restriction digestion or a combination of the above-discussed methods. For example, fragments of LBP can be made by expressing LBP DNA which has been manipulated in vitro to encode the desired fragment, e.g., by restriction digestion of any of the DNA sequences of FIGS. 2A, 7A, [0095] 8A, 10-18 (SEQ ID NOS: 10-18, 45, 46, and 48).
  • Fragments can also be chemically synthesized using techniques known in the art, e.g., conventional Merrifield solid phase f-Moc or t-Boc chemistry for example, peptides of the present invention can be arbitrarily divided into fragments of desired length with no overlap of the fragments, or divided into overlapping fragments of a desired length. [0096]
  • An LBP or a biologically active fragment or analog thereof, or a binding molecule or a biologically active fragment or analog thereof, can, e.g., compete with its cognate molecule for the binding site on the complementary molecule, and thereby reduce or eliminate binding between LBP and the cellular binding molecule. LBP or a binding molecule can be obtained, e.g., from purification or secretion of naturally occurring LBP or binding molecule, from recombinant LBP or binding molecule, or from synthesized LBP or binding molecule. [0097]
  • Therefore, methods for generating analogs and fragments and testing them for activity are known to those skilled in the art. [0098]
  • An agent can also be a nucleic acid used as an antisense molecule. Antisense therapy is meant to include, e.g., administration or in situ generation of oligonucleotides or their derivatives which specifically hybridize, e.g., bind, under cellular conditions, with the cellular mRNA and/or genomic DNA encoding an LBP polypeptide, or mutant thereof, so as to inhibit expression of the encoded protein, e.g., by inhibiting transcription and/or translation. The binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix. [0099]
  • In certain embodiments, the antisense construct binds to a naturally-occurring sequence of an LBP gene which, e.g., is involved in expression of the gene. These sequences include, e.g., promoter, start codons, stop codons, and RNA polymerase binding sites. In other embodiments, the antisense construct binds to a nucleotide sequence which is not present in the wild type gene. For example, the antisense construct can bind to a region of an LBP gene which contains an insertion of an exogenous, non-wild type sequence. Alternatively, the antisense construct can bind to a region of an LBP gene which has undergone a deletion, thereby bringing two regions of the gene together which are not normally positioned together and which, together, create a non-wild type sequence. When administered in vivo to a subject, antisense constructs which bind to non-wild type sequences provide the advantage of inhibiting the expression of a mutant LBP gene, without inhibiting expression of any wild type LBP gene. [0100]
  • An antisense construct of the present invention can be delivered, e.g., as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the cellular mRNA which encodes an LBP polypeptide. An alternative is that the antisense construct is an oligonucleotide which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA (duplexing) and/or genomic sequences (triplexing) of an LBP gene. Such oligonucleotides are preferably modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and are therefore stable in vivo. Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate, phosphorodithioates and methylphosphonate analogs of DNA and peptide nucleic acids (PNA). (See also U.S. Pat. Nos. 5,176,996; 5,264,564; and 5,256,775). Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed. (See, e.g., Van der Krol et al., Biotechniques 6: 958-976, (1988); Stein et al., Cancer Res. 48: 2659-2668 (1988)). [0101]
  • By mimetic is meant a molecule which resembles in shape and/or charge distribution LBP or a binding molecule. The mimetic can be a peptide or a non-peptide. Mimetics can act as therapeutic agents because they can, e.g., competitively inhibit binding of LBP to a binding molecule. By employing, e.g., scanning mutagenesis, e.g., alanine scanning mutagenesis, linker scanning mutagenesis or saturation mutagenesis, to map the amino acid residues of a particular LBP polypeptide involved in binding a binding molecule, peptide mimetics, e.g., diazepine or isoquinoline derivatives, can be generated which mimic those residues in binding to a binding molecule, and which therefore can inhibit binding of the LBP to a binding molecule and thereby interfere with the function of LBP. Non-hydrolyzable peptide analogs of such residues can be generated using, e.g., benzodiazepine (see, e.g., Freidinger et al., in Peptides: Chemistry and Biology, G. R. Marshall ed., ESCOM Publisher: Leiden, Netherlands (1988)); azepine (see, e.g., Huffman et al., in Peptides: Chemistry and Biology, G. R. Marshall ed., ESCOM Publisher: Leiden, Netherlands (1988)); substituted gamma lactam rings (see, e.g., Garvey et al., in Peptides: Chemistry and Biology, G. R. Marshall ed., ESCOM Publisher: Leiden, Netherlands (1988)); keto-methylene pseudopeptides (see, eg., Ewenson et al., J. Med. Chem. 29: 295 (1986); Ewenson et al., in Peptides: Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co. Rockland, Ill. (1985)); β-turn dipeptide cores (see, e.g., Nagai et al., Tetrahedron Lett. 26: 647 (1985); Sato et al., J. Chem. Soc. Perkin Trans. 1: 1231 (1986)); or β-aminoalcohols (see, eg., Gordon et al., Biochem. Biophys. Res. Commun. 126:419 (1985); Dann et al., Biochem. Biophys. Res. Commun. 134:71 (1986)). [0102]
  • Antibodies are meant to include antibodies against any moiety that directly or indirectly affects LBP metabolism. The antibodies can be directed against, e.g., LBP or a binding molecule, or a subunit or fragment thereof. For example, antibodies include anti-LBP-1, LBP-2 or LBP-3 antibodies; and anti-binding molecule antibodies. Antibody fragments are meant to include, e.g., Fab fragments, Fab′ fragments, F(ab′)[0103] 2 fragments, F(v) fragments, heavy chain monomers, heavy chain dimers, heavy chain trimers, light chain monomers, light chain dimers, light chain trimers, dimers consisting of one heavy and one light chain, and peptides that mimic the activity of the anti-LBP or anti-binding molecule antibodies. For example, Fab2′ fragments of the inhibitory antibody can be generated through, e.g., enzymatic cleavage. Both polyclonal and monoclonal antibodies can be used in this invention. Preferably, monoclonal antibodies are used. Natural antibodies, recombinant antibodies or chimeric-antibodies, e.g., humanized antibodies, are included in this invention. Preferably, humanized antibodies are used when the subject is a human. Most preferably, the antibodies have a constant region derived from a human antibody and a variable region derived from an inhibitory mouse monoclonal antibody. Production of polyclonal antibodies to LBP is described in Example 6. Monoclonal and humanized antibodies are generated by standard methods known to those skilled in the art. Monoclonal antibodies can be produced, e.g., by any technique which provides antibodies produced by continuous cell lines cultures. Examples include the hybridoma technique (Kohler and Milstein, Nature 256: 495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72 (1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., in Monoclonal Antibodies and Cancer Therapy, A. R. Lisa, Inc., pp. 77-96 (1985)). Preferably, humanized antibodies are raised through conventional production and harvesting techniques (Berkower, I., Curr. Opin. Biotechnol. 7:622-628 (1996); Ramharayan and Skaletsky, Am. Biotechnol. Lab 13:26-28 (1995)). In certain preferred embodiments, the antibodies are raised against the LBP, preferably the LDL-binding site, and the Fab fragments produced. These antibodies, or fragments derived therefrom, can be used, e.g., to block the LDL-binding sites on the LBP molecules.
  • Agents also include inhibitors of a molecule that are required for synthesis, post-translational modification, or functioning of LBP. and/or a binding molecule, or activators of a molecule that inhibits the synthesis or functioning of LBP and/or the binding molecule. Agents include, e.g., cytokines, chemokines, growth factors, hormones, signaling components, kinases, phosphatases, homeobox proteins, transcription factors, editing factors, translation factors and post-translation factors or enzymes. Agents are also meant to include ionizing radiation, non-ionizing radiation, ultrasound and toxic agents which can, e.g., at least partially inactivate or destroy LBP and/or the binding molecule. [0104]
  • An agent is also meant to include an agent which is not entirely LBP specific. For example, an agent may alter other genes or proteins related to arterial plaque formation. Such overlapping specificity may provide additional therapeutic advantage. [0105]
  • The invention also includes the agent so identified as being useful in treating atherosclerosis. [0106]
  • The invention also includes a method for evaluating an agent for the ability to alter the binding of LBP polypeptide to a binding molecule. An agent is provided. An LBP polypeptide is provided. A binding molecule is provided. The agent, LBP polypeptide and binding molecule are combined. The formation of a complex comprising the LBP polypeptide and binding molecule is detected. An alteration in the formation of the complex in the presence of the agent as compared to in the absence of the agent is indicative of the agent altering the binding of the LBP polypeptide to the binding molecule. [0107]
  • In preferred embodiments, the LBP polypeptide is LBP-1, LBP-2 or LBP-3. Examples of a binding molecule include native LDL, modified LDL, e.g., methylated LDL or oxidized LDL, and arterial extracellular matrix structural components. [0108]
  • Altering the binding includes, e.g., inhibiting or promoting the binding. The efficacy of the agent can be assessed, e.g., by generating dose response curves from data obtained using various concentrations of the agent. Methods for determining formation of a complex are standard and are known to those skilled in the art, e.g., affinity coelectrophoresis (ACE) assays or ELISA assays as described herein. [0109]
  • The invention also includes the agent so identified as being able to alter the binding of an LBP polypeptide to a binding molecule. [0110]
  • The invention also includes a method for evaluating an agent for the ability to bind to an LBP polypeptide. An agent is provided. An LBP polypeptide is provided. The agent is contacted with the LBP polypeptide. The ability of the agent to bind to the LBP polypeptide is evaluated. Preferably, the LBP polypeptide is LBP-1, LBP-2 or LBP-3. Binding can be determined, e.g., by measuring formation of a complex by standard methods known to those skilled in the art, e.g., affinity coelectrophoresis (ACE) assays or ELISA assays as described herein. [0111]
  • The invention also includes the agent so identified as being able to bind to LBP polypeptide. [0112]
  • The invention also includes a method for evaluating an agent for the ability to bind to a nucleic acid encoding an LBP regulatory sequence. An agent is provided. A nucleic acid encoding an LBP regulatory sequence is provided. The agent is contacted with the nucleic acid. The ability of the agent to bind to the nucleic acid is evaluated. Preferably, the LBP regulatory sequence is an LBP-1, LBP-2 or LBP-3 regulatory sequence. Binding can be determined, e.g., by measuring formation of a complex by standard methods known to those skilled in the art, e.g., DNA mobility shift assays, DNase I footprint analysis Molecular Biology, The invention being able to bind sequence. (Ausubel et al., ed., Current Protocols in John Wiley & Sons, New York, N.Y., (1989)). [0113]
  • The invention also includes the agent so identified as to a nucleic acid encoding an LBP regulatory sequence. [0114]
  • The invention also includes a method for treating atherosclerosis in an animal. An animal in need of treatment for atherosclerosis is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the animal in a therapeutically effective amount such that treatment of the atherosclerosis occurs. [0115]
  • In certain preferred embodiments, the agent is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof. The agent can be, e.g., the polypeptide as set forth in SEQ ID NOS: 1-9, 43, 44, and 47. Preferably, the agent is a polypeptide of no more than about 100 amino acid residues in length, more preferably of no more than about 50 amino acid residues, more preferably yet of no more than about 30 amino acid residues, more preferably yet of no more than about 20 amino acid residues, more preferably yet of no more than about 10 amino acid residues, more preferably yet of no more than about 5 amino acid residues, more preferably yet of no more than about 4 amino acid residues, more preferably yet of no more than about 3 amino acid residues, and most preferably of no more than about 2 amino acid residues. Preferably, the polypeptide includes at least about 20% acidic amino acid residues, more preferably yet at least about 40% acidic amino acid residues, more preferably yet at least about 60% acidic amino acid residues, more preferably yet at least about 80% acidic amino acid residues, more preferably yet at least about 90% acidic amino acid residues, more preferably yet at least about 95% acidic amino acid residues, and most preferably at least about 98% acidic amino acid residues. Acidic amino acid residues include aspartic acid and glutamic acid. An example of such an LBP poly-peptide is BHF-1, which is a 20 amino acid length fragment of human or rabbit LBP-1 which contains amino acid residues 14 through 33. See FIG. 9 (SEQ ID NO: 9). 45% of the amino acid residues of BHF-1 are acidic. The invention also includes biologically active fragments and analogs of BHF-1. [0116]
  • Other preferred acidic regions from the LBPs are amino acid residues 329 through 343 (SEQ ID NO: 19), 329 through 354 (SEQ ID NO: 20), 344 through 354 (SEQ ID NO: 21), and 529 through 538 (SEQ ID NO: 22) of human LBP-2 as depicted in FIG. 7A (SEQ. ID NO: 43); amino acid residues 14 through 43 (SEQ ID NO: 23)and 38 through 43 (SEQ ID NO: 24) of rabbit or human LBP-1 as depicted in FIG. 1 (SEQ ID NO: 1) and FIG. 6 (SEQ ID NO: 6); [0117] amino acid residues 338 through 353 (SEQ ID NO: 25), 338 through 365 (SEQ ID NO: 26), 354 through 365 (SEQ ID NO: 27), and 444 through 453 (SEQ ID NO: 28) of rabbit LBP-2 as depicted in FIG. 2A (SEQ ID NO: 47); amino acid residues 96 through 110 (SEQ ID NO: 29) of rabbit LBP-3 as depicted in FIG. 5 (SEQ ID NO: 5); and amino acid residues 69-75 (SEQ ID NO: 41) of human LBP-3 as depicted in FIG. 8A (SEQ ID NO: 44). The invention is also meant to include biologically active fragments and analogs of any of these polypeptides.
  • Other examples of agents include homopolymers and heteropolymers of any amino acid or amino acid analog. In certain preferred embodiments, the agent is a homopolymer of an acidic amino acid or analog thereof. In certain embodiments, the agent is a heteropolymer of one or more acidic amino acids and one or more other amino acids, or analogs thereof. For example, agents include poly(glu), poly(asp), poly(glu asp), poly(glu N), poly (asp N) and poly(glu asp N). By N is meant any amino acid, or analog thereof, other than glu or asp. By poly(glu asp) is meant all permutations of glu and asp for a given length peptide. A preferred peptide is poly(glu) of no more than about 10 amino acids in length, preferably about 7 amino acids in length. [0118]
  • In certain preferred embodiments, the agent is an LBP nucleic acid or a biologically active fragment or analog thereof, e.g., a nucleic acid encoding LBP-1, LBP-2 or LBP-3 polypeptide, or a biologically active fragment or analog thereof. The agent can be, e.g., a nucleic acid comprising a nucleotide sequence as set forth in SEQ ID NOS: 10-18, 45, 46, and 48. In other embodiments, the agent is an antisense molecule, e.g., one which can bind to an LBP gene sequence. [0119]
  • Treating is meant to include, e.g., preventing, treating, reducing the symptoms of, or curing the atherosclerosis. Administration of the agent can be accomplished by any method which allows the agent to reach the target area, e.g., a target cell or the extracellular matrix. These methods include, e.g., injection, deposition, implantation, suppositories, oral ingestion, inhalation, topical administration, or any other method of administration where access to the target area by the agent is obtained. Injections can be, e.g., intravenous, intradermal, subcutaneous, intramuscular or intraperitoneal. Implantation includes inserting implantable drug delivery systems, e.g., microspheres, hydrogels, polymeric reservoirs, cholesterol matrices, polymeric systems, e.g., matrix erosion and/or diffusion systems and non-polymeric systems, e.g., compressed, fused or partially fused pellets. Suppositories include glycerin suppositories. Oral ingestion doses can be enterically coated. Inhalation includes administering the agent with an aerosol in an inhalator, either alone or attached to a carrier that can be absorbed. [0120]
  • Administration of the agent can be alone or in combination with other therapeutic agents. In certain embodiments, the agent can be combined with a suitable carrier, incorporated into a liposome, or incorporated into a polymer release system. [0121]
  • In certain embodiments of the invention, the administration can be designed so as to result in sequential exposures to the agent over some time period, e.g., hours, days, weeks, months or years. This can be accomplished by repeated administrations of the agent by one of the methods described above, or alternatively, by a controlled release delivery system in which the agent is delivered to the animal over a prolonged period without repeated administrations. By a controlled release delivery system is meant that total release of the agent does not occur immediately upon administration, but rather is delayed for some time. Release can occur in bursts or it can occur gradually and continuously. Administration of such a system can be, e.g., by long acting oral dosage forms, bolus injections, transdermal patches or subcutaneous implants. [0122]
  • Examples of systems in which release occurs in bursts include, e.g., systems in which the agent is entrapped in liposomes which are encapsulated in a polymer matrix, the liposomes being sensitive to a specific stimulus, e.g., temperature, pH, light, magnetic field, or a degrading enzyme, and systems in which the agent is encapsulated by an ionically-coated microcapsule with a microcapsule core-degrading enzyme. Examples of systems in which release of the agent is gradual and continuous include, e.g., erosional systems in which the agent is contained in a form within a matrix, and diffusional systems in which the agent permeates at a controlled rate, e.g., through a polymer. Such sustained release systems can be, e.g., in the form of pellets or capsules. [0123]
  • The agent can be suspended in a liquid, e.g., in dissolved form or colloidal form. The liquid can be a solvent, partial solvent or non-solvent. In many cases water or an organic liquid can be used. [0124]
  • The agent can be administered prior to or subsequent to the appearance of atherosclerosis symptoms. In certain embodiments, the agent is administered to patients with familial histories of atherosclerosis, or who have phenotypes that may indicate a predisposition to atherosclerosis, or who have been diagnosed as having a genotype which predisposes the patient to atherosclerosis, or who have other risk factors, e.g., hypercholesterolemia, hypertension or smoking. [0125]
  • The agent is administered to the animal in a therapeutically effective amount. By therapeutically effective amount is meant that amount which is capable of at least partially preventing or reversing atherosclerosis. A therapeutically effective amount can be determined on an individual basis and will be based, at least in part, on consideration of the species of animal, the animal's size, the animal's age, the agent used, the type of delivery system used, the time of administration relative to the onset of atherosclerosis symptoms, and whether a single, multiple, or controlled release dose regimen is employed. A therapeutically effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation. [0126]
  • Preferably, the concentration of the agent is at a dose of about 0.1 to about 1000 mg/kg body weight/day, more preferably at about 0.1 to about 500 mg/kg/day, more preferably yet at about 0.1 to about 100 mg/kg/day, and most preferably at about 0.1 to about 5 mg/kg/day. The specific concentration partially depends upon the particular agent used, as some are more effective than others. The dosage concentration of the agent that is actually administered is dependent at least in part upon the final concentration that is desired at the site of action, the method of administration, the efficacy of the particular agent, the longevity of the particular agent, and the timing of administration relative to the onset of the atherosclerosis symptoms. Preferably, the dosage form is such that it does not substantially deleteriously affect the animal. The dosage can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation. [0127]
  • In certain embodiments, various gene constructs can be used as part of a gene therapy protocol to deliver nucleic acids encoding an agent, e.g., either an agonistic or antagonistic form of an LBP polypeptide. For example, expression vectors can be used for in vivo transfection and expression of an LBP polypeptide in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of, LBP polypeptide in a cell in which non-wild type LBP is expressed. Expression constructs of the LBP polypeptide, and mutants thereof, may be administered in any biologically effective carrier, e.g., any formulation or composition capable of effectively delivering the LBP gene to cells in vivo. Approaches include, e.g., insertion of the subject gene in viral vectors including, e.g., recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids. Viral vectors infect or transduce cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectin™ (Life Technologies, Inc., Gaithersburg, Md.) or derivatized (e.g. antibody conjugated), polylysine conjugates, gramacidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or Ca[0128] 3,(P04)2 precipitation carried out in vivo. The above-described methods are known to those skilled in the art and can be performed without undue experimentation. Since transduction of appropriate target cells represents the critical first step in gene therapy, choice of the particular gene delivery system will depend on such factors as the phenotype of the intended target and the route of administration, e.g., locally or systemically. Administration can be directed to one or more cell types, and to one or more cells within a cell type, so as to be therapeutically effective, by methods that are known to those skilled in the art. In a preferred embodiment, the agent is administered to arterial wall cells of the animal. For example, a genetically engineered LBP gene is administered to arterial wall cells. In certain embodiments, administration is done in a prenatal animal or embryonic cell. It will be recognized that the particular gene construct provided for in vivo transduction of LBP expression is also useful for in vitro transduction of cells, such as for use in the diagnostic assays described herein.
  • In certain embodiments, therapy of atherosclerosis is performed with antisense nucleotide analogs of the genes which code for the LBPs. Preferably, the antisense nucleotides have non-hydrolyzable “backbones,” e.g., phosphorothioates, phosphorodithioates or methylphosphonates. The nucleoside base sequence is complementary to the sequence of a portion of the gene coding for, e.g., LBP-1, 2 or 3. Such a sequence might be, e.g., ATTGGC if the gene sequence for the LBP is TAACCG. One embodiment of such therapy would be incorporation of an antisense analog of a portion of one of the LBP genes in a slowrelease medium, e.g., polyvinyl alcohol, which is administered, e.g., by subcutaneous injection, so as to release the antisense nucleotide analog over a period of weeks or months. In another embodiment, the antisense analog is incorporated into a polymeric matrix, e.g., polyvinyl alcohol, such that the gel can be applied locally to an injured arterial wall to inhibit LBP synthesis and prevent LDL accumulation, e.g., after angioplasty or atherectomy. [0129]
  • The invention also includes a method for treating an animal at risk for atherosclerosis. An animal at risk for atherosclerosis is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the animal in a therapeutically effective amount such that treatment of the animal occurs. Being at risk for atherosclerosis can result from, e.g., a family history of atherosclerosis, or phenotypic symptoms which predispose to atherosclerosis, e.g., having hypercholesterolemia, hypertension or smoking. [0130]
  • The invention also includes a method for treating a cell having an abnormality in structure or metabolism of LBP. A cell having an abnormality in structure or metabolism of LBP is provided. An agent capable of altering an aspect of LBP structure or metabolism is provided. The agent is administered to the cell in a therapeutically effective amount such that treatment of the cell occurs. [0131]
  • In certain embodiments, the cell is obtained from a cell culture or tissue culture or an embryo fibroblast. The cell can be, e.g., part of an animal, e.g., a natural animal or a nonhuman transgenic animal. Preferably, the LBP is LBP-1, LBP-2 or LBP-3. [0132]
  • The invention also includes a pharmaceutical composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers include, e.g., saline, liposomes and lipid emulsions. [0133]
  • In certain preferred embodiments, the agent of the pharmaceutical composition is an LBP polypeptide, e.g., LBP-1, LBP-2 or LBP-3, or a biologically active fragment or analog thereof. The agent can be, e.g., the polypeptide as set forth in SEQ ID NOS: 1-9, 43, 44, and 47. Preferably, the agent is a polypeptide of no more than about 100 amino acid residues in length, more preferably of no more than about 50 amino acid residues, more preferably yet of no more than about 30 amino acid residues, more preferably yet of no more than about 20 amino acid residues, more preferably yet of no more than about 10 amino acid residues, more preferably yet of no more than about 5 amino acid residues, more preferably yet of no more than about 4 amino acid residues, more preferably yet of no more than about 3 amino acid residues, and most preferably of no more than about 2 amino acid residues. Preferably, the polypeptide includes at least about 20% acidic amino acid residues, more preferably yet at least about 40% acidic amino acid residues, more preferably yet at least about 60% acidic amino acid residues, more preferably yet at least about 80% acidic amino acid residues, more preferably yet at least about 90% acidic amino acid residues, more preferably yet at least about 95% acidic amino acid residues, and most preferably at least about 98%acidic amino acid residues. [0134]
  • In certain preferred embodiments, the agent is an LBP nucleic acid, e.g., a nucleic acid encoding LBP-1, LBP-2 or LBP-3 polypeptide, or a biologically active fragment or analog thereof. The agent can be, e.g., a nucleic acid comprising a nucleotide sequence as set forth in SEQ ID NOS: 10-18, 45, 46, and 48. [0135]
  • The invention also includes a vaccine composition for treating atherosclerosis in an animal comprising a therapeutically effective amount of an agent, the agent being capable of altering an aspect of LBP metabolism or structure in the animal so as to result in treatment of the atherosclerosis, and a pharmaceutically acceptable carrier. [0136]
  • The invention also includes a method for diagnosing atherosclerotic lesions in an animal. An animal is provided. A labeled agent capable of binding to LBP present in atherosclerotic lesions is provided. The labeled agent is administered to the animal under conditions which allow the labeled agent to interact with the LBP so as to result in labeled LBP. The localization or quantification of the labeled LBP is determined by imaging so as to diagnose the presence of atherosclerotic lesions in the animal. [0137]
  • Preferably, the LBP is LBP-1, LBP-2 or LBP-3. The imaging can be performed by standard methods known to those skilled in the art, including, e.g., magnetic resonance imaging, gamma camera imaging, single photon emission computed tomographic (SPECT) imaging, or positron emission tomography (PET). [0138]
  • Preferably, agents that bind tightly to LBPs in atherosclerotic lesions are used for atherosclerotic imaging and diagnosis. The agent is radiolabeled with, e.g., [0139] 99mTc or another isotope suitable for clinical imaging by gamma camera, SPECT, PET scanning or other similar technology. Since LBPs occur in very early lesions, such imaging is more sensitive than angiography or ultrasound for locating very early lesions which do not yet impinge on the arterial lumen to cause a visible bulge or disturbed flow. In addition to locating both early and more developed lesions, the imaging agents which bind to LBPs can also be used to follow the progress of atherosclerosis, as a means of evaluating the effectiveness of both dietary and pharmacological treatments.
  • Thus, a diagnostic embodiment of the invention is the adaptation of, e.g., a peptide complementary to one of the LBPs, by radiolabeling it and using it as an injectable imaging agent for detection of occult atherosclerosis. The peptide is selected from those known to bind to LBPs, e.g., RRRRRRR or KKLKLXX, or any other polycationic peptide which binds to the highly electronegative domains of the LBPs. For extracorporeal detection with a gamma scintillation (Anger) camera, technetium-binding ligands, e.g., CGC, GGCGC, or GGCGCF, can be incorporated into the peptides at the N-terminus or C-terminus for [0140] 99mTc labeling. For external imaging by magnetic resonance imaging (MRI), e.g., the gadolinium-binding chelator, diethylene triamine penta-acetic acid (DTPA), is covalently bound to the N- or C-terminus of the peptides. In yet other embodiments, the LBP-binding peptides are covalently bound, e.g., to magnetic ion oxide particles by standard methods known to those skilled in the art, e.g., conjugating the peptides with activated polystyrene resin beads containing magnetic ion oxide.
  • The invention also includes a method for immunizing an animal against an LBP, e.g., LBP-1, LBP-2 or LBP-3, or fragment or analog thereof. An animal having LDL is provided. An LBP or fragment or analog thereof is provided. The LBP or fragment or analog thereof is administered to the animal so as to stimulate antibody production by the animal to the LBP or fragment or analog thereof such that binding of the LBP to the LDL is altered, e.g., decreased or increased. [0141]
  • The invention also includes a method of making a fragment or analog of LBP polypeptide, the fragment or analog having the ability to bind to modified LDL and native LDL. An LBP polypeptide is provided. The sequence of the LBP polypeptide is altered. The altered LBP polypeptide is tested for the ability to bind to modified LDL, e.g., methylated LDL, oxidized LDL, acetylated LDL, cyclohexanedione-treated LDL (CHD-LDL), and to native LDL. [0142]
  • The fragments or analogs can be generated and tested for their ability to bind to these modified LDLs and to native LDL, by methods known to those skilled in the art, e.g., as described herein. Preferably, they are tested for their ability to bind to methylated LDL and native LDL. The binding activity of the fragment or analog can be greater or less than the binding activity of the native LBP. Preferably, it is greater. In preferred embodiments, the LBP is LBP-1, LBP-2 or LBP-3. [0143]
  • The invention also includes a method for isolating a cDNA encoding an LBP. A cDNA library is provided. The cDNA library is screened for a cDNA encoding a polypeptide which binds to native LDL and modified LDL, e.g., methylated LDL or oxidized LDL. The cDNA which encodes this polypeptide is isolated, the cDNA encoding an LBP. [0144]
  • Atherosclerosis in a hyperlipidemic subject can be reduced following the generation of an immune response in the subject by immunization with LBPs. Numerous immunotherapeutic products can be used to generate antibodies that will block the binding between LDL and LBPs. [0145]
  • The injection of one or more LBPs can result in the production of anti-LBP antibodies, resulting in a reduction in ,e.g., aortic atherosclerosis. This effect is thought to be mediated by an inhibition of LBP binding to LDL. LBP immunogens that can be used in the invention include human LBPs, non-human LBPs, recombinant LBPs, and proteins structurally related to the LBPs described herein, e.g. non-naturally occurring proteins that differ from a naturally occurring LBP at one or more amino acid residues. In addition to full length proteins, injecting one or more peptides that include an LBP domain can generate an effective immune response. For example, the injection of a peptide comprising an LBP domain having LDL-binding activity can cause an organism to make antibodies to the LBP binding sites for LDL. These peptide immunogens can include sequences derived from human LBPs, non-human LBPs, recombinant LBPs, and proteins structurally related to the LBPs described herein. [0146]
  • Modifications can be made to a protein or peptide immunogen of the invention to increase its immunogenicity. The immunogen can be conjugated or coupled with a carrier, e.g. a Cholera toxin B chain or monoclonal antibodies. The immunogen can be precipitated with aluminum salts or cross-linked with formaldehyde or other aldehydes. The protein may be mixed with a physiologically acceptable diluent such as water, phosphate buffered saline, or saline. The composition may further include an adjuvant. In addition to RIBI adjuvant, adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide are all well known in the art. Adjustments in the adjuvant of the invention can be made to affect the immunogenicity of the peptide or protein. Examples of such modifications include using: aluminum salts; cytokines; MF59 (microfluidized emulsion of oil and surfactants); SAF-1 (oil-based emulsion); saponin derivatives; polymers (such as polyphosphazene); and bacterial toxins. Additional descriptions of antigenic protein-adjuvant combinations are described in WO 99/54452 (herein incorporated by reference) and WO 99/49890 (herein incorporated by reference). [0147]
  • In addition to delivery of the proteins and peptides described above, numerous other delivery systems can be used to generate the anti-atherosclerotic immunity of the invention. The LBP immunogen can be delivered either directly as a protein antigen or alternatively as a nucleic acid that encodes the protein antigen. The immunotherapeutic products of the invention, either protein or nucleic acid, can be delivered by numerous delivery routes. These include injection, deposition, implantation, suppositories, oral ingestion, inhalation (e.g., delivery via a nasal spray), and topical administration (e.g., delivery via a skin patch). [0148]
  • A nucleic acid encoding an immunogen of the invention can be directly administered, for example by injection, to tissues and expressed as a protein. The DNA or RNA can be either associated with a delivery vehicle (e.g., viruses, bacteria, liposomes, and gold beads) or naked (free from association with transfection-facilitating proteins, viral particles, liposomal formulations, charged lipids and calcium phosphate precipitating). The nucleic acid can optionally include a promoter, e.g. a viral promoter. The immunogen encoded by the nucleic acid is produced in the host, resulting in the generation of an immune response. Methods for the delivery of nucleic acid sequences encoding therapeutic proteins and peptides are described in detail by Felgner et al. (U.S. Pat. No. 25 5,580,859; herein incorporated by reference) and Barbet et al. (U.S. Pat. No. 6,025,338:; herein incorporated by reference). Vaccine compositions of viral liposomes comprising a nucleic acid, e.g. an RNA, encoding a protein antigen are described in WO 99/52503 (herein incorporated by reference). Proteins and nucleic acids encoding peptides can also be delivered to an individual by their encapsulation in liposomes, microparticles, and ISCOMS, all of which are well known in the art (see, e.g., U.S. Pat. No. 6,013,258, herein incorporated by reference). [0149]
  • A nucleic acid encoding an immunogen of the invention can also be included in the genome of a plant, so as to result in the production of the immunogen by plant tissues. The genetically modified plant may then consumed by an individual, resulting in the ingestion of the immunogen and the generation of an anti-LBP immune response. Methodology for the generation and usage of edible plant vaccines is described in WO 99/54452 (herein incorporated by reference). [0150]
  • Numerous plants may be useful for the production of an edible vaccine, including: tobacco, tomato, potato. eggplant, pepino, yam, soybean, pea, sugar beet, lettuce, bell pepper, celery, carrot, asparagus, onion, grapevine, muskmelon, strawberry, rice, sunflower, rapeseed/canola, wheat, oats, maize, cotton, walnut, spruce/conifer, poplar and apple. The edible vaccine can include a plant cell transformed with a nucleic acid construct comprising a promoter and a sequence encoding an LBP. The sequence may optionally encode a chimeric protein, comprising a cholera toxin subunit B peptide fused to the LBP peptide. Preferred plant promoters of the invention include CaMV 35S, patatin, mas, and granule-bound starch synthase promoters. Additional useful promoters and enhancers are described in WO 99/54452. [0151]
  • The edible vaccine of the invention can be administered to a mammal suffering from or at risk of atherosclerosis. Preferably, an edible vaccine is administered orally, e.g. consuming a transgenic plant of the invention. The transgenic plant can be in the form of a plant part, extract, juice, liquid, powder, or tablet. The edible vaccine can also be administered via an intranasal route. [0152]
  • Microorganisms, e.g., attenuated viruses or bacteria, can be used in the invention by including a nucleic acid encoding an LBP immunogen in the genome of the microorganism. This modified vector can then be delivered to a host, resulting in the in vivo production of the immunogen. The immune response generated by these vectors is expected to result in anti-atherosclerotic immunity. Nucleic acid molecules are inserted into microorganism genomes by standard methods known in the art (U.S. Pat. No. 5,866,136 and U.S. Pat. No. 6,025,164, both of which are herein incorporated by reference) [0153]
  • The anti-atherosclerotic methods of the invention are directed to treating a subject, e.g., a human, primate, horse, dog, cat, or goat, at risk for atherosclerosis by stimulating an anti-LBP response in the subject by immunotherapy. The LBP proteins and peptides of the invention may be delivered to the subject by the numerous delivery systems described herein. The immunotherapy may comprise an initial immunization followed by additional, e.g. one, two, or three, boosters. [0154]
  • The invention also includes a method of treating a subject at risk for atherosclerosis by (1) providing a subject at risk for atherosclerosis and (2) administering to the subject one or more of the following: (a) an LBP protein or fragment or analog thereof and an adjuvant; (b) a nucleic acid encoding an LBP protein; (c) a virus or bacteria comprising a nucleic acid encoding an LBP protein; and (d) an edible plant comprising a nucleic acid encoding an LBP protein. The LBP protein used in this method can be any LBP described herein, e.g., LBP-1, LBP-2, or LBP-3. A combination of more than one nucleic acid or LBP protein or fragment or analog thereof can be administered to the subject. For example, combinations of LBP proteins, or nucleic acids encoding LBP proteins, include: (1) LBP-I and LBP-2; (2) LBP-I and LBP-3; (3)LBP-2 and LBP-3; and (4) LBP-1, LBP-2, and LBP-3. This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis. [0155]
  • Also provided by the invention is a method of treating a subject at risk for atherosclerosis whereby a non-autologous LBP protein or a nucleic acid encoding a non-autologous LBP protein is delivered to the subject to generate an immune response to an autologous LBP. Specifically, this method entails identifying one or more autologous LBP proteins, e.g., LBP-1, LBP-2, or LBP-3, produced by the subject. The identification can by, e.g., DNA sequence analysis, protein sequence analysis, antibody reactivity, hybridization analysis, or nucleic acid amplification. Next, a non-autologous LBP protein, e.g., allogeneic, xenogeneic, or a genetically modified, non-naturally occurring protein that differs at one or more amino acid residues from the one or more LBP proteins, is administered to the subject. Alternatively, a nucleic acid encoding a non-autologous LBP protein is administered to the subject. The anti-atherosclerotic effectiveness of this immunotherapeutic product is determined by its ability to induce an immune response against one or more autologous LBP proteins when administered to the subject. It is therefore expected that extensive differences between a non-autologous and autologous LBP protein will not result in cross immunoreactivity. This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis. [0156]
  • Another method of the invention is a method of treating a subject at risk for atherosclerosis by increasing the levels of one or more LBP proteins circulating in the plasma. According to this method, either autologous or non-autologous LBP levels may be increased. Non-autologous LBP proteins include, e.g., allogeneic LBP, xenogeneic LBP, and genetically modified LBP. The plasma levels of one or more LBP proteins can be increased by the delivery of a nucleic acid encoding an LBP protein. Because LBP generally does not normally occur as a circulating protein, the endogenous molecule is expected to be susceptible to immune recognition when delivered in a soluble form. This method optionally includes a step of diagnosing the subject as being at risk for atherosclerosis. [0157]
  • Also included in the invention is a pharmaceutical composition containing one or more LBP proteins, e.g., LBP-1, LBP-2, or LBP-3, mixed with an adjuvant, suitable for use in humans. The pharmaceutical composition can contain a combination of more than one LBP protein. For example, compositions can include any of the following: (1) LBP-1 and LBP-2; (2) LBP-I and LBP-3; (3) LBP-2 and LBP-3; and (4) LBP-1, LBP-2, and LBP-3. [0158]
  • Also included in the invention is a cell therapy system, whereby a cell expressing an LBP is delivered to a subject at risk for atherosclerosis. This cell can be engineered to express either an autologous or non-autologous LBP protein or peptide of the invention. Delivery of this engineered cell to a subject results in the in vivo production of an LBP protein and the associated immunotherapy produced when either the protein or a nucleic acid encoding the protein is provided to an individual. Cell therapy methods are described in U.S. Pat. No. 5,955,095 (herein incorporated by reference). [0159]
  • The following non-limiting examples further illustrate the present invention. [0160]
  • EXAMPLES Example 1
  • Construction of a Rabbit cDNA Library [0161]
  • This example illustrates the construction of a rabbit cDNA library using mRNA from balloon-deendothelialized healing rabbit abdominal aorta. Balloon-catheter deendothelialized rabbit aorta has been shown to be a valid model for atherosclerosis (Minick et al., Am. J. Pathol. 95:131-158 (1979). [0162]
  • The RNA was obtained four weeks after ballooning to maximize focal LDL binding in the ballooned rabbit aorta. First strand cDNA synthesis was carried out in a 50 μl reaction mixture containing 4 μg mRNA; 2 μg oligo d(T)primer; methylation dNTP mix (10 mM each); 10 mM DTT; 800 units superscript II RT (Life Technologies, Gaithersburg, Md.); 1×first strand cDNA synthesis buffer (50 mM Tris-HCl, pH 8.3; 75 mM KCl; 5 mM MgCl[0163] 2), which was incubated for I hr at 37° C. The reaction mixture was then adjusted to 250 μl through the addition of 1×second strand buffer (30 mM Tris-HCl, pH 7.5; 105 mMKCl;5.2 mM MgCl2); 0.1 mM DTT; methylation dNTP mix (10 mM each); 50 units E. coli DNA polymerase I, 3 units RNase H; 15 units E. coli DNA ligase (all enzymes from Life Technologies), which was incubated for an additional 2.5 hr at 15° C. The resulting double-stranded cDNAs (dscDNA) were then treated with 1.5 units T4 DNA polymerase (Novagen Inc., Madison, Wis.) for 20 min at 11° C. to make blunt-ended dscDNA. These were then concentrated by ethanol precipitation and EcoRl/Hind III linkers were attached to the ends by T4 DNA ligase (Novagen Inc.). The linker-ligated cDNAs were treated with EcoRl and Hind111 restriction enzymes to produce EcoRl and Hind III recognition sequences at their 5′ and 3′ ends, respectively. After the removal of linker DNA by gel exclusion chromatography, the dscDNAs were inserted into λEXlox phage arms (Novagen Inc.) in a unidirectional manner by T4 DNA ligase and packaged into phage particles according to the manufacturer's protocol (Novagen Inc.). A phage library of cDNAs containing 2×106 independent clones was established from 4 μg of mRNA.
  • Example 2
  • Identification of Rabbit cDNAs Encoding LDL Binding Proteins (LBPs) [0164]
  • This example illustrates a method of functionally screening a rabbit cDNA library so as to identify cDNAs encoding LBPs which bind to both native LDL and methyl LDL. Methyl LDL is not recognized by previously reported cell surface receptors. See, e.g., Weisgraber et al., J. Biol. Chem. 253:9053-9062 (1978). [0165]
  • A fresh overnight culture of [0166] E. coli ER1 647 cells (Novagen Inc.) was infected with the cDNA phage obtained from Example 1, and plated at a density of 2×104 plaque-forming units (pfu) in 150 mm diameter plates containing 2×YT agar. A total of 50 plates, equivalent to 1×106 phage, were plated and incubated at 37° C. until the plaques reached 1 mm in diameter (5-6 hr). A dry nitrocellulose membrane, which had previously been saturated with 10 mM IPTG solution, was layered on top of each plate to induce the production of recombinant protein, as well as to immobilize the proteins on the membranes. The plates were incubated at 37° C. for an additional 3-4 hr, and then overnight at 4° C.
  • The next day, the membranes were lifted from each plate and processed as follows. Several brief rinses in TBST solution (10 mM Tris-HCl, pH 8.0; 150 mM NaCl, 0.05% Tween 20); two 10-min rinses with 6 M guanidine-HCl in HBB (20 mM HEPES, pH 7.5; 5 mM MgCl[0167] 2, 1 mM DTT, and 5 mM KCl); two 5-min rinses in 3 M guanidine-HCl in HBB; a final brief rinse in TBSEN (TBS, 1 mM EDTA, 0.02% NaN3).
  • The membranes were then blocked for 30 min at room temperature in a solution of TBSEN with 5% non-fat dry milk, followed by 10 min in TBSEN with 1% non-fat dry milk. Following blocking, the membranes were incubated with native human LDL (obtained as described in Example 11 or methylated human LDL (meLDL) (see Weisgraber et al., J. Biol. Chem. 253:9053-9062 (1978)), at a concentration of 4 μg/ml, in a solution containing 1×TBSEN, 1% non-fat dry milk, [0168] 1mM PMSF, 0.5×protease inhibitor solution (1 mM ε-amino caproic acid/1 mM benzamidine). Incubation was for 4 hr at room temperature in a glass Petri dish with gentle stirring on a stirring table, followed by overnight at 4° C. with no stirring.
  • Specifically bound meLDL and native LDL were detected on the nitrocellulose membranes by antibodies against human LDL. Sheep anti-human LDL polyclonal antibodies (Boehringer Mannheim, Indianapolis, Ind.) were adsorbed with [0169] E. coli plys E cell extracts to abolish background. For adsorption, E. coli plys E cells were grown to log phase, spun down and resuspended in PBS containing 1 mM PMSF, 2 mM ε-amino caproic acid, and 1 mM benzamidine. The cell suspension then underwent 8 freeze-thaw cycles via immersion in liquid nitrogen and cold running tap water, respectively. The anti LDL antibodies/cell extract solution were incubated with gentle stirring for 1 hr at 4° C. (1 ml of antibody solution/3 mg crude cell extract). Following incubation, the mixture was centrifuged (10,000×g; 10 min; 4° C.) and the supernatant was stored at 4° C. in the presence of 0.02% NaN3, until use. The membranes were processed for immunoscreening as follows: (i) three 5-min washes at room temperature in TBSEN containing 1% gelatin; (ii) 30 min incubation in PBS, pH 7.4 with 1% gelatin; (iii) two-hr room temperature incubation with gentle stirring in fresh PBS/gelatin solution containing adsorbed sheep anti-human LDL antibodies (Boehringer Manheim, Indianapolis, Ind.) (1:1000 dilution); (iv) three brief washes in TBS, pH 7.4; (v) one-hr room temperature incubation with gentle stirring in PBS/gelatin solution containing donkey anti-sheep alkaline phosphatase-conjugated antibodies (Sigma, St. Louis, Mo.) (1:10,000 dilution); (vi) three brief washes with TBS, PH 7.4.; and (vii) development according to the manufacturer's instructions, using an alkaline phosphatase substrate development kit (Novagen Inc.). Phage plaques which produced LBPs appeared as blue-colored “donuts” on the membranes.
  • The phage from Example 1 containing the LBP cDNAs were plaque-purified and converted into plasmid subclones by following a protocol called “Autosubcloning by Cremediated Plasmid Excision” provided by Novagen Inc. DNA sequences were obtained by the dideoxynucleotide chain-termination method (Sanger et al., Proc. Natl. Acad. Sci., USA 74: 5463-5467 (1977), and analyzed by an Applied Biosystems automated sequencer. The open reading frame (ORF) of each cDNA was determined from consensus sequences obtained from both the sense and antisense strands of the cDNAs. Sequencing confirmed that three previously unknown genes had been isolated. Since the genes were selected by functional screening for LDL binding, the proteins coded by these genes were termed LDL binding proteins (LBPs), specifically, LBP-1, LBP-2 and LBP-3. The cDNA sequences for rabbit LBP-1, LBP-2 and LBP-3 and the corresponding proteins are set forth in SEQ ID NOS: 10-14 and 48. [0170]
  • Based on their respective cDNA coding sequences, the sizes of the recombinant proteins were determined to be 16.2 kDa for LBP-1, 40 kDa for LBP-2, and 62.7 kDa for LBP-3. [0171]
  • Example 3
  • Northern Blot Analysis of Rabbit RNA Using LBP cDNA or cRNA [0172]
  • This example illustrates the size and tissue distribution of LBP mRNAs. Total RNA was isolated from different rabbit tissues: adrenals, thoracic aorta, abdominal aorta, ballooned and reendothelialized abdominal aorta, heart, kidney, smooth muscle cells, lung and liver, by Trizol reagent (Life Technologies) and concentrated by ethanol precipitation. Gel electrophoresis of RNA was carried out in 1.2% agarose gel containing 1×MOPS buffer (0.2 M MOPS, pH 7.0; 50 mM sodium acetate; 5 mM EDTA, pH 8.0) and 0.37 M formaldehyde. Gels were loaded with 20 μg total RNA from each tissue examined and electrophoresed at 100 volts for 2 hr in 1×MOPS buffer. RNAs were blotted onto supported nitrocellulose membranes (Schleicher & Schuell, Keene, N.H.) and immobilized by baking at 80° C. for 2 hr. Hybridization to radiolabeled LBP-1, LBP-2 and LBP-3 cDNA or cRNA probes was carried out by standard procedures known to those skilled in the art (see, e.g., Ausubel et al., Current Protocols in Molecular Biology; John Wiley & Sons (1989)); signals were detected by autoradiography. [0173]
  • The results were as follows: the sizes of the mRNAs were about 1.3 kb for LBP-1, about 2.3-2.5 kb for LBP-2, and about 4.7 kb for LBP-3. LBP-1, LBP-2 and LBP-3 mRNA were found in all tissues tested, but the highest amount was in ballooned abdominal aorta. [0174]
  • Example 4
  • Isolation of Human LBP cDNAs and Genomic Clones [0175]
  • This example illustrates isolation of human LBP cDNAs. Human LBP cDNA clones were isolated from three cDNA libraries. A human fetal brain cDNA library was obtained from Stratagene, LaJolla, Calif., a human liver and a human aorta cDNA library were obtained from Clontech, Palo Alto, Calif., and screened with a radiolabeled cDNA probe derived from rabbit LBP-1, LBP-2 or LBP-3, according to the method described in Law et al., Gene Expression 4:77-84 (1994). Several strongly hybridizing clones were identified and plaque-purified. Clones were confirmed to be human LBP-1, LBP-2 and LBP-3, by DNA sequencing using the dideoxynucleotide chain-termination method and analysis by an Applied Biosystems automated sequencer. The cDNA sequences and the corresponding proteins for human LBP-1, LBP-2 and LBP-3 are set forth in SEQ ID NOS: 15, 16 and 17, respectively. [0176]
  • A human genomic library was screened with each of the LBP-1, LBP-2, and LBP-3 clones obtained from the cDNA library screening. Clones hybridizing to each of the three cDNAs were isolated and sequenced. The genomic sequence for LBP-1, LBP-2, and LBP-3 are set forth in FIGS. [0177] 22-24, respectively. The LBP-1 open reading frame spans four exons of the LBP-1 gene (FIG. 22; SEQ ID NO: 49). The LBP-1 protein predicted by the genomic sequence is identical to that predicted by the cDNA clone described above. The LBP-2 open reading frame spans five exons of the LBP-2 gene (FIG. 23; SEQ ID NO: 50). The LBP-2 protein predicted by the genomic sequence differs from that predicted by the cDNA clone in that it contains an additional 321 amino acids at its amino terminus (the LBP-2 cDNA is a 5′ truncation). The LBP-3 open reading frame spans ten exons of the LBP-3 gene (FIG. 24; SEQ ID NO: 51). The LBP-3 protein predicted by the genomic sequence differs from that predicted by the cDNA clone in that it contains an additional 16 amino acids at its amino terminus (the LBP-3 cDNA is a 5′ truncation) and an Asn at amino acid position 130 (the cDNA predicts a Tyr at this position). A comparison between the corresponding LBP-1, LBP-2 and LBP-3 protein sequences for rabbit and human are shown in FIGS. 19, 20 and 21.
  • Example 5
  • Isolation of Recombinant LBP-1 LBP-2 and LBP-3 Rabbit Proteins from E. Coli [0178]
  • LBP cDNA was isolated from the original pEXlox plasmids obtained as described in Examples 1 and 2, and subcloned into the pPRoEX-HT vector (Life Technologies) for recombinant protein expression. Induction of the recombinant protein by IPTG addition to transformed [0179] E. coli DH10B cultures resulted in the expression of recombinant protein containing a 6-histidine tag (N-terminal). This tagged protein was then purified from whole cell proteins by binding to Ni-NTA (nickel nitrilo-triacetic acid) as described in the protocol provided by the manufacturer (Qiagen, Inc., Santa Clara, Calif.). The preparation obtained after the chromatography step was approximately 90% pure; preparative SDS-PAGE was performed as the final purification step.
  • When required by the characterization procedure, iodination of LBPs was carried out using Iodobeads (Pierce, Rockford, Ill.). The Iodobeads were incubated with 500 μCi of Na[0180] 125I solution (17 Ci/mg) (New England Nuclear, Boston, Mass.) in a capped microfuge tube for 5 min at room temperature. The protein solution was added to the Iodobeads-Na125I microfuge tube and incubated for 15 min at room temperature. At the end of this incubation, aliquots were removed for the determination of total soluble and TCA precipitable counts. The radiolabeled protein was then precipitated with cold acetone (2.5 vol; −20° C.; 2.5 hr). Following this incubation, precipitated protein was collected by centrifugation (14,000 g; 1 hr; room temperature) and resuspended in sample buffer (6 M urea/50 mM Tris, pH 8.0/2 mM EDTA). Integrity of the protein preparation was assessed by SDS-PAGE.
  • The identities of the recombinant LBPs were confirmed using standard protein sequencing protocols known to those skilled in the art. (A Practical Guide for Protein and Peptide Purification for Microsequencing, Matsudaira, ed., Academic Press, Inc., 2d edition (1993)). Analysis was performed using an Applied Biosystems Model 477A Protein Sequencer with on-[0181] line Model 120 PTH amino acid analyzer.
  • Example 6
  • Production of Antibodies to LBP-1. LBP-2 and LBP-3 [0182]
  • This example illustrates the production of polyclonal antibodies to LBP-1, LBP-2 and LBP-3. A mixture of purified recombinant LBP protein (0.5 ml; 200 μg) and RIBI adjuvant (RIB1 ImmunoChem. Research, Inc., Hamilton, Mont.) was injected subcutaneously into male guinea pigs (Dunkin Hartley; Hazelton Research Products, Inc., Denver, Pa.) at 3-5 sites along the dorsal thoracic and abdominal regions of the guinea pig. Blood was collected by venipuncture on days 1 (pre-immune bleeding), 28, 49 and 70. Booster injections were administered on days 21 (100 μg; SC), 42 (50 μg; SC), and 63 (25 μg; SC). The titer of the guinea pig antiserum was evaluated by serial dilution “dot blotting.” Preimmune antiserum was evaluated at the same time. After the third booster of LBP protein, the titer against the recombinant protein reached a maximal level with a detectable calorimetric response on a dot blot assay of 156 pg. [0183]
  • Specificity of the polyclonal antibody for recombinant LBP-1, LBP-2 or LBP-3 was demonstrated using Western blot analysis. (Towbin et al., Proc. Natl. Acad. Sci. USA 76: 4350 (1979)). The protein-antibody complex was visualized immunochemically with alkaline phosphatase-conjugated goat antiguinea pig IgG, followed by staining with nitro blue tetrazolium (BioRad Laboratories, Hercules, Calif.). Non-specific binding was blocked using 3% non-fat dry milk in Tris buffered saline (100 mM Tris; 0.9% NaCl, pH 7.4). [0184]
  • Example 7
  • Immunohistochemical Characterization [0185]
  • This example illustrates the presence of LBPs in or on endothelial cells covering plaques, in or on adjacent smooth muscle cells, and in the extracellular matrix. In addition, co-localization of LDL and LBPs was demonstrated. These results were obtained by examining ballooned rabbit arterial lesions and human atherosclerotic plaques by immunohistochemical methods. [0186]
  • Ballooned deendothelialized aorta was obtained from rabbits which had received a bolus injection of human LDL (3 mg; i.v.) 24 hr prior to tissue collection. Human aortas containing atherosclerotic plaques were obtained from routine autopsy specimens. Tissues were fixed in 10% buffered formalin (≦24 hr) and imbedded in paraffin using an automated tissue-imbedding machine. Tissue sections were cut (5-7 μ) and mounted onto glass slides by incubating for 1 hr at 60° C. Sections were deparaffinized. After a final wash with deionized H[0187] 2O, endogenous peroxidase activity was eliminated by incubating the sections with 1% H2O2/H2O buffer for 5 min at room temperature. Sections were rinsed with phosphate buffered saline (PBS) for 5 min at room temperature and nonspecific binding was blocked with 5% normal goat serum or 5% normal rabbit serum depending on the source of the secondary antibody (Sigma, St. Louis, Mo.) (1 hr; room temperature). Sections were then incubated with a 1:50 dilution (in 5% normal goat serum/PBS) of a guinea pig polyclonal antibody against the rabbit form of recombinant LBP-1, LBP-2 or LBP-3. Controls included preimmune serum as well as specific antisera to LBP-1, LBP-2, or LBP-3 in which the primary antibody was completely adsorbed and removed by incubation with recombinant LBP-1, LBP-2 or LBP-3 followed by centrifugation prior to incubation with the tissue sections. An affinity purified rabbit polyclonal antibody against human apolipoprotein B (Polysciences Inc.; Warrington, Pa.) was used at a dilution of 1:100 (in 5% normal rabbit serum/PBS). Sections were incubated for 2 hr at room temperature in a humidified chamber. At the end of incubation, sections were rinsed with PBS and incubated with a 1:200 dilution (in 5% normal goat serum/PBS) of goat anti-guinea pig biotinylated IgG conjugate (Vector Laboratories, Burlingame, Calif.) or a 1:250 dilution (in 5% normal rabbit serum/PBS)of rabbit anti-goat biotinylated IgG conjugate (Vector Laboratories, Burlingame, Calif.) for 1 hr at room temperature in a humidified chamber. Sections were then rinsed with PBS and antigen-antibody signal amplified using avidin/biotin HRP conjugate (Vectastain ABC kit; Vector Laboratories, Burlingame, Calif.). Sections were developed using DAB substrate (4-6 min; room temperature) and counterstained with hematoxylin. In the ballooned rabbit artery, immunohistochemistry with the anti-LBP-1, LBP-2 and LBP-3 antibodies showed that LBP-1, LBP-2 and LBP-3 were located in or on functionally modified endothelial cells at the edges of regenerating endothelial islands, the same location in which irreversible LDL binding has been demonstrated (Chang et al., Arteriosclerosis and Thrombosis 12:1088-1098 (1992)). LBP-1, LBP-2 and LBP-3 were also found in or on intimal smooth muscle cells underneath the functionally modified endothelial cells, and to a lesser extent, in extracellular matrix. No LBP-1, LBP-2 or LBP-3 was detected in still deendothelialized areas, where LDL binding had been shown to be reversible (Chang et al., Arteriosclerosis and Thrombosis 12:1088-1098 (1992)). Immunohistochemistry of ballooned rabbit aorta with anti-human apolipoprotein B antibodies showed the presence of LDL at the same locations as that found for LBP-1, LBP-2 and LBP-3.
  • In the human atherosclerotic plaques taken at routine autopsies, immunohistochemistry with the anti-LBP-1,. anti-LBP-2 and anti-LBP-3 antibodies showed that LBP-1, LBP-2, and LBP-3 were also found in or on endothelial cells covering plaques and in or on adjacent smooth muscle cells. In the human tissue, there was greater evidence of LBP-1, LBP-2 and LBP-3 in extracellular matrix. [0188]
  • The results obtained with paraffin sections were identical to those of frozen sections. [0189]
  • Example 8
  • Affinity Coelectrophoresis (ACE) Assays of LBPs and LDL or HDL [0190]
  • This example illustrates that binding occurs between LBP-1, LBP-2 or LBP-3 and LDL, and that this binding is specific, as illustrated by the fact that binding does not occur between LBP-1, LBP-2 or LBP-3 and HDL (high density lipoprotein). Analysis of the affinity and specificity of recombinant rabbit LBP-1, LBP-2 or LBP-3 binding to LDL was carried out using the principle of affinity electrophoresis (Lee and Lander, Proc. Natl. Acad. Sci. USA 88:2768-2772 (1991)). Melted agarose (1%; 65° C.) was prepared in 50 mM sodium MOPS, pH 7.0; 125 mM sodium acetate, 0.5% CHAPS. A teflon comb consisting of nine parallel bars (45×4×4 mm/3 mm spacing between bars) was placed onto GelBond film (FMC Bioproducts, Rockland, Me.) fitted to a plexiglass casting tray with the long axis of the bars parallel to the long axis of the casting tray. A teflon strip (66×1×1 mm) was placed on edge with the long axis parallel to the short axis of the casting tray, at a distance of 4 mm from the edge of the teflon comb. Melted agarose (>65° C.) was then poured to achieve a height of approximately 4 mm. Removal of the comb and strip resulted in a gel containing nine 45×4×4 mm rectangular wells adjacent to a 66×1 mm slot. LDL or HDL samples were prepared in gel buffer (50 mM sodium MOPS, pH 7.0, 125 mM sodium acetate) at twice the desired concentration. Samples were then mixed with an equal volume of melted agarose (in 50 mM MOPS, pH 7.0; 125 mM sodium acetate; 50° C.), pipetted into the appropriate rectangular wells and allowed to gel. The binding affinity and specificity of LBP-1 and LBP-3 was tested using several concentrations of LDL (540 to 14 nM) and HDL (2840177 nM). A constant amount (0.003 aM -0.016 M) of t[0191] 25I-labeled LBP-1, LBP-2 or LBP-3 (suspended in 50 mM sodium MOPS, pH 7.0; 125 mM sodium acetate; 0.5% bromphenol blue; 6% (wt/vol) sucrose) was loaded into the slot. Gels were electrophoresed at 70 v/2 hr/20° C. At the end of the run, the gels were air dried and retardation profiles were visualized by exposure of X-ray films to the gels overnight at −70° C., with intensifying screens.
  • LDL retarded LBP-1, LBP-2 and LBP-3 migration through the gel in a concentration-dependent, saturable manner, indicating that LBP-1, LBP-2 and LBP-3 binding to LDL was highly specific. This conclusion is supported by the fact that HDL did not retard LBP-1, LBP-2 or LBP-3. A binding curve generated from the affinity coelectrophoresis assay indicated that LBP-1 binds to LDL with a K[0192] d of 25.6 nM, that LBP-2 (rabbit clone 26) binds to LDL with a Kd of 100 MM, and that LBP-3 (80 kDa fragment) binds to LDL with a Kd of 333 nM.
  • In addition to testing affinity and specificity of LBP-1, LBP-2 and LBP-3 binding to LDL, the ability of “cold” (i.e., non-radiolabeled) LBP-1, LBP-2 or LBP-3 to competitively inhibit radiolabeled LBP-1, LBP-2 or LBP-3 binding to LDL, respectively, was tested. Competition studies were carried out using fixed concentrations of cold LDL and radiolabeled LBP-1 and increasing amounts of cold recombinant LBP-1 (6-31 μM). The ACE assay samples and gel were prepared as described herein. Cold LBP-1 inhibited binding of radiolabeled LBP-1 to LDL in a concentration-dependent manner, cold LBP-2 inhibited binding of radiolabeled LBP-2 to LDL in a concentration-dependent manner, and cold LBP-3 inhibited binding of radiolabeled LBP-3 to LDL in a concentration-dependent manner. [0193]
  • Rabbit and human LBP-2 contain a long stretch of acidic amino acids at the amino terminal (rabbit LBP-2 [0194] amino acid residues 338 through 365 and human LBP-2 amino acid residues 329 through 354). The possibility that this segment of LBP-2 was the LDL binding domain was tested by subcloning two rabbit LBP-2 clones which differ from each other by the presence or absence of this acidic region (clone 26 and clone 45, respectively) into expression vectors, by standard methods known to those skilled in the art. ACE assays were then conducted in order to assess the affinity and specificity of the binding of these two clones to LDL. LDL retarded clone 26 derived radiolabeled LBP-2 migration through the gel in a concentration-dependent, saturable, manner while clone 45 derived radiolabeled LBP-2 migration was not retarded.
  • Competition studies using fixed concentrations of cold LDL and clone 26 derived radiolabeled LBP-2 and increasing concentrations of cold recombinant LBP-2/clone 26 and LBP-a/[0195] clone 45 were carried out. Cold clone 26 derived LBP-2 inhibited binding of clone 26 derived radiolabeled LBP-2 to LDL in a concentration-dependent manner. Clone 45 derived LBP-2, on the other hand, did not affect the binding of clone 26 derived radiolabeled LBP-2 to LDL. These results indicate that the long stretch of acidic amino acids contain a binding domain of LBP-2 to LDL.
  • Example 9
  • Affinity Coelectrophoreses (ACE) Assays of LBP-1 or LBP-2 and LDL in the Presence of Inhibitors [0196]
  • This example illustrates that binding between LBP-1 or LBP-2 and LDL is inhibited by polyglutamic acid or BHF-1. The ability of a third compound to inhibit binding between two proteins previously shown to interact was tested by a modification of the ACE assays described in Example 8. The third compound was added to the top or wells together with the radiolabeled protein. If the third compound inhibited binding, the radiolabeled protein would run through the gel. If the third compound did not inhibit binding, migration of the radiolabeled protein was retarded by the protein cast into the gel. [0197]
  • Inhibition of LBP-1/LDL or LBP-2/LDL binding by polyglutamic acid (average MW about 7500, corresponding to about 7 monomers) was shown by casting a constant amount of LDL (148 nM) in all the rectangular lanes. A constant amount (1 μl) of [0198] 125I-labeled LBP-1 or LBP-2 (0.003 nM -0.016 nM) was loaded in the wells at the top of the gel, together with increasing concentrations of polyglutamic acid (obtained from Sigma) (0-0.4 nM). The gel was electrophoresed at 70 volts for 2 hr, dried and placed on X-ray film, with intensifying screens, overnight at −70° C. before the film was developed to determine the retardation profile of LBP-1 and LBP-2. As the concentration of polyglutamic acid increased, retardation of radiolabeled LBP-1 and LBP-2 migration by LDL decreased in a concentration-dependent manner, which showed that polyglutamic acid inhibited binding between LBP-1, LBP-2 and LDL.
  • Inhibition of LBP-1/LDL binding by BHF-1 was shown by casting a constant amount of LDL (148 nM) in all the rectangular lanes. A constant amount of [0199] 125I-labeled LBP-1 (0.003 nM -0.016 nM) was loaded in the wells at the top of the gel, together with increasing concentrations of BHF-1 (0-10 nM), obtained as described in Example 15. The gel was electrophoresed at 70 volts for 2 hr, dried and placed on X-ray film, with intensifying screens, overnight at −70° C. The film was then developed to determine the retardation profile of 125I-LBP-1. As the concentration of BHF-1 increased, retardation of LBP-1 by LDL decreased in a concentration-dependent manner, which demonstrated that BHF-1 inhibited binding between LBP-1 and LDL.
  • Example 10
  • Affinity Coelectrophoreses (ACE) Assays for Identifying Fragments, Analogs and Mimetics of LBPs which Bind to LDL [0200]
  • This example illustrates a method for identifying fragments, analogs or mimetics of LBPs which bind to LDL, and which thus can be used as inhibitors of LDL binding to LBP in the arterial walls, by occupying binding sites on LDL molecules, thereby rendering these sites unavailable for binding to LBP in the arterial wall. [0201]
  • Fragments of LBPs are generated by chemical cleavage or synthesized from the known amino acid sequences. Samples of these fragments are individually added (cold) to radiolabeled LBP as described in Example 8, to assess the inhibitory potency of the various fragments. By iterative application of this procedure on progressively smaller portions of fragments identified as inhibitory, the smallest active polypeptide fragment or fragments are identified. In a similar manner, analogs of the LBPs are tested to identify analogs which can act as inhibitors by binding to LDL. And, similarly, mimetics of LBP (molecules which resemble the conformation and/or charge distributions of the LDL-binding sites on LBP molecules) are tested in a similar fashion to identify molecules exhibiting affinities for the LDL-binding sites on LBP. [0202]
  • The affinities of the inhibitors so identified are at least as strong as the affinity of LDL itself for the LDL-binding sites on LBP. The inhibitors bind at least competitively, and some irreversibly and preferentially as well, to the LDL-binding sites, thereby rendering such sites unavailable for binding to humoral LDL. [0203]
  • Example 11
  • ELISA Assays [0204]
  • This example illustrates the use of ELISA plate assays for the quantification of a test compound's capacity to inhibit the binding of LDL to a specific LBP. [0205]
  • In one example, the ELISA assay was carried out as follows: LDL was diluted in 50 mM Na[0206] 2HCO3, pH 9.6/0.02% NaN3 and added to the wells of a 96-well plate (Immuno Ware 96-Well Reacti-Bind EIA Polystyrene Plates; Pierce (Rockford, Ill.)) to achieve a final concentration ranging from 0.1 to 1 μg/well. The plates were incubated for 6 hr at room temperature. At the end of the incubation period, the wells were washed 3 times with Tris-buffered saline, pH 7.4 (TBS), and blocked overnight with 200 μl of 1% bovine serum albumin (BSA) in TBS/0.02% NaN3 (Sigma; St. Louis Mo.) at room temperature. The wells were then incubated with 200 μl of LBP protein (5-10 μg/well) in TBS and varying concentrations of the test compound. Plates were incubated for 1 hr at room temperature. The wells were then washed three times with TBS and blocked for 2 hr with 200 μl of 1% BSA in TBS/0.02% NaN3 at room temperature. At the end of the incubation period, the wells were washed 3 times with TBS and a 1:1000 dilution (in TBS/0.05% Tween 20) of the appropriate guinea pig anti-LBP protein polyclonal antibody was added to the wells and incubated for 1 hr at room temperature. The wells were then washed 3 times with TBS/0.05% Tween 20; a 1:30,000 dilution of goat anti-guinea pig IgG alkaline phophatase conjugate (Sigma) was added to each well. Plates were incubated for 1 hr at room temperature. The wells were washed 3 times with TBS/0.05% Tween 20 and a calorimetric reaction was carried out by adding 200 ml of p-nitrophenyl phosphate substrate (Sigma; St. Louis Mo.) to the wells. The reaction was allowed to proceed for 30 min at room temperature and stopped with 50 μl of 3 N NaOH. The absorbance was determined at 405 nm using an ELISA plate reader. The test compound's effectiveness in blocking the binding of LDL to the recombinant protein was assessed by comparing the absorbance values of control and treated groups.
  • In a second example, the ELISA assay was carried out as follows: LDL was diluted in Tris-buffered saline, pH 7.4 (TBS) and added to the wells of a 96-well plate (Immuno Ware 96-Well Reacti-Bind EIA Polystyrene Plates; Pierce (Rockford, Ill.)) to give a plate-saturating concentration of 0.2 μg/well. The plate was incubated for 1 hr at room temperature, after which the wells were washed three times with TBS, before being blocked for 1 hr at room temperature with 1% bovine serum albumin (BSA in TBS). The wells were then washed twice with TBS before LBP-1 or LBP-2 (0.025 μg/well), or LBP-3 (0.01 μg/well) were added, without and with varying concentrations of the test inhibitor compound. Each condition was set up in quadruplicate. The plate was incubated for 1 hr at room temperature, then washed three times with TBS/0.02% Tween (TBS/Tween). An appropriate dilution of guinea pig anti-LBP polyclonal antibody (1:750 to 1:1500, depending on the antibody) was added to three wells for each condition and incubated for 1 hr. Anti-LBP antibody was replaced by buffer for the fourth well of each condition, as a negative control. After 1 hr, the plate was again washed three times with TBS/Tween before a 1:10,000 dilution (in TBS/Tween) of goat anti-guinea pig IgG alkaline phosphatase-conjugated antibody (Sigma) was added to each well. The plate was incubated for 1 hr at room temperature, then washed three times with TBS/Tween. A fresh solution of substrate was prepared from an Alkaline Phosphatase Substrate Kit (Bio-Rad, Hercules, Calif.) as follows: [0207] Mix 1 ml 5×concentrated diethanolamine buffer with 4 ml distilled water. Add one tablet of p-nitrophenylphosphate (5 mg) and vortex until tablet is completely dissolved. Subtrate solution was added to wells immediately. Increasing concentrations of diluted alkaline phosphatase-conjugated goat anti-guinea pig IgG (1:100,000 dilution in TBS/Tween) were added to five empty wells, followed by substrate, as a positive control. Following addition of substrate, the plate was immediately placed in an ELISA plate reader, allowed to stand at 37° C., generally for 75 min, before absorbance was measured at 405 nm. Incubation in the ELISA reader at 37° C. was sometimes adjusted to optimize absorbance (60-90 min). The effectiveness of the test inhibitor was determined, after subtracting absorbance of negative controls, by comparing absorbance in wells where an LBP was mixed with test inhibitor to absorbance in wells containing LBP with no inhibitor.
  • Alternatively, LBPs, rather than LDL, were bound to the plate. Recombinant LBP protein binding to LDL and the effect of varying concentration of the inhibitor on LBP-LDL binding was determined through the use of antibodies against LDL. This interaction was visualized through the use of a secondary antibody conjugated to a reporter enzyme (e.g. alkaline phosphatase). [0208]
  • ELISA plate assays were used to screen agents which can affect the binding of LBP proteins to LDL. For example, peptides derived from LBP-1 and human LBP-3 protein sequences (BHF-1 and BHF-2, respectively) were synthesized and have been shown to reduce the binding of LDL to recombinant LBP-1 and LBP-2 in this format. These results were in agreement with those obtained with the ACE assays. [0209]
  • Example 12
  • Administration of Humanized Antibodies Against LBPs so as to Block LDL-Binding Sites on the LBPs [0210]
  • This example illustrates administration to patients of humanized antibodies against LBP-1, LBP-2 or LBP-3 so as to block LDL-binding sites on arterial LBP molecules. Mouse monoclonal antibodies are humanized by recombinant DNA techniques and produced by standard procedures known to those skilled in the art (Berkower, I., Curr. Opin. Biotechnol. 7:622-628 (1996); Ramharayan and Skaletsky, Am. Biotechnol. Lab 13: 26-28 (1995)) against LBPs and/or the LDL-binding sites on the LBPs. The corresponding Fab fragments are also produced, as described in Goding, J. W., Monoclonal Antibodies: Principles and Practice, Academic Press, New York, N.Y. (1986). These antibodies are administered parenterally in sufficient quantity so as to block LDL-binding sites on the LBP molecules, i.e., 1-10 mg/kg daily. This prevents the irreversible arterial uptake of LDL that is required to facilitate oxidation of the LDL. [0211]
  • Example 13
  • Preparation of LDL [0212]
  • This example illustrates the preparation of LDL. LDL was prepared from the plasma of normolipemic donors (Chang et al., Arterioscler. Thromb. 12:1088-1098 (1992)). 100 ml of whole blood was placed into tubes containing 100 mM disodium EDTA. Plasma was separated from red blood cells by low-speed centrifugation (2,000 g; 30 min; 4° C.). Plasma density was adjusted to 1.025 gm/ml with a solution of KBr and centrifuged for 18-20 hr, 100,000×g, 12° C. Very low density lipoproteins (VLDL) were removed from the tops of the centrifuge tubes with a Pasteur pipette. The density of the infranate was raised to 1.050 gm/ml with KBr solution and centrifuged for 22-24 hr, 100,000×g, 12° C. LDL was removed from the tops of the centrifuge tubes with a drawn out Pasteur pipette tip. Purity of the LDL preparation was checked by Ouchterlony double immunodiffusion against antibodies to human LDL, human HDL, human immunoglobulins, and human albumin. KBr was removed from the LDL solution by dialysis (1L,×2, approximately 16 hr) against 0.9% saline, pH 9.0, containing 1 mM EDTA and 10 μM butylated hydroxytoluene (BHT), the latter to prevent oxidation of LDL. Following dialysis, LDL protein was measured by the method of Lowry (Lowry et al., J. Biol. Chem. 193:265-275 (1951)), and the LDL was stored at 4° C. until use. LDL preparations were kept for no more than 4-6 weeks. [0213]
  • Example 14
  • Preparation of HDL [0214]
  • This example illustrates the preparation of HDL. HDL was prepared from plasma of normolipemic donors. 100 ml of whole blood was placed into tubes containing 100 mM disodium EDTA and plasma was collected by centrifugation (2000 g; 30 min; 4° C.). Apolipoprotein B containing lipoproteins present in plasma were then precipitated by the sequential addition of sodium heparin (5,000 units/ml) and MnCl[0215] 2 (1 M) to achieve a final concentration of 200 units/ml and 0.46 M, respectively (Warnick and Albers, J. Lipid Res. 19:65-76 (1978)). Samples were then centrifuged, (2000 g; 1 hr; 4° C.). The supernatant was collected and density adjusted to 1.21 g/ml by the slow addition of solid KBr. HDL was separated by ultracentrifugation (100,000 g; >46 hr; 12° C.). Purity of the HDL preparation was assessed via Ouchterlony double immunodiffusion test using antibodies against human HDL, human LDL, human immunoglobulins, and human albumin. HDL samples were dialyzed against saline pH 9.0/1 mM EDTA/10 μM BHT (4L; 24 hr/4° C.) and total protein was determined by the Lowry protein assay (Lowry et al., J. Biol. Chem. 193:265-275 (1951)). HDL was stored at 4° C. until use. HDL preparations were kept for no longer than 2 weeks.
  • Example 15
  • Synthesis of BHF-1 [0216]
  • This example illustrates the synthesis of BHF-1, a fragment of human or rabbit LBP-1 which contains amino acid residues 14 through 33. BHF-1 was synthesized using an Applied Biosystems Model 430A peptide synthesizer with standard T-Boc NMP chemistry cycles. The sequence of BHF-1 is as follows: [0217]
  • val-asp-val-asp-glu-tyr-asp-glu-asn-lys-phe-val-asp-glu-glu-asp-gly-gly-asp-gly (SEQ ID NO: 9). [0218]
  • After synthesis, the peptide was cleaved with hydrofluoric acid/anisole (10/1 v/v) for 30 min at −10° C. and then incubated for 30 min at 0° C. BHF-1 was then precipitated and washed three times with cold diethyl ether. Amino acid coupling was monitored with the ninhydrin test (>99%). [0219]
  • The BHF-1 peptide was purified to homogeneity by high performance liquid chromatography on a reverse phase Vydac C[0220] 4 column (2.24×25 cm) using a linear gradient separation (2-98%B in 60 min) with a flow rate of 9 ml/min. Buffer A consisted of 0.1% trifluoroacetic acid (TFA)/Milli Q water and Buffer B consisted of 0.085% TFA/80% acetonitrile. The gradient was run at room temperature and absorbance monitored at 210 and 277 nm.
  • Fast atom bombardment-mass spectrometry gave a protonated molecular ion peak (M+H)[0221] + at m/z=2290.2, in good agreement with the calculated value. On amino acid analysis, experimental values for the relative abundance of each amino acid in the peptide were in good agreement with theoretical values. The lyophilized peptide was stored at −20° C.
  • Example 16
  • In Vitro Screening for Agents Which Inhibit Binding Between LDL and LBPs [0222]
  • This example illustrates in vitro screening for agents which inhibit binding between LDL and LBPs. [0223]
  • A candidate polypeptide for being an agent is chosen, e.g., LBP-1, LBP-2. LBP-3, BHF-1 or any other polypeptide. The shortest fragment of the polypeptide that inhibits LDL binding to LBPs in vitro is determined. Peptides are synthesized by standard techniques described herein. Inhibition assays are performed using standard ELISA techniques for screening, and affinity coelectrophoresis (ACE) assays to confirm the ELISA results, as described herein. Additional assays that can be used in this screening method include, e.g., fluorescence polarization and pulsed ultra-filtration electrospray mass spectrometry. Short peptides ranging, e.g., from dimers to 20-mers are constructed across sequences of the candidate polypeptide whose chemical characteristics make them likely LDL binding sites, e.g., acidic regions. The ability of shorter and shorter lengths of the peptides to inhibit LDL binding in vitro and to mammalian cells in culture is tested. For example, the effect of the peptide on inhibiting LDL binding in mammalian cells transfected to express an LBP gene is tested. Each of the peptides so identified as an inhibitor is tested with each of LBP-1, LBP-2 and LBP-3, to determine whether a single inhibitor works against all three LBPs. [0224]
  • Once the minimum active sequence is determined, the peptide backbone is modified so as to inhibit proteolysis, as discussed herein. For example, modification is accomplished by substitution of a sulfoxide for the carbonyl, by reversing the peptide bond, by substituting a methylene for the carbonyl group, or other similar standard methodology. See Spatola, A.F., “Peptide Backbone Modifications: A Structure-Activity Analysis of Peptides Containing Amide Bond Surrogates, Conformational Constraints, and Related Backbone Replacements,” in Chemistry and Biochemistry of Amino Acids, Peptides and Proteins, Vol. 7, pp. 267-357, B. Weinstein (ed.), Marcel Dekker, Inc., New York (1983). The ability of these analogs to inhibit LDL binding to the LBPs in vitro is tested in a similar manner as for the natural peptides described above, e.g., by ELISA, ACE, fluorescence polarization, and/or pulsed ultra-filtration electrospray mass spectrometry. [0225]
  • Example 17
  • In Vitro Screening With Cultured Mammalian Cells for Agents Which Inhibit Binding between LDL and LBPs [0226]
  • This example illustrates cell-based in vitro screening of agents which have been shown by in vitro tests such as ACE assay and ELISA to be potential inhibitors of binding between LDL and LBPs. [0227]
  • Mammalian cells, such as 293 cells, which are commonly used for expression of recombinant gene constructs, are used to develop cell lines which express LBPs on the cell surface. This is done by subcloning LBP open reading frames (ORFS) into a mammalian expression plasmid vector, pDisplay (Invitrogen, Carlsbad, Calif.), which is designed to express the gene of interest on the cell surface. The use of mammalian cells to produce LBPs allows for their expression in a functionally active, native conformation. Therefore, stably transfected mammalian cell lines with surface expression of LBPs individually, or in combination, are particularly suitable for assaying and screening inhibitors that block LDL binding in cell culture, as well as to evaluate the cytotoxicity of these compounds. [0228]
  • Specifically, LBP ORFs are amplified by PCR (Perkin Elmer, Foster City, Calif.) from cDNA templates using Taq polymerase (Perkin Elmer) and appropriate primers. The amplified LBP ORFs are purified by agarose gel electrophoresis and extracted from gel slices with the Bio-Rad DNA Purification kit (Bio-Rad, Hercules, Calif.). The purified DNAs are then cut with the restriction enzymes Bgl II and Sal I (New England Biolabs, Beverly, Mass.) to generate cohesive ends, and purified again by agarose gel electrophoresis and DNA extraction as described above. The LBP ORFs are then subcloned into the Bgl II/Sal I sites in the mammalian expression vector, pDisplay (Invitrogen) by ligation. Recombinant plasmids are established by transformation in [0229] E. coli strains TOP10 (Invitrogen) or DH5 α (Life Technologies, Grand Island, N.Y.). Recombinant pDisplay/LBP plasmid DNA is isolated from overnight E. coli cultures with the Bio-Rad Plasmid Miniprep kit, cut with Bgl II/Sal I, and analyzed by agarose gel electrophoresis. LBP ORFs in successfully transformed clones are verified by automated dideoxy DNA sequencing. To transfect human kidney 293 cells, 1-2 μg of DNA is mixed with 6 μl lipofectamine reagent (Life Technologies) and incubated with the cells as described in the Life Technologies protocol. LBP expression in transfected cells is confirmed by Western blot analysis of cell extracts obtained 48 hr after transfection. To select for stably transfected 293 cells, the antibiotic G418 (Life Technologies) is added to the growth medium at a concentration of 800 μg/ml. Colonies resistant to G418 are tested for recombinant LBP expression by Western blot, and recombinant clones expressing LBPs are expanded, assayed for LDL binding and used to test compounds for their ability to inhibit LDL binding.
  • Example 18
  • In Vivo Screening for Agents Which Inhibit Binding Between LDL and LBPs [0230]
  • This example illustrates in vivo screening of agents which have been shown by in vitro tests to be promising candidate inhibitors of binding between LDL and LBPs. [0231]
  • In vivo inhibitory activity is first tested in the healing balloon-catheter deendothelialized rabbit aorta model of arterial injury (Roberts et al., J. Lipid Res. 24:1160-1167 (1983); Chang et al., Arterioscler. Thromb. 12:1088-1098 (1992)). This model was shown to be an excellent analog for human atherosclerotic lesions. Other useful animal models for human atherosclerosis include Apo E knockout mice and LDL receptor knockout mice. Both of these mouse models are characterized by high levels of plasma cholesterol and the development of naturally-occurring atherosclerotic-like lesions. [0232]
  • Each candidate inhibitor is tested in five to ten ballooned rabbits, while an equal number of rabbits receive a control peptide, or placebo. Four weeks following aortic deendothelialization, when reendothelialization (healing) is partially complete, daily parenteral (intravenous or subcutaneous) or intragastric administration of the peptides and the analogs begins at an initial concentration of 10 mg/kg body weight, which is varied down, or up to 100 mg/kg depending on results. 30 min later, a bolus of intravenously injected 125I (or [0233] 99mTc-) labeled LDL is given to test the candidate inhibitor's ability in short term studies to inhibit LDL sequestration in healing arterial lesions. If 125I-LDL is used, the animals are sacrificed 8-24 hr later, the aortas excised, washed and subjected to quantitative autoradiography of excised aortas, as previously described (Roberts et al., J. Lipid Res. 24:1160-1167 (1983); Chang et al., Arterioscler. Thromb. 12:1088-1098 (1992)). If 99mTc-LDL is used, analysis is by external gamma camera imaging of the live anesthetized animal at 2-24 hr, as previously described (Lees and Lees, Syndromes of Atherosclerosis, in Fuster, ed., Futura Publishing Co., Armonk, N.Y., pp. 385-401 (1996)), followed by sacrifice, excision and imaging of the excised aorta. Immediately before the end of testing, the animals have standard toxicity tests, including CBC, liver enzymes, and urinalysis.
  • The compounds which are most effective and least toxic are then tested in short term studies of rabbits fed a 2% cholesterol diet (Schwenke and Carew, Arteriosclerosis 9:895-907 (1989)). Each candidate inhibitor is tested in five to ten rabbits, while an equal number of rabbits receive a control peptide, or placebo. Animals receive one or more doses per day of the candidate inhibitor, or placebo, for up to two weeks. Daily frequency of doses is determined by route of administration. If active drug or placebo are administered parenterally, they are given 1-3 times daily and the 2% cholesterol diet is continued. If drug or placebo are given orally, they are mixed with the 2% cholesterol diet. Schwenke and Carew (Arteriosclerosis 9:895-907 (1989)) have shown that the LDL concentration in lesion-prone areas of the rabbit aorta is increased 22-fold above normal in rabbits fed a 2% cholesterol diet for 16 days, and that the increased LDL content precedes the histological evidence of early atherosclerosis. Therefore, analysis of the effect of the candidate inhibitors is tested two weeks after the start of cholesterol feeding by injecting [0234] 125I-LDL, allowing it to circulate for 8-24 hr, and then performing quantitative autoradiography on the excised aortas of both test and control animals. If appropriate, quantitation of aortic cholesterol content is also carried out (Schwenke and Carew, Arteriosclerosis 9:895-907 (1989); Schwenke and Carew, Arteriosclerosis 9:908-918 (1989).
  • The above procedures identify the most promising candidate inhibitors, as well as the best route and frequency of their administration. Inhibitors so identified are then tested in long-term studies of cholesterol-fed rabbits. These tests are carried out in the same way as the short-term cholesterol feeding studies, except that inhibitor effectiveness is tested by injection of [0235] 125I-LDL at longer intervals following the initiation of cholesterol feeding, and lesion-prone areas of the aorta are examined histologically for evidence of atherosclerosis. Testing times are at two, four, and six months. Major arteries are examined grossly and histologically or evidence and extent of atherosclerosis. If necessary, other accepted animal models, such as atherosclerosis-susceptible primates (Williams et al., Arterioscler. Thromb. Vast. Biol. 15:827-836 (1995)), genetically altered mice, and/or Watanabe rabbits are tested with short- and long-term cholesterol feeding.
  • Example 19
  • In Vivo Inhibition of Radiolabeled LDL Accumulation in the Ballooned Deendothelialized Rabbit Aorta via Induction of Active Immunity Against LBP Protein [0236]
  • This example illustrates the effect that induction of immunity against LBP protein has on the accumulation of radiolabeled LDL in the ballooned deendothelialized rabbit aorta model of atherosclerosis. [0237]
  • Immunity was induced in male New Zealand White rabbits (Hazelton Research Products, Denver, Pa.) as follows: A mixture of purified human recombinant LBP-2 or BHF-1 peptide (1 ml; 1 mg) and RIBI adjuvant (RIBI ImmunoChem Research, Inc., Hamilton, Mont.) was injected subcutaneously at 2-5 sites along the dorsal thoracic and abdominal regions of the rabbits. Blood was collected by venipuncture on days 1 (preimmune bleeding), 35, 63, and 91. Booster injections were administered on days 28 (500 μg; SC), 56 (250 μg; SC), and 84 (125 μg; SC). [0238]
  • The titer of the rabbits was evaluated by serial dilution using an ELISA plate format. Preimmune serum was evaluated at the same time. After the third booster of LBP protein or peptide, the titer reached a maximal level with a detectable calorimetric response on an ELISA plate of 156 pg. Titer is defined as the maximum dilution of antibody which generates an absorbance reading of 0.5 above control in 30 min. Specificity of the polyclonal antibodies was demonstrated using Western blot analysis as described in Example 6. [0239]
  • On day 93, the abdominal aorta of immunized and control rabbits was deendothelialized using a Fogarty number 4 embolectomy catheter (Chang et al., Arteriosclerosis and Thrombosis 12:1088-1098 (1992)). Four weeks after ballooning, rabbits received a bolus injection of [0240] 125I-labeled LDL (1 ml; i.v.). Blood samples were collected at 1 hr intervals for 8 hr, and 24 hr post injection. Blood samples were centrifuged for 30 min at 2000 rpm (40° C.)and total activity present in the serum was determined using a Gamma counter. Total TCA precipitable counts were determined by addition of TCA to the serum to a final concentration of 10% followed by incubation for min at 4° C. Serum samples were then centrifuged (2000 rpm; 30 min; 40° C.) and total activity present in the supernate was determined. TCA precipitable counts were calculated by substration: total soluble counts minus counts present in the supernate after TCA precipitation. Blood samples for the determination of antibody titers were collected prior to the injection of the radiolabeled LDL.
  • After 24 hr, the rabbits were injected intravenously with 5% Evan's blue dye which was allowed to circulate for 15 min. Areas of the aorta in which the endothelial covering is absent stain blue while those areas covered by endothelium remain unstained. At the end of the incubation period, the rabbits were euthanized and the abdominal and thoracic aorta were dissected out, rinsed, and fixed overnight in 10% TCA at room temperature. The aortas were then rinsed exhaustively with physiological saline, weighed, counted, blotted dry and placed onto X-ray film in order to visualize the pattern of radiolabeled LDL accumulation in the deendothelialized rabbit abdominal aorta. [0241]
  • Immunization of rabbits against recombinant human LBP-2 or BHF-1 peptide altered the pattern of radiolabeled LDL accumulation in the ballooned deendothelialized abdominal aorta. When corrected for dosage, and percent reendothelialization, immunized-ballooned rabbits had lower accumulation of radiolabeled LDL compared to nonimmune-ballooned rabbits. These results indicate that active immunization against LBP provides an effective means by which the accumulation of LDL in the injured arterial wall can be modified. [0242]
  • Example 20
  • Screening Agents in Humans Which Inhibit Binding Between LDL and LBPs [0243]
  • Human studies are carried out according to standard FDA protocols for testing of new drugs for safety (Phase I), efficacy (Phase II), and efficacy compared to other treatments (Phase III). Subjects, who are enrolled into studies after giving informed consent, are between the ages of 18 and 70. Women who are pregnant, or likely to become pregnant, or subjects with diseases other than primary atherosclerosis, such as cancer, liver disease, or diabetes, are excluded. Subjects selected for study in FDA Phase II and Phase III trials have atherosclerotic disease previously documented by standard techniques, such as ultrasound and/or angiography, or are known to be at high risk of atherosclerosis by virtue of having at least one first degree relative with documented atherosclerosis. Subjects themselves have normal or abnormal plasma lipids. Initial testing includes 20-50 subjects on active drug and 20-50 subjects, matched for age, sex, and atherosclerotic status, on placebo. The number of subjects is pre-determined by the number needed for statistical significance. Endpoints for inhibitor efficacy includes ultrasound measurements of carotid artery thickness in high risk subjects, as well as in subjects with known carotid or coronary disease; atherosclerotic events; atherosclerotic deaths; and all-cause deaths in all subjects. Non-invasive analysis (carotid artery thickness by ultrasound) as per Stadler (Med. and Biol. 22:25-34 (1996)) are carried out at 6- to 12-month intervals for 3 years. Atherosclerotic events and deaths, as well as all-cause deaths are tabulated at 3 years. [0244]
  • Oral dosage of drug in FDA Phase I trials ranges from 0.01 to 10 gm/day, and is determined by results of animal studies, extrapolated on a per kg basis. Based on data obtained from Phase I studies, the dose range and frequency are narrowed in Phase II and III trials. If parenteral administration of drug is determined by animal studies to be the only effective method, parenteral administration in human subjects is tested by injection, as well as by the transdermal and nasal insufflation routes. Testing of parenteral drug follows the same outline as that for oral administration. [0245]
  • The optimal treatment schedule and dosage for humans is thus established. [0246]
  • Example 21
  • Treating an Individual Having Atherosclerosis with BHF-1 [0247]
  • This example illustrates a method for treating an individual having atherosclerosis with an LBP fragment, e.g., BHF-1, so as to decrease the levels of arterially bound LDL in the individual. BHF-1 is obtained as described herein. The BHF-1 is administered to the mammal intravenously as a bolus or as an injection at a concentration of 0.5-10 mg/kg body weight. Such administrations are repeated indefinitely in order to prevent the development or progression of symptomatic atherosclerosis, just as is done currently with cholesterol lowering drugs. Stable subjects are examined twice yearly to evaluate the extent of any atherosclerotic disease by physical exam and non-invasive studies, such as carotid artery thickness, ultrasound, and/or gamma camera imaging of the major arteries, to determine if atherosclerotic lesions are present, and, if previously present, have regressed or progressed. Such a regimen results in treatment of the atherosclerosis. [0248]
  • Example 22
  • In Vivo Reduction of Atherosclerosis in Apo E Knockout Mice by Immunization with LBPs [0249]
  • Separate immunization experiments were performed with each of LBP-1, LBP-2, and LBP-3. Immunity was induced by injecting apo E knockout mice with the LBP protein (LBP-1, LBP-2, or LBP-3) together with an RIBI adjuvant (RIBI ImmunoChem Research, Inc., Hamilton, Mont.). Apo E knockout mice (Jackson Laboratories, Bar Harbor, Me.) are hyperlipidemic and thus a model for human atherosclerosis. Apo E knockout mice have high levels of plasma cholesterol and develop naturally-occurring atherosclerotic-like lesions. [0250]
  • Four week old apo E knockout mice (Jackson Laboratories, Bar Harbor, Maine) were ear tagged, randomly assigned to different cages and weighed. Body weights were determined weekly. Animals were allowed to habituate for 1 week. Normal rodent chow was provided ad libitum and animals were maintained in a 12:12 light:dark cycle. The following four groups of mice were treated with either recombinant LBP proteins (40 μg of recombinant protein/mouse) plus RIBI adjuvant or RIBI adjuvant alone (control group). [0251]
  • LBP-1: Immunized with rabbit recombinant LBP-1 (6-His tag). [0252]
  • LBP-2: Immunized with rabbit recombinant LBP-2 clone 26 (6-His tag). [0253]
  • LBP-3: Immunized with rabbit recombinant LBP-3 (6-His tag). [0254]
  • Control: Received adjuvant. [0255]
  • Blood samples (pre-immune serum) were collected prior to the initial injection of recombinant protein and RIBI adjuvant (as described in the manufacturer's manual). After 21 days, mice received a booster injection (half-initial dose) and were then bled seven days later. Titer was defined as the maximum dilution of serum that yielded a change in absorbance equivalent to 2× that of control serum (60 min; 37° C.). The amount of recombinant protein per well was 100 ng. [0256]
  • Booster injections took place at 21 day intervals until an average titer value of 1:10,000 was reached. At this time, mice were switched to western type diet (Harland Teklad, Madison, Wis.) and fed ad libitum. Blood samples were collected at this time (retro-orbital sinus bleeding technique) and monthly thereafter. [0257]
  • Blood samples were analyzed for total cholesterol, HDL cholesterol, and triglyceride concentration with a commercially available total cholesterol and triglycerides assay kits (Sigma; St. Louis Mo.) using an ELISA format. HDL concentration was determined after Apo B containing lipoproteins were precipitated using heparin/MnCl[0258] 2.
  • Apo E knockout mice were sacrificed at 26 weeks of age. The mice were anesthetized with methoxyfluorane and exanguinated via cardiac puncture. A midline thoracotomy was performed, a cannula inserted into the right ventricle and perfusate allowed unrestricted flow via an incision into the right atrium. The mice were perfused with saline, followed by 10% phosphate buffered formalin until fasciculations stopped. At this time, the aorta was exposed and adventitial fat removed in situ. The aorta was then removed from the heart down to the iliac bifurcation and placed in 10% phosphate buffered formalin overnight. [0259]
  • The aorta was stained as follows: after a brief 70% ethanol rinse, it was immersed in a filtered solution of 0.5% (weight/volume) Sudan IV in 35% ethanol/50% acetone with continuous shaking for 10 minutes at room temperature. Unbound dye was removed by incubating the aorta in an 80% ethanol solution with shaking until the background color was clear. The vessel was then rinsed in distilled water, placed in physiological saline and opened longitudinally from the aortic arch down to the iliac bifurcation. The vessel was pinned out and photographed. Photographs were then digitized using an Astra 1200S scanner (UMAX Technologies Inc., Freemont, Calif.) and a commercially available graphics program (Canvas; Deneba Software, Miami Fla.). Total and lesion areas were determined using the signal processing toolbox of MATLAB (The Mathworks Inc., Natick, Mass.). Percent involvement was calculated by dividing lesion area by total area. [0260]
  • A second analysis was done to measure aortic atherosclerosis by a cholesterol extraction method whereby cholesterol is determined as a unit weight of artery. This method may be more accurate in measuring lesion size than attempting to measure the thickness of many sections. Specifically, the weight of an artery was measured, then the cholesterol was extracted. Aortic cholesterol content was then measured by gas-liquid chromatography. The amount of cholesterol per unit weight of artery was then determined. [0261]
  • After the first booster injection some of the apo E knockout mice immunized against LBP-1 had relatively high anti-LBP-1 titers (≦1:5000) while others in the same group exhibited moderate levels (>1:500 to <1:1000). LBP-2/26 titers were low in the apoE knockout mice (<1:500) at this time. LBP-3 titers ranged from moderate to low (≧1:500 to <1:1000) to low (<1:500) in the apoE knockout mice. [0262]
  • After the second booster injection, Apo E knockout mice immunized against LBP-1 had moderate to high titers (>1:1000 to ≦1:8000). Apo E knockout mice immunized against LBP-2/26 had moderate titer levels (>1:2000). LBP-3 titers range from moderate to high (>1:1000 to >1:8000) in the Apo knockout mice. [0263]
  • After the third booster injection, most of the mice immunized against LBP-1 had relatively high titers (>1:10,000) while others had moderate to high titers (>1000 to <1:10,000). Some of the Apo E knockout mice had moderate (<1:5000) to low (<1:1000) titers. LBP-3 titers ranged from high (>1:5000 to ≦1:10,000) to moderate (>1:1000 to <1:5000). [0264]
  • Data were analyzed using T-tests and Wilcoxons. Immunization against LBP-1, LBP-2/26 or LBP-3 did not have a significant effect (P>0.05) on body weight of Apo E knockout mice. Due to the small sample size and the large variability present in the Apo E knockout mice, it was not possible to determine whether immunization against LBP-1, LBP-2/26 or LBP-3 had an effect on total cholesterol, HDL cholesterol or triglycerides concentration, but it did not appear to. [0265]
  • Immunization against LBP-1 or LBP-3 did not have a significant effect (P>0.05) on lesions of the apo E knockout mice or LDL receptor negative knockout mice. However, immunization of the apo E knockout mice against LBP-2 had a significant effect on lesion area (Table 2), and, once outliers were deleted, a significant effect on arterial wall cholesterol content (Table 3). The LBP-2 immunized apo E knockout mice had significantly reduced aortic atherosclerosis as compared to the control, non-immunized mice. Without being bound to any particular theory, the circulating antibodies generated against LBP-2 proteins are thought to block LDL binding to the artery wall. [0266]
    TABLE 2
    Lesion Area in LBP-Immunized Apo E Mice
    Lesion Area Treated Area P-Value
    Apo E Mice % Coverage Change Wilcoxon
    Control 9.40
    LBP-1 6.05 −0.36% 0.07
    LBP-2 6.01 −0.36% 0.01
    LBP-3 7.14 −0.24% 0.36
  • [0267]
    TABLE 3
    Arterial Cholesterol Content in LBP-Immunized Apo E Mice
    Arterial Wall Treated Area
    Cholesterol Cholesterol P-Value
    Apo E Mice cholesterol/mg aorta) Change Wilcoxon
    Control 6.33
    LBP-1 3.82 −0.40% 0.14
    LBP-2 3.28 −0.48% 0.07
    LBP-2 (outliers 1.83 −0.71% 0.01
    deleted)
    LBP-3 4.48 −0.29% 0.20
  • Example 23
  • In Vivo Reduction of Atherosclerosis in LDL Receptor Knockout Mice by Immunization with BHF-1 [0268]
  • An immunization experiment was performed with the BHF-1 peptide. LDL receptor (LDLR) knockout mice (B6,129S-Ldlr[0269] tmlHer, Jackson Laboratories, Bar Harbor, Me.) were injected with the BHF-1 peptide (see Example 15 for methods of synthesizing the BHF-1 peptide) together with an RIBI adjuvant (RIBI ImmunoChem Research, Inc., Hamilton, Mont.). LDLR knockout mice are hyperlipidemic and thus a model for human atherosclerosis. LDLR knockout mice have high levels of plasma cholesterol and develop naturally-occurring atherosclerotic-like lesions.
  • Four week old LDLR knockout mice were ear tagged, randomly assigned to different cages and weighed. Body weights were subsequently determined weekly. Animals were allowed to habituate for one week prior to experimentation. Normal rodent chow was provided ad libitum and animals were maintained in a 12:12 light:dark cycle. Animals were divided into experimental and control groups, as follows: (1) experimental, 16 mice were immunized with the BHF-1.20.L peptide; (2) control, 8 mice were immunized against bovine serum albumin. [0270]
  • Mice in the experimental group received subcutaneous injections (9.99 μg/g body weight; 200 μl final volume) of the BHF-1.20.L peptide daily for 2 weeks, from 5 to 7 weeks of age, prior to the initial injection with the peptide and adjuvant. Blood samples (pre-immune serum) were collected prior to the initial injection of BHF-1.20.L and RIBI adjuvant (50 μg of peptide/mouse) (as described in the manufacturer's manual) at 7 weeks of age. After 21 days, mice received a booster injection (half-initial dose) and were then bled 7 days later. Titer was defined as the maximum dilution of serum that yielded a change in absorbance equivalent to 2× that of control serum (60 min; 37° C.). The amount of peptide per well was 100 ng. Booster injections took place at 21 days interval. [0271]
  • Blood samples were analyzed for total cholesterol, HDL cholesterol, and triglyceride concentration, using commercially available total cholesterol and triglycerides assay kits (Sigma; St. Louis Mo.) (ELISA). HDL concentration was determined after Apo B containing lipoproteins were precipitated using heparin/MnCl[0272] 2.
  • When fed a normal rodent chow, total serum cholesterol concentration in LDLR knockout mice remains relatively low. A high fat diet, on the other hand, results in an increase in total serum cholesterol concentration in these mice. The animals were thus switched at 16 weeks of age to a modified “Western Type” diet (0.1% cholesterol content) (Harland Teklad, Madison, Wis.) and fed ad libitum. This diet was expected to increase the total serum cholesterol concentration to a range of 600-800 mg/dl, thereby increasing the rate of lesion formation. Blood samples were collected at 18 weeks of age (retro-orbital sinus bleeding technique) and monthly thereafter. [0273]
  • At 30 weeks of age, the mice were sacrificed and aorta were removed as described in Example 22. Aortic atherosclerosis was measured by the cholesterol extraction method described in Example 22, whereby cholesterol is determined as a unit weight of artery. [0274]
  • Immunization against BHF-1 .20.L had no effect on body weight of LDLR knockout mice. Consumption of the modified “Western Type” diet for 12 weeks significantly (P<.05) increased total serum cholesterol, HDL cholesterol and triglycerides in both experimental and control animals. Levels of total serum cholesterol, HDL serum cholesterol and serum triglyceride concentration were not significantly different (P>.05) between experimental and control animals. [0275]
  • Mice that were immunized with the BHF-1 peptide had 24% less aortic cholesterol content (P>0.037) as compared to the control, non-immunized mice. Without being bound to any particular theory, the immunization is thought to generate circulating antibodies against the BHF-1 peptide. These antibodies are thought to block LDL binding to the artery wall, thereby reducing aortic cholesterol content. [0276]
  • Those skilled in the art will be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. These and all other equivalents are intended to be encompassed by the following claims. [0277]
  • 1 53 1 151 PRT Oryctolagus cuniculus 1 Met Ser Lys Asn Thr Val Ser Ser Ala Arg Phe Arg Lys Val Asp Val 1 5 10 15 Asp Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp Gly Gly Asp 20 25 30 Gly Gln Ala Gly Pro Asp Glu Gly Glu Val Asp Ser Cys Leu Arg Gln 35 40 45 Gly Asn Met Thr Ala Ala Leu Gln Ala Ala Leu Lys Asn Pro Pro Ile 50 55 60 Asn Thr Arg Ser Gln Ala Val Lys Asp Arg Ala Gly Ser Ile Val Leu 65 70 75 80 Lys Val Leu Ile Ser Phe Lys Ala Gly Asp Ile Glu Lys Ala Val Gln 85 90 95 Ser Leu Asp Arg Asn Gly Val Asp Leu Leu Met Lys Tyr Ile Tyr Lys 100 105 110 Gly Phe Glu Ser Pro Ser Asp Asn Ser Ser Ala Val Leu Leu Gln Trp 115 120 125 His Glu Lys Ala Leu Ala Ala Gly Gly Val Gly Ser Ile Val Arg Val 130 135 140 Leu Thr Ala Arg Lys Thr Val 145 150 2 317 PRT Oryctolagus cuniculus VARIANT (1)...(317) Xaa = Any Amino Acid 2 Asp Cys Arg Ser Ser Ser Asn Asn Arg Xaa Pro Lys Gly Gly Ala Ala 1 5 10 15 Arg Ala Gly Gly Pro Ala Arg Pro Val Ser Leu Arg Glu Val Val Arg 20 25 30 Tyr Leu Gly Gly Ser Ser Gly Ala Gly Gly Arg Leu Thr Arg Gly Arg 35 40 45 Val Gln Gly Leu Leu Glu Glu Glu Ala Ala Ala Arg Gly Arg Leu Glu 50 55 60 Arg Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly 65 70 75 80 Arg Ala Pro Pro Ala Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala 85 90 95 Gly Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu 100 105 110 Asp Asp Glu Asp Asp Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val 115 120 125 Pro Glu Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly 130 135 140 Gly Glu Arg Gly Pro Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser 145 150 155 160 Leu Cys Gly Pro His Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala 165 170 175 Gly Ser Gly Thr Arg Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu 180 185 190 Gly Gly Ser Ala Ser Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val 195 200 205 Pro Leu Pro Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro 210 215 220 Phe Gly Cys Pro Ala Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu 225 230 235 240 Trp Thr Val Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro 245 250 255 Glu Gln Ala Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu 260 265 270 Leu Leu Met Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu 275 280 285 Gly Pro Ala Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln 290 295 300 Gly His Phe Glu Asp Asp Asp Pro Glu Gly Phe Leu Gly 305 310 315 3 232 PRT Oryctolagus cuniculus 3 Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly Glu Glu Arg Val 1 5 10 15 Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp 20 25 30 Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp Arg 35 40 45 Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Gly Glu Arg Gly Pro 50 55 60 Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu Cys Gly Pro His 65 70 75 80 Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly Ser Gly Thr Arg 85 90 95 Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu Gly Gly Ser Ala Ser 100 105 110 Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro Gly 115 120 125 Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro Ala 130 135 140 Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu Trp Thr Val Met Asp 145 150 155 160 Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr Ala 165 170 175 Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln Arg 180 185 190 Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu Lys 195 200 205 Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu Asp 210 215 220 Asp Asp Pro Glu Gly Phe Leu Gly 225 230 4 252 PRT Oryctolagus cuniculus 4 Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly Arg 1 5 10 15 Ala Pro Pro Ala Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly 20 25 30 Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp 35 40 45 Asp Glu Asp Asp Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val Pro 50 55 60 Glu Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Gly 65 70 75 80 Glu Arg Gly Pro Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu 85 90 95 Cys Gly Pro His Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly 100 105 110 Ser Gly Thr Arg Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu Gly 115 120 125 Gly Ser Ala Ser Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val Pro 130 135 140 Leu Pro Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe 145 150 155 160 Gly Cys Pro Ala Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu Trp 165 170 175 Thr Val Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu 180 185 190 Gln Ala Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu 195 200 205 Leu Met Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly 210 215 220 Pro Ala Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly 225 230 235 240 His Phe Glu Asp Asp Asp Pro Glu Gly Phe Leu Gly 245 250 5 557 PRT Oryctolagus cuniculus 5 Met Lys Asn Gln Asp Lys Lys Asn Gly Ala Ala Lys Gln Pro Asn Pro 1 5 10 15 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Ala Glu Gly Ala Gln Gly 20 25 30 Arg Pro Gly Arg Pro Ala Pro Ala Arg Glu Ala Glu Gly Ala Ser Ser 35 40 45 Gln Ala Pro Gly Arg Pro Glu Gly Ala Gln Ala Lys Thr Ala Gln Pro 50 55 60 Gly Ala Leu Cys Asp Val Ser Glu Glu Leu Ser Arg Gln Leu Glu Asp 65 70 75 80 Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Ala Pro Gly Glu 85 90 95 Asp Gly Val Gln Gly Glu Pro Pro Glu Pro Glu Asp Ala Glu Lys Ser 100 105 110 Arg Ala Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Gly Thr Pro Val 115 120 125 Val Asn Gly Glu Lys Glu Thr Ser Lys Ala Glu Pro Gly Thr Glu Glu 130 135 140 Ile Arg Thr Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro Gln 145 150 155 160 Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu Met 165 170 175 Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala Leu 180 185 190 Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln Lys 195 200 205 Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu Lys 210 215 220 Asp His Leu Arg Gly Glu His Ser Lys Ala Ile Leu Ala Arg Ser Lys 225 230 235 240 Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu Lys 245 250 255 Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys Glu 260 265 270 Val Thr Ser His Phe Gln Met Thr Leu Asn Asp Ile Gln Leu Gln Met 275 280 285 Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met Glu 290 295 300 Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg Glu 305 310 315 320 Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln Leu 325 330 335 Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala Glu 340 345 350 Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val Glu 355 360 365 Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu Lys 370 375 380 Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn Thr 385 390 395 400 Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met Glu 405 410 415 Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met Tyr 420 425 430 Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala Glu 435 440 445 Glu Lys Thr Leu Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys Ile 450 455 460 Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn Asp 465 470 475 480 Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Pro Val 485 490 495 Ser Asp Ser Gly Pro Glu Arg Arg Pro Glu Pro Ala Thr Thr Ser Lys 500 505 510 Glu Gln Gly Val Glu Gly Pro Gly Ala Gln Val Pro Asn Ser Pro Arg 515 520 525 Ala Thr Asp Ala Ser Cys Cys Ala Gly Ala Pro Ser Thr Glu Ala Ser 530 535 540 Gly Gln Thr Gly Pro Gln Glu Pro Thr Thr Ala Thr Ala 545 550 555 6 151 PRT Homo sapiens 6 Met Ser Lys Asn Thr Val Ser Ser Ala Arg Phe Arg Lys Val Asp Val 1 5 10 15 Asp Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp Gly Gly Asp 20 25 30 Gly Gln Ala Gly Pro Asp Glu Gly Glu Val Asp Ser Cys Leu Arg Gln 35 40 45 Gly Asn Met Thr Ala Ala Leu Gln Ala Ala Leu Lys Asn Pro Pro Ile 50 55 60 Asn Thr Lys Ser Gln Ala Val Lys Asp Arg Ala Gly Ser Ile Val Leu 65 70 75 80 Lys Val Leu Ile Ser Phe Lys Ala Asn Asp Ile Glu Lys Ala Val Gln 85 90 95 Ser Leu Asp Lys Asn Gly Val Asp Leu Leu Met Lys Tyr Ile Tyr Lys 100 105 110 Gly Phe Glu Ser Pro Ser Asp Asn Ser Ser Ala Met Leu Leu Gln Trp 115 120 125 His Glu Lys Ala Leu Ala Ala Gly Gly Val Gly Ser Ile Val Arg Val 130 135 140 Leu Thr Ala Arg Lys Thr Val 145 150 7 217 PRT Homo sapiens 7 Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Asp Asp Asp Glu Asp 1 5 10 15 Glu Asp Glu Glu Asp Asp Val Ser Glu Gly Ser Glu Val Pro Glu Ser 20 25 30 Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Glu Arg Gly 35 40 45 Pro Gln Ser Ala Lys Glu Arg Val Lys Glu Trp Thr Pro Cys Gly Pro 50 55 60 His Gln Gly Gln Asp Glu Gly Arg Gly Pro Ala Pro Gly Ser Gly Thr 65 70 75 80 Arg Gln Val Phe Ser Met Ala Ala Met Asn Lys Glu Gly Gly Thr Ala 85 90 95 Ser Val Ala Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro 100 105 110 Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro 115 120 125 Pro Gly Arg Lys Glu Lys Pro Ser Asp Pro Val Glu Trp Thr Val Met 130 135 140 Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr 145 150 155 160 Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln 165 170 175 Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu 180 185 190 Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu 195 200 205 Asp Asp Asp Pro Asp Gly Phe Leu Gly 210 215 8 530 PRT Homo sapiens 8 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Pro Glu Gly Ala Gln Glu 1 5 10 15 Arg Pro Ser Gln Ala Ala Pro Ala Val Glu Ala Glu Gly Pro Gly Ser 20 25 30 Ser Gln Ala Pro Arg Lys Pro Glu Gly Ala Gln Ala Arg Thr Ala Gln 35 40 45 Ser Gly Ala Leu Arg Asp Val Ser Glu Glu Leu Ser Arg Gln Leu Glu 50 55 60 Asp Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Gly Pro Gly 65 70 75 80 Glu Asp Gly Ala Gln Gly Glu Pro Ala Glu Pro Glu Asp Ala Glu Lys 85 90 95 Ser Arg Thr Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Thr Pro Val 100 105 110 Val Tyr Gly Glu Lys Glu Pro Ser Lys Gly Asp Pro Asn Thr Glu Glu 115 120 125 Ile Arg Gln Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro Gln 130 135 140 Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu Met 145 150 155 160 Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala Leu 165 170 175 Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln Lys 180 185 190 Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu Lys 195 200 205 Asp His Leu Arg Gly Glu His Ser Lys Ala Val Leu Ala Arg Ser Lys 210 215 220 Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu Lys 225 230 235 240 Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys Glu 245 250 255 Val Thr Ser His Phe Gln Val Thr Leu Asn Asp Ile Gln Leu Gln Met 260 265 270 Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met Glu 275 280 285 Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg Glu 290 295 300 Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln Leu 305 310 315 320 Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala Glu 325 330 335 Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val Glu 340 345 350 Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu Lys 355 360 365 Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn Thr 370 375 380 Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met Glu 385 390 395 400 Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met Tyr 405 410 415 Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala Glu 420 425 430 Glu Lys Thr Val Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys Ile 435 440 445 Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn Asp 450 455 460 Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Ser Leu 465 470 475 480 Thr Asp Ser Gly Pro Glu Arg Arg Pro Glu Gly Pro Gly Ala Gln Ala 485 490 495 Pro Ser Ser Pro Arg Val Thr Glu Ala Pro Cys Tyr Pro Gly Ala Pro 500 505 510 Ser Thr Glu Ala Ser Gly Gln Thr Gly Pro Gln Glu Pro Thr Ser Ala 515 520 525 Arg Ala 530 9 20 PRT Homo sapiens 9 Val Asp Val Asp Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp 1 5 10 15 Gly Gly Asp Gly 20 10 1404 DNA Oryctolagus cuniculus CDS (58)...(510) 10 aagcctcgca gcggtcgggg cggcgccgcg gaggctcgag ggcggcgggc ggcggcg atg 60 Met 1 tcg aag aac acg gtg tcg tcg gcg cgg ttc cgg aag gtg gac gtg gat 108 Ser Lys Asn Thr Val Ser Ser Ala Arg Phe Arg Lys Val Asp Val Asp 5 10 15 gag tac gac gag aac aag ttc gtg gac gag gaa gac ggc ggc gac ggc 156 Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp Gly Gly Asp Gly 20 25 30 cag gcg ggg ccg gac gag ggc gag gtg gac tcg tgc ctg cgg caa ggg 204 Gln Ala Gly Pro Asp Glu Gly Glu Val Asp Ser Cys Leu Arg Gln Gly 35 40 45 aac atg aca gcc gcc ctg cag gcg gcg ctg aag aac cct ccc atc aac 252 Asn Met Thr Ala Ala Leu Gln Ala Ala Leu Lys Asn Pro Pro Ile Asn 50 55 60 65 acc agg agc cag gcg gtg aag gac cgg gca ggc agc atc gtg ctg aag 300 Thr Arg Ser Gln Ala Val Lys Asp Arg Ala Gly Ser Ile Val Leu Lys 70 75 80 gtg ctc atc tcc ttc aag gcc ggc gac ata gaa aag gcc gtg cag tcc 348 Val Leu Ile Ser Phe Lys Ala Gly Asp Ile Glu Lys Ala Val Gln Ser 85 90 95 ctg gac agg aac ggc gtg gac ctg ctc atg aag tac atc tac aag ggc 396 Leu Asp Arg Asn Gly Val Asp Leu Leu Met Lys Tyr Ile Tyr Lys Gly 100 105 110 ttc gag agc ccc tcc gac aac agc agc gcc gtg ctc ctg cag tgg cac 444 Phe Glu Ser Pro Ser Asp Asn Ser Ser Ala Val Leu Leu Gln Trp His 115 120 125 gag aag gcg ctg gct gca gga gga gtg ggc tcc atc gtc cgt gtc ctg 492 Glu Lys Ala Leu Ala Ala Gly Gly Val Gly Ser Ile Val Arg Val Leu 130 135 140 145 act gca agg aaa acc gtg tagcctggca ggaacgggtg cctgccgggg 540 Thr Ala Arg Lys Thr Val 150 agcgggagct gccggtacaa agaccaaaac gcccagatgc cgccgctgcc ctgtgggcgg 600 cgtctgttcc cagcttcgct ttttcccttt cccgtgtctg tcaggattac ataaggtttc 660 ccttcgtgag aatcggagtg gcgcagaggg tcctgttcat acgcgccgtg cgtccggctg 720 tgtaagaccc ctgccttcag tgtccttgag caacggtagc gtgtcgccgg ctgggtttgg 780 ttttgtcgtg gagggatctg gtcagaattt gaggccagtt tcctaactca ttgctggtca 840 ggaaatgatc ttcatttaaa aaaaaaaaaa agactggcag ctattatgca aaactggacc 900 ctcttccctt atttaagcag agtgagtttc tggaaccagt ggtgcccccc cccccgcccc 960 ggccgccgtc ctgctcaagg gaagcctccc tgcagagcag cagagcccct gggcaggagc 1020 gccgcgtccc gctcccagga gacagcatgc gcggtcacgc ggcacttcct gtgcctccca 1080 gccccagtgc cccggagttc ttcagggcga cagggacctc agaagactgg atccgatcca 1140 gacagacgcc cattcttggt tcagctcagt gttttcaaaa ggaacgtgct accgtgggta 1200 gagcacactg gttctcagaa cacggccggc gcttgacggt tgtcacagct ccagaacaaa 1260 tcctgggaga caggcgagcg cgagtcgccg ggcaggaatt ccacacactc gtgctgtttt 1320 tgatacctgc tttttgtttt gttttgtaaa aatgatgcac ttgagaaaat aaaacgtcag 1380 tgttgacaaa aaaaaaaaaa aaaa 1404 11 1617 DNA Oryctolagus cuniculus CDS (1)...(951) 11 gac tgc cgc agc agc agc aac aac cgc tag ccg aag ggt ggc gcg gcg 48 Asp Cys Arg Ser Ser Ser Asn Asn Arg * Pro Lys Gly Gly Ala Ala 1 5 10 15 cgg gcc ggc ggc ccg gcg cgg ccc gtg agc ctg cgg gaa gtc gtg cgc 96 Arg Ala Gly Gly Pro Ala Arg Pro Val Ser Leu Arg Glu Val Val Arg 20 25 30 tac ctc ggg ggt agc agc ggc gct ggc ggc cgc ctg acc cgc ggc cgc 144 Tyr Leu Gly Gly Ser Ser Gly Ala Gly Gly Arg Leu Thr Arg Gly Arg 35 40 45 gtg cag ggt ctg ctg gaa gag gag gcg gcg gcg cgg ggc cgc ctg gag 192 Val Gln Gly Leu Leu Glu Glu Glu Ala Ala Ala Arg Gly Arg Leu Glu 50 55 60 cgc acc cgt ctc gga gcg ctt gcg ctg ccc cgc ggg gac agg ccc gga 240 Arg Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly 65 70 75 cgg gcg cca ccg gcc gcc agc gcc cgc gcg gcg cgg aac aag aga gct 288 Arg Ala Pro Pro Ala Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala 80 85 90 95 ggc gag gag cga gtg ctt gaa aag gag gag gag gag gag gag gag gaa 336 Gly Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu 100 105 110 gac gac gag gac gac gac gac gac gtc gtg tcc gag ggc tcg gag gtg 384 Asp Asp Glu Asp Asp Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val 115 120 125 ccc gag agc gat cgt ccc gcg ggt gcg cag cat cac cag ctg aat ggc 432 Pro Glu Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly 130 135 140 ggc gag cgc ggc ccg cag acc gcc aag gag cgg gcc aag gag tgg tcg 480 Gly Glu Arg Gly Pro Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser 145 150 155 ctg tgt ggc ccc cac cct ggc cag gag gaa ggg cgg ggg ccg gcc gcg 528 Leu Cys Gly Pro His Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala 160 165 170 175 ggc agt ggc acc cgc cag gtg ttc tcc atg gcg gcc ttg agt aag gag 576 Gly Ser Gly Thr Arg Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu 180 185 190 ggg gga tca gcc tct tcg acc acc ggg cct gac tcc ccg tcc ccg gtg 624 Gly Gly Ser Ala Ser Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val 195 200 205 cct ttg ccc ccc ggg aag cca gcc ctc cca gga gcc gat ggg acc ccc 672 Pro Leu Pro Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro 210 215 220 ttt ggc tgc cct gcc ggg cgc aaa gag aag ccg gca gac ccc gtg gag 720 Phe Gly Cys Pro Ala Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu 225 230 235 tgg aca gtc atg gac gtc gtg gag tac ttc acc gag gcg ggc ttc cct 768 Trp Thr Val Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro 240 245 250 255 gag caa gcc acg gct ttc cag gag cag gag atc gac ggc aag tcc ctg 816 Glu Gln Ala Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu 260 265 270 ctg ctc atg cag cgc acc gat gtc ctc acc ggc ctg tcc atc cgc ctg 864 Leu Leu Met Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu 275 280 285 ggg cca gcg ttg aaa atc tat gag cac cat atc aag gtg ctg cag cag 912 Gly Pro Ala Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln 290 295 300 ggt cac ttc gag gac gat gac ccg gaa ggc ttc ctg gga tgagcacaga 961 Gly His Phe Glu Asp Asp Asp Pro Glu Gly Phe Leu Gly 305 310 315 gccgccgcgc cccttgtccc cacccccacc ccgcctggac ccattcctgc ctccatgtca 1021 cccaaggtgt cccagaggcc aggagctgga ctgggcaggc gaggggtgcg gacctaccct 1081 gattctggta gggggcgggg ccttgctgtg ctcattgcta cccccccacc ccgtgtgtgt 1141 ctctgcacct gcccccagca cacccctccc ggagcctgga tgtcgcctgg gactctggcc 1201 tgctcatttt gcccccagat cagccccctc cctccctcct gtcccaggac attttttaaa 1261 agaaaaaaag gaaaaaaaaa aattggggag ggggctggga aggtgcccca agatcctcct 1321 cggcccaacc aggtgtttat tcctatatat atatatatat gttttgttct gcctgttttt 1381 cgttttttgg tgcgtggcct ttcttccctc ccaccaccac tcatggcccc agccctgctc 1441 gccctgtcgg cgggagcagc tgggaatggg aggagggtgg gaccttgggt ctgtctccca 1501 ccctctctcc cgttggttct gttgtcgctc cagctggctg tattgctttt taatattgca 1561 ccgaagggtt gttttttttt ttttaaataa aattttaaaa aaaggaaaaa aaaaaa 1617 12 1362 DNA Oryctolagus cuniculus CDS (1)...(696) 12 gcc agc gcc cgc gcg gcg cgg aac aag aga gct ggc gag gag cga gtg 48 Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly Glu Glu Arg Val 1 5 10 15 ctt gaa aag gag gag gag gag gag gag gag gaa gac gac gag gac gac 96 Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp 20 25 30 gac gac gac gtc gtg tcc gag ggc tcg gag gtg ccc gag agc gat cgt 144 Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp Arg 35 40 45 ccc gcg ggt gcg cag cat cac cag ctg aat ggc ggc gag cgc ggc ccg 192 Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Gly Glu Arg Gly Pro 50 55 60 cag acc gcc aag gag cgg gcc aag gag tgg tcg ctg tgt ggc ccc cac 240 Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu Cys Gly Pro His 65 70 75 80 cct ggc cag gag gaa ggg cgg ggg ccg gcc gcg ggc agt ggc acc cgc 288 Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly Ser Gly Thr Arg 85 90 95 cag gtg ttc tcc atg gcg gcc ttg agt aag gag ggg gga tca gcc tct 336 Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu Gly Gly Ser Ala Ser 100 105 110 tcg acc acc ggg cct gac tcc ccg tcc ccg gtg cct ttg ccc ccc ggg 384 Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro Gly 115 120 125 aag cca gcc ctc cca gga gcc gat ggg acc ccc ttt ggc tgc cct gcc 432 Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro Ala 130 135 140 ggg cgc aaa gag aag ccg gca gac ccc gtg gag tgg aca gtc atg gac 480 Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu Trp Thr Val Met Asp 145 150 155 160 gtc gtg gag tac ttc acc gag gcg ggc ttc cct gag caa gcc acg gct 528 Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr Ala 165 170 175 ttc cag gag cag gag atc gac ggc aag tcc ctg ctg ctc atg cag cgc 576 Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln Arg 180 185 190 acc gat gtc ctc acc ggc ctg tcc atc cgc ctg ggg cca gcg ttg aaa 624 Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu Lys 195 200 205 atc tat gag cac cat atc aag gtg ctg cag cag ggt cac ttc gag gac 672 Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu Asp 210 215 220 gat gac ccg gaa ggc ttc ctg gga tgagcacaga gccgccgcgc cccttgtccc 726 Asp Asp Pro Glu Gly Phe Leu Gly 225 230 cacccccacc ccgcctggac ccattcctgc ctccatgtca cccaaggtgt cccagaggcc 786 aggagctgga ctgggcaggc gaggggtgcg gacctaccct gattctggta gggggcgggg 846 ccttgctgtg ctcattgcta cccccccacc ccgtgtgtgt ctctgcacct gcccccagca 906 cacccctccc ggagcctgga tgtcgcctgg gactctggcc tgctcatttt gcccccagat 966 cagccccctc cctccctcct gtcccaggac attttttaaa agaaaaaaag gaaaaaaaaa 1026 aattggggag ggggctggga aggtgcccca agatcctcct cggcccaacc aggtgtttat 1086 tcctatatat atatatatat gttttgttct gcctgttttt cgttttttgg tgcgtggcct 1146 ttcttccctc ccaccaccac tcatggcccc agccctgctc gccctgtcgg cgggagcagc 1206 tgggaatggg aggagggtgg gaccttgggt ctgtctccca ccctctctcc cgttggttct 1266 gttgtcgctc cagctggctg tattgctttt taatattgca ccgaagggtt gttttttttt 1326 ttttaaataa aattttaaaa aaaggaaaaa aaaaaa 1362 13 1422 DNA Oryctolagus cuniculus CDS (1)...(756) 13 acc cgt ctc gga gcg ctt gcg ctg ccc cgc ggg gac agg ccc gga cgg 48 Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly Arg 1 5 10 15 gcg cca ccg gcc gcc agc gcc cgc gcg gcg cgg aac aag aga gct ggc 96 Ala Pro Pro Ala Ala Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly 20 25 30 gag gag cga gtg ctt gaa aag gag gag gag gag gag gag gag gaa gac 144 Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp 35 40 45 gac gag gac gac gac gac gac gtc gtg tcc gag ggc tcg gag gtg ccc 192 Asp Glu Asp Asp Asp Asp Asp Val Val Ser Glu Gly Ser Glu Val Pro 50 55 60 gag agc gat cgt ccc gcg ggt gcg cag cat cac cag ctg aat ggc ggc 240 Glu Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Gly 65 70 75 80 gag cgc ggc ccg cag acc gcc aag gag cgg gcc aag gag tgg tcg ctg 288 Glu Arg Gly Pro Gln Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu 85 90 95 tgt ggc ccc cac cct ggc cag gag gaa ggg cgg ggg ccg gcc gcg ggc 336 Cys Gly Pro His Pro Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly 100 105 110 agt ggc acc cgc cag gtg ttc tcc atg gcg gcc ttg agt aag gag ggg 384 Ser Gly Thr Arg Gln Val Phe Ser Met Ala Ala Leu Ser Lys Glu Gly 115 120 125 gga tca gcc tct tcg acc acc ggg cct gac tcc ccg tcc ccg gtg cct 432 Gly Ser Ala Ser Ser Thr Thr Gly Pro Asp Ser Pro Ser Pro Val Pro 130 135 140 ttg ccc ccc ggg aag cca gcc ctc cca gga gcc gat ggg acc ccc ttt 480 Leu Pro Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe 145 150 155 160 ggc tgc cct gcc ggg cgc aaa gag aag ccg gca gac ccc gtg gag tgg 528 Gly Cys Pro Ala Gly Arg Lys Glu Lys Pro Ala Asp Pro Val Glu Trp 165 170 175 aca gtc atg gac gtc gtg gag tac ttc acc gag gcg ggc ttc cct gag 576 Thr Val Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu 180 185 190 caa gcc acg gct ttc cag gag cag gag atc gac ggc aag tcc ctg ctg 624 Gln Ala Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu 195 200 205 ctc atg cag cgc acc gat gtc ctc acc ggc ctg tcc atc cgc ctg ggg 672 Leu Met Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly 210 215 220 cca gcg ttg aaa atc tat gag cac cat atc aag gtg ctg cag cag ggt 720 Pro Ala Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly 225 230 235 240 cac ttc gag gac gat gac ccg gaa ggc ttc ctg gga tgagcacaga 766 His Phe Glu Asp Asp Asp Pro Glu Gly Phe Leu Gly 245 250 gccgccgcgc cccttgtccc cacccccacc ccgcctggac ccattcctgc ctccatgtca 826 cccaaggtgt cccagaggcc aggagctgga ctgggcaggc gaggggtgcg gacctaccct 886 gattctggta gggggcgggg ccttgctgtg ctcattgcta cccccccacc ccgtgtgtgt 946 ctctgcacct gcccccagca cacccctccc ggagcctgga tgtcgcctgg gactctggcc 1006 tgctcatttt gcccccagat cagccccctc cctccctcct gtcccaggac attttttaaa 1066 agaaaaaaag gaaaaaaaaa aattggggag ggggctggga aggtgcccca agatcctcct 1126 cggcccaacc aggtgtttat tcctatatat atatatatat gttttgttct gcctgttttt 1186 cgttttttgg tgcgtggcct ttcttccctc ccaccaccac tcatggcccc agccctgctc 1246 gccctgtcgg cgggagcagc tgggaatggg aggagggtgg gaccttgggt ctgtctccca 1306 ccctctctcc cgttggttct gttgtcgctc cagctggctg tattgctttt taatattgca 1366 ccgaagggtt gttttttttt ttttaaataa aattttaaaa aaaggaaaaa aaaaaa 1422 14 4722 DNA Oryctolagus cuniculus CDS (61)...(1731) 14 gtggaaaata gcaactgtgt ttctcaagga tccaatccca acctaaggtg gcagcgcaca 60 atg aag aat caa gac aaa aag aac ggg gct gcc aaa cag ccc aac ccc 108 Met Lys Asn Gln Asp Lys Lys Asn Gly Ala Ala Lys Gln Pro Asn Pro 1 5 10 15 aaa agc agc ccg gga cag ccg gaa gca gga gcg gag gga gcc cag ggg 156 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Ala Glu Gly Ala Gln Gly 20 25 30 cgg ccc ggc cgg ccg gcc ccc gcc cga gaa gcc gaa ggt gcc agc agc 204 Arg Pro Gly Arg Pro Ala Pro Ala Arg Glu Ala Glu Gly Ala Ser Ser 35 40 45 cag gct ccc ggg agg ccg gag ggg gct caa gcc aaa act gct cag cct 252 Gln Ala Pro Gly Arg Pro Glu Gly Ala Gln Ala Lys Thr Ala Gln Pro 50 55 60 ggg gcg ctc tgt gat gtc tct gag gag ctg agc cgc cag ttg gaa gac 300 Gly Ala Leu Cys Asp Val Ser Glu Glu Leu Ser Arg Gln Leu Glu Asp 65 70 75 80 ata ctc agt aca tac tgt gtg gac aac aac cag ggg gcc ccg ggt gag 348 Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Ala Pro Gly Glu 85 90 95 gat ggg gtc cag ggt gag ccc cct gaa cct gaa gat gca gag aag tct 396 Asp Gly Val Gln Gly Glu Pro Pro Glu Pro Glu Asp Ala Glu Lys Ser 100 105 110 cgc gcc tat gtg gca agg aat ggg gag ccg gag ccg ggc acc cca gta 444 Arg Ala Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Gly Thr Pro Val 115 120 125 gtc aat ggc gag aag gag acc tcc aag gca gag ccg ggc acg gaa gag 492 Val Asn Gly Glu Lys Glu Thr Ser Lys Ala Glu Pro Gly Thr Glu Glu 130 135 140 atc cgg acg agc gat gag gtc gga gac cga gac cac cgg agg cca cag 540 Ile Arg Thr Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro Gln 145 150 155 160 gaa aag aag aag gcc aag ggt ctg gga aag gag atc acg ctg ctg atg 588 Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu Met 165 170 175 cag aca ctg aac acg ctg agc acc cca gag gag aag ctg gcg gct ctg 636 Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala Leu 180 185 190 tgc aag aag tat gcg gaa ctg ctc gag gag cac cgg aac tcg cag aag 684 Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln Lys 195 200 205 cag atg aag ctg ctg cag aag aag cag agc cag ctg gtg cag gag aag 732 Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu Lys 210 215 220 gac cac ctg cgt ggc gag cac agc aag gcc atc ctg gcc cgc agc aag 780 Asp His Leu Arg Gly Glu His Ser Lys Ala Ile Leu Ala Arg Ser Lys 225 230 235 240 ctc gag agc ctg tgc cgg gag ctg cag cgg cac aac cgc tcg ctc aag 828 Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu Lys 245 250 255 gaa gaa ggt gtg cag cga gcc cga gag gag gag gag aag cgc aag gag 876 Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys Glu 260 265 270 gtg acg tca cac ttc cag atg acg ctc aac gac att cag ctg cag atg 924 Val Thr Ser His Phe Gln Met Thr Leu Asn Asp Ile Gln Leu Gln Met 275 280 285 gag cag cac aac gag cgc aac tcc aag ctg cgc cag gag aac atg gag 972 Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met Glu 290 295 300 ctg gcc gag cgg ctc aag aag ctg att gag cag tac gag ctg cga gaa 1020 Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg Glu 305 310 315 320 gag cac atc gac aaa gtc ttc aaa cac aag gat ctg cag cag cag ctg 1068 Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln Leu 325 330 335 gtg gac gcc aag ctc cag cag gcc cag gag atg ctg aag gag gca gag 1116 Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala Glu 340 345 350 gag cgg cac cag cgg gag aag gac ttt ctc ctg aag gag gcc gtg gag 1164 Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val Glu 355 360 365 tcc cag agg atg tgc gag ctg atg aag caa cag gag acc cac ctg aag 1212 Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu Lys 370 375 380 cag cag ctt gcc cta tac aca gag aag ttt gag gag ttc cag aac act 1260 Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn Thr 385 390 395 400 ctt tcc aaa agc agc gag gtg ttc acc aca ttc aaa cag gaa atg gaa 1308 Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met Glu 405 410 415 aag atg aca aag aag atc aag aag ctg gag aaa gag acc acc atg tac 1356 Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met Tyr 420 425 430 cgt tcc cgg tgg gag agc agc aac aag gcc ctg ctt gag atg gct gag 1404 Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala Glu 435 440 445 gag aaa aca ctc cgg gac aaa gag ctg gaa ggc ctg cag gtg aaa atc 1452 Glu Lys Thr Leu Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys Ile 450 455 460 cag cgg ctg gag aag ctg tgc cgg gca ctg cag aca gag cgc aat gac 1500 Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn Asp 465 470 475 480 ctg aac aag agg gtg cag gac ctg agt gcc ggt ggc cag ggc ccc gtc 1548 Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Pro Val 485 490 495 tcc gac agc ggt cct gag cgg agg cca gag ccc gcc acc acc tcc aag 1596 Ser Asp Ser Gly Pro Glu Arg Arg Pro Glu Pro Ala Thr Thr Ser Lys 500 505 510 gag cag ggt gtc gag ggc ccc ggg gct caa gta ccc aac tct cca agg 1644 Glu Gln Gly Val Glu Gly Pro Gly Ala Gln Val Pro Asn Ser Pro Arg 515 520 525 gcc aca gac gct tcc tgc tgc gca ggt gca ccc agc aca gag gca tca 1692 Ala Thr Asp Ala Ser Cys Cys Ala Gly Ala Pro Ser Thr Glu Ala Ser 530 535 540 ggc cag aca ggg ccc cag gag ccc acc act gcc act gcc tagagagctt 1741 Gly Gln Thr Gly Pro Gln Glu Pro Thr Thr Ala Thr Ala 545 550 555 ggtgctgggg tgtgccagga agggagcagg cagcccagcc aggcctggcc cagcccaggc 1801 tcccatgcta agcagtccgg tgctgaggcc aggatgttct gacctggctg gcacctgacc 1861 ctctgcagtc ttggattttg tgggtcagtt ttacatgcat atggcacaca tgcaaggcct 1921 cacacatttg tgtctctaag tgtactgtgg gcttgcatcg ggggtgacga tggacagatg 1981 aagccagcgg ctcccttgtg agctgaagtc ttacggagga gacggcgtct gcactgccat 2041 cgcagtgacc tgcaggacga gttccttgag ctttccctgc ctgctttgag gctgagaccc 2101 ctcccggccc ttcagagctc ctgacaggtg atacacaccc agccttgacc gcacttctct 2161 tgggtagctg ggctctccta gcctccccca gaggcgccat tgcttctctt gacttggaga 2221 ggggatgccc aggcgtggcc ttggcaggca ctgggagcta gtgattgggc tgctctcctg 2281 cctcgagcag gggcaggagt gtttctggtg ggatgatgcg ctcgctggtc aggagccccg 2341 tgggcgctgc ttcccccgcc ctctggtgat gccaggacca ggccagtgat gcttctcagt 2401 agccttacca ttcacaggtg cctctccagc ccgcacagtg agtgacaaga tcatccaaag 2461 gattccttct gaaggtgttc gtttcgtttt gttttgttgc acgtgacggt ttgtattgag 2521 gaccctctga ggaagagggg tgctgtagca gtggtccctg cgtgcctggc tccagtgtcc 2581 tgccctcccc cccctcgcca tggctcctcg gccgccttgg tgctgaggtt tctgtttggt 2641 gagatcaggt tgtctgttca gagagaagag gcgtctgatg gctttgccgc cagcttgcct 2701 gcgggcctca atcccgggag gccgcccggt tcccgtcact gttgtccccg tgcagtgcgt 2761 tgctggtccc caggaccagc tgctcgtttg ctgtatgggt cagtttctgc ttcctgcccc 2821 ccactccacc taactgcaat ccttggggtt tccctggttc tcgtccctgg tacctctgtg 2881 cccaagaagt agccttcttt gggattcttg ttctgcccat gcgggagctg ctgctgtctg 2941 acaggtgagg cctgagactc agcggctgac agagctgcag agctctgcac ggtggctccc 3001 ggggcggcct ctgtgtgctg cacaccgctg ctctgctggc actggccagt ctgtgcagag 3061 catttgagta ctggctcagg agggagggct ctgctggcct cgagggacag cgccacgtct 3121 ccagctgggc tcagggagag ccccagactg gctgcgtagg gtgcttgggg tttgcttctt 3181 gcagtatttc ttggaagctg ttttgttgtc ctgctattcc ttcatcttcc acagtccacg 3241 ctcagccttt aacttggatc cctcacataa cagggttcat gagacccgca agtacgccca 3301 agctacgtat ggctgaggcc agctggcagg tgaatggcac gccattgctg ctgctaatcc 3361 ctggcatatc tttagttcac ctcgaaatgc ccccgccaca gtgcaagcag tgagtccacg 3421 tgccaccctg ggctgaatcc caccccctgt gagtgttgcc cgagattgtg tctcttctga 3481 atgccttcac tgggaatggc ctctgccgcc tcctgctcag ggaggctttc cccttccctc 3541 agcccctgtg ccagactgag gtacaagaac cgccaagccc atgcaaggtg tggctaggcg 3601 ccagggtgca ggaaggaggc aggtagctgc ctgcaccctt gaaagccaag aggcctacgg 3661 tggcctccat cctggcttgc ctcacttcag ctacctcgca tagcccaggg gtggggctat 3721 tggattccag ggtgggggga tgggaagctg cagggggcag gtggctctca ctaggcttcc 3781 cagctcagga atgtgggcct caggtagggg agagcctttg ctccactcca cccatttgca 3841 ggcatctagg ccagtctaga tggcgacccc ttctcttcct ctccattgac caaatcgtac 3901 ctgtctctcc agctgctcgc ttgctctgct ttccaaagtc agcccaggta cccaggtgcc 3961 gcccacattg gcctggaacc tggaccagag gcaagggagg tggcctatcc ttgagtgata 4021 gccagtgcct tcctcacccg gtggcttcca tgcctgtgac ctcagattta ggaccaagag 4081 ctgtgttggt ttcttacgtt gtgagctttc cctccagggg accacagcag gtgaggctcg 4141 gagcccagag cccttggcgc cgccagcagt aacttgtgtc cggaccttgt ccagctgagc 4201 gcttcgtgta tgactcagct tcgtgtgtga gtccagcgga gtgcgtcacg tgacctagac 4261 tcagcggtgt cagccgcact ttgatttgtt tgttttccat gaggtttttg gaccatgggc 4321 ttagctcagg caacttttct gtaaggagaa tgttaacttt ctgtaaagat gcttatttaa 4381 ctaacgcctg cttcccccac tcccaaccag gtggccaccg agagctcacc aggaggccaa 4441 tagagctgct ccagctctcc catcttgcac cgcacaaagg tggccgcccc agggacagcc 4501 aggcacctgc ctgggggagg ggcttctctt ccttatggcc tggccatcta gattgtttaa 4561 agttgtgctg acagcttttt ttggtttttt ggtttttgtt tttgtttttg tttttgtttt 4621 tgtctacttt tggtattcac aacagccagg gacttgattt tgatgtattt taagccacat 4681 taaataaaga gtctgttgcc ttaaaaaaaa aaaaaaaaaa a 4722 15 1925 DNA Homo sapiens CDS (118)...(570) 15 gacgcctcag agcggaacag ggaagtgaat caggcgccgg gtagtgggtt gctgggctgg 60 gcttgctgag gtagaggcag cgccaagaag aggcctttgc cgctggtcgg gattggg atg 120 Met 1 tcg aag aac aca gtg tcg tcg gcc cgc ttc cgg aag gtg gac gtg gat 168 Ser Lys Asn Thr Val Ser Ser Ala Arg Phe Arg Lys Val Asp Val Asp 5 10 15 gaa tat gac gag aac aag ttc gtg gac gaa gaa gat ggg ggc gac ggc 216 Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp Gly Gly Asp Gly 20 25 30 cag gcc ggg ccc gac gag ggc gag gtg gac tcc tgc ctg cgg caa gga 264 Gln Ala Gly Pro Asp Glu Gly Glu Val Asp Ser Cys Leu Arg Gln Gly 35 40 45 aac atg aca gct gcc cta cag gca gct ctg aag aac ccc cct atc aac 312 Asn Met Thr Ala Ala Leu Gln Ala Ala Leu Lys Asn Pro Pro Ile Asn 50 55 60 65 acc aag agt cag gca gtg aag gac cgg gca ggc agc att gtc ttg aag 360 Thr Lys Ser Gln Ala Val Lys Asp Arg Ala Gly Ser Ile Val Leu Lys 70 75 80 gtg ctc atc tct ttt aaa gct aat gat ata gaa aag gca gtt caa tct 408 Val Leu Ile Ser Phe Lys Ala Asn Asp Ile Glu Lys Ala Val Gln Ser 85 90 95 ctg gac aag aat ggt gtg gat ctc cta atg aag tat att tat aaa gga 456 Leu Asp Lys Asn Gly Val Asp Leu Leu Met Lys Tyr Ile Tyr Lys Gly 100 105 110 ttt gag agc ccg tct gac aat agc agt gct atg tta ctg caa tgg cat 504 Phe Glu Ser Pro Ser Asp Asn Ser Ser Ala Met Leu Leu Gln Trp His 115 120 125 gaa aag gca ctt gct gct gga gga gta ggg tcc att gtt cgt gtc ttg 552 Glu Lys Ala Leu Ala Ala Gly Gly Val Gly Ser Ile Val Arg Val Leu 130 135 140 145 act gca aga aaa act gtg tagtctggca ggaagtggat tatctgcctc 600 Thr Ala Arg Lys Thr Val 150 gggagtggga attgctggta caaagaccaa aacaaccaaa tgccaccgct gccctgtggg 660 tagcatctgt ttctctcagc tttgccttct tgctttttca tatctgtaaa gaaaaaaatt 720 acatatcagt tgtcccttta atgaaaattg ggataatata gaagaaattg tgttaaaata 780 gaagtgtttc atcctttcaa aaccatttca gtgatgttta taccaatctg tatatagtat 840 aatttacatt caagttttaa ttgtgcaact tttaaccctg ttggctggtt tttggttctg 900 tttggttttg tattattttt aactaatact gaaaaatttg gtcagaattt gaggccagtt 960 tcctagctca ttgctagtca ggaaatgata tttataaaaa atatgagaga ctggcagcta 1020 ttaacattgc aaaactggac catatttccc ttatttaata agcaaaatat gtttttggaa 1080 taagtggtgg gtgaatacca ctgctaagtt atagctttgt ttttgcttgc ctcctcatta 1140 tctgtactgt gggtttaagt atgctacttt ctctcagcat ccaataatca tggcccctca 1200 atttatttgt ggtcacgcag ggttcagagc aagaagtctt gctttataca aatgtatcca 1260 taaaatatca gagcttgttg ggcatgaaca tcaaactttt gttccactaa tatggctctg 1320 tttggaaaaa actgcaaatc agaaagaatg atttgcagaa agaaagaaaa actatggtgt 1380 aatttaaact ctgggcagcc tctgaatgaa atgctacttt ctttagaaat ataatagctg 1440 ccttagacat tatgaggtat acaactagta tttaagatac catttaatat gccccgtaaa 1500 tgtcttcagt gttcttcagg gtagttggga tctcaaaaga tttggttcag atccaaacaa 1560 atacacattc tgtgttttag ctcagtgttt tctaaaaaaa gaaactgcca cacagcaaaa 1620 aattgtttac tttgttggac aaaccaaatc agttctcaaa aaatgaccgg tgcttataaa 1680 aagttataaa tatcgagtag ctctaaaaca aaccacctga ccaagaggga agtgagcttg 1740 tgcttagtat ttacattgga tgccagtttt gtaatcactg acttatgtgc aaactggtgc 1800 agaaattcta taaactcttt gctgtttttg atacctgctt tttgtttcat tttgttttgt 1860 tttgtaaaaa tgataaaact tcagaaaata aaatgtcagt gttgaataat taaaaaaaaa 1920 aaaaa 1925 16 1208 DNA Homo sapiens CDS (1)...(651) 16 gaa gag cga gta ctt gag aaa gaa gag gaa gaa gat gat gat gaa gat 48 Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Asp Asp Asp Glu Asp 1 5 10 15 gaa gat gaa gaa gat gat gtg tca gag ggc tct gaa gtg ccc gag agt 96 Glu Asp Glu Glu Asp Asp Val Ser Glu Gly Ser Glu Val Pro Glu Ser 20 25 30 gac cgt cct gca ggt gcc cag cac cac cag ctt aac ggc gag cgg gga 144 Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Glu Arg Gly 35 40 45 cct cag agt gcc aag gag agg gtc aag gag tgg acc ccc tgc gga ccg 192 Pro Gln Ser Ala Lys Glu Arg Val Lys Glu Trp Thr Pro Cys Gly Pro 50 55 60 cac cag ggc cag gat gaa ggg cgg ggg cca gcc ccg ggc agc ggc acc 240 His Gln Gly Gln Asp Glu Gly Arg Gly Pro Ala Pro Gly Ser Gly Thr 65 70 75 80 cgc cag gtg ttc tcc atg gca gcc atg aac aag gaa ggg gga aca gct 288 Arg Gln Val Phe Ser Met Ala Ala Met Asn Lys Glu Gly Gly Thr Ala 85 90 95 tct gtt gcc acc ggg cca gac tcc ccg tcc ccc gtg cct ttg ccc cca 336 Ser Val Ala Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro 100 105 110 ggc aaa cca gcc cta cct ggg gcc gac ggg acc ccc ttt ggc tgt cct 384 Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro 115 120 125 ccc ggg cgc aaa gag aag cca tct gat ccc gtc gag tgg acc gtg atg 432 Pro Gly Arg Lys Glu Lys Pro Ser Asp Pro Val Glu Trp Thr Val Met 130 135 140 gat gtc gtc gaa tat ttt act gag gct gga ttc ccg gag cag gcg aca 480 Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr 145 150 155 160 gct ttc caa gag cag gaa att gat ggc aaa tct ttg ctg ctc atg cag 528 Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln 165 170 175 cgc aca gat gtg ctc acc ggc ctg tcc atc cgc ctc ggg cca gcc ctg 576 Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu 180 185 190 aaa atc tac gag cac cac atc aag gtg ctt cag caa ggc cac ttt gag 624 Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu 195 200 205 gat gat gac ccc gat ggc ttc tta ggc tgagcgccca gcctcacccc 671 Asp Asp Asp Pro Asp Gly Phe Leu Gly 210 215 tgccccagcc cattccggcc cccatctcac ccaagatccc ccagagtcca ggagctggac 731 ggggacaccc tcagccctca taacagattc caaggagagg gcaccctctt gtccttatct 791 ttgccccttg tgtctgtctc acacacatct gctcctcagc acgtcggtgt ggggagggga 851 ttgctcctta aaccccaggt ggctgaccct ccccacccag tccaggacat tttaggaaaa 911 aaaaaatgaa atgtgggggg cttctcatct ccccaagatc ctcttccgtt cagccagatg 971 tttcctgtat aaatgtttgg atctgcctgt ttattttggt gggtggtctt tcctccctcc 1031 cctaccaccc atgcccccct tctcagtctg cccctggcct ccagccccta ggggactagc 1091 tgggttgggg ttcctcgggc cttttctctc ctccctcttt tctttctgtt gattgtcgct 1151 ccagctggct gtattgcttt ttaatattgc accgaaggtt ttttaaataa aatttta 1208 17 4697 DNA Homo sapiens CDS (3)...(1592) 17 ca aaa agc agc cca gga caa ccg gaa gca gga ccc gag gga gcc cag 47 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Pro Glu Gly Ala Gln 1 5 10 15 gag cgg ccc agc cag gcg gct cct gca gta gaa gca gaa ggt ccc ggc 95 Glu Arg Pro Ser Gln Ala Ala Pro Ala Val Glu Ala Glu Gly Pro Gly 20 25 30 agc agc cag gct cct cgg aag ccg gag ggg gct caa gcc aga acg gct 143 Ser Ser Gln Ala Pro Arg Lys Pro Glu Gly Ala Gln Ala Arg Thr Ala 35 40 45 cag tct ggg gcc ctt cgt gat gtc tct gag gag ctg agc cgc caa ctg 191 Gln Ser Gly Ala Leu Arg Asp Val Ser Glu Glu Leu Ser Arg Gln Leu 50 55 60 gaa gac ata ctg agc aca tac tgt gtg gac aat aac cag ggg ggc ccc 239 Glu Asp Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Gly Pro 65 70 75 ggc gag gat ggg gca cag ggt gag ccg gct gaa ccc gaa gat gca gag 287 Gly Glu Asp Gly Ala Gln Gly Glu Pro Ala Glu Pro Glu Asp Ala Glu 80 85 90 95 aag tcc cgg acc tat gtg gca agg aat ggg gag cct gaa cca act cca 335 Lys Ser Arg Thr Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Thr Pro 100 105 110 gta gtc tat gga gag aag gaa ccc tcc aag ggg gat cca aac aca gaa 383 Val Val Tyr Gly Glu Lys Glu Pro Ser Lys Gly Asp Pro Asn Thr Glu 115 120 125 gag atc cgg cag agt gac gag gtc gga gac cga gac cat cga agg cca 431 Glu Ile Arg Gln Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro 130 135 140 cag gag aag aaa aaa gcc aag ggt ttg ggg aag gag atc acg ttg ctg 479 Gln Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu 145 150 155 atg cag aca ttg aat act ctg agt acc cca gag gag aag ctg gct gct 527 Met Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala 160 165 170 175 ctg tgc aag aag tat gct gaa ctg ctg gag gag cac cgg aat tca cag 575 Leu Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln 180 185 190 aag cag atg aag ctc cta cag aaa aag cag agc cag ctg gtg caa gag 623 Lys Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu 195 200 205 aag gac cac ctg cgc ggt gag cac agc aag gcc gtc ctg gcc cgc agc 671 Lys Asp His Leu Arg Gly Glu His Ser Lys Ala Val Leu Ala Arg Ser 210 215 220 aag ctt gag agc cta tgc cgt gag ctg cag cgg cac aac cgc tcc ctc 719 Lys Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu 225 230 235 aag gaa gaa ggt gtg cag cgg gcc cgg gag gag gag gag aag cgc aag 767 Lys Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys 240 245 250 255 gag gtg acc tcg cac ttc cag gtg aca ctg aat gac att cag ctg cag 815 Glu Val Thr Ser His Phe Gln Val Thr Leu Asn Asp Ile Gln Leu Gln 260 265 270 atg gaa cag cac aat gag cgc aac tcc aag ctg cgc caa gag aac atg 863 Met Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met 275 280 285 gag ctg gct gag agg ctc aag aag ctg att gag cag tat gag ctg cgc 911 Glu Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg 290 295 300 gag gag cat atc gac aaa gtc ttc aaa cac aag gac cta caa cag cag 959 Glu Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln 305 310 315 ctg gtg gat gcc aag ctc cag cag gcc cag gag atg cta aag gag gca 1007 Leu Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala 320 325 330 335 gaa gag cgg cac cag cgg gag aag gat ttt ctc ctg aaa gag gca gta 1055 Glu Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val 340 345 350 gag tcc cag agg atg tgt gag ctg atg aag cag caa gag acc cac ctg 1103 Glu Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu 355 360 365 aag caa cag ctt gcc cta tac aca gag aag ttt gag gag ttc cag aac 1151 Lys Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn 370 375 380 aca ctt tcc aaa agc agc gag gta ttc acc aca ttc aag cag gag atg 1199 Thr Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met 385 390 395 gaa aag atg act aag aag atc aag aag ctg gag aaa gaa acc acc atg 1247 Glu Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met 400 405 410 415 tac cgg tcc cgg tgg gag agc agc aac aag gcc ctg ctt gag atg gct 1295 Tyr Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala 420 425 430 gag gag aaa aca gtc cgg gat aaa gaa ctg gag ggc ctg cag gta aaa 1343 Glu Glu Lys Thr Val Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys 435 440 445 atc caa cgg ctg gag aag ctg tgc cgg gca ctg cag aca gag cgc aat 1391 Ile Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn 450 455 460 gac ctg aac aag agg gta cag gac ctg agt gct ggt ggc cag ggc tcc 1439 Asp Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Ser 465 470 475 ctc act gac agt ggc cct gag agg agg cca gag ggg cct ggg gct caa 1487 Leu Thr Asp Ser Gly Pro Glu Arg Arg Pro Glu Gly Pro Gly Ala Gln 480 485 490 495 gca ccc agc tcc ccc agg gtc aca gaa gcg cct tgc tac cca gga gca 1535 Ala Pro Ser Ser Pro Arg Val Thr Glu Ala Pro Cys Tyr Pro Gly Ala 500 505 510 ccg agc aca gaa gca tca ggc cag act ggg cct caa gag ccc acc tcc 1583 Pro Ser Thr Glu Ala Ser Gly Gln Thr Gly Pro Gln Glu Pro Thr Ser 515 520 525 gcc agg gcc tagagagcct ggtgttgggt catgctggga agggagcggc 1632 Ala Arg Ala 530 agcccagcca ggcctggccc ataaaaggct cccatgctga gcagcccatt gctgaagcca 1692 ggatgttctt gacctggctg gcatctggca cttgcaattt tggattttgt gggtcagttt 1752 tacgtacata gggcattttg caaggccttg caaatgcatt tatacctgta agtgtacagt 1812 gggcttgcat tggggatggg ggtgtgtaca gatgaagtca gtggcttgtc tgtgagctga 1872 agagtcttga gaggggctgt catctgtagc tgccatcaca gtgagttggc agaagtgact 1932 tgagcatttc tctgtctgat ttgaggctca gacccctccc tgccctttca gagctcaaaa 1992 caagtaatac accaaggtct tgactgcatt tgtcttgtga gcagggcttg cttggtcagc 2052 tcaggccctc ctagctgctt ggaggctcct ttgattctct agacctggaa aaggtgtccc 2112 taggcagagc cctggcaggg cgctcagagc tgggatttcc tgcctggaac aagggacctg 2172 gagaatgttt ttgcgtggga tgatgtgctg gtcaggagcc ccttgggcat cgcttcccct 2232 gccctttggt agtgccagga ccaggccaat gatgcttctc agtagcctta tcattcacag 2292 gtgcctctct agcctgcaca aatgattgac aagagatcac ccaaaggatt atttctgaag 2352 gtgttttttt ctttatttct ttttcttttt ttttttttct ttttcttttt tttttgcaca 2412 tgacagtgtt tgtattgagg accttccaag gaaaagggat gctgtaccag tggtgcctgg 2472 gtgcctggcc tccagtgtcc cacctccttc accaccccac ttggctcctt tgccatcttg 2532 atgctgaggt ttcctgtttg gtgagatcag gttgtttgtg gtaaaagaaa ggaaagggct 2592 tctgatggct ttgccacaag cttacctgtg ggtttcagtc ctgagaggcc accaccagtt 2652 cccatcagca ctgtctccat gcagcagttg ctgggtccca tgtccagctg cctctttggc 2712 ttcatgggtt tttctgcttc ctgcccccac ccccacatgt gcaatcctca agatttgtcc 2772 tgattctatt tcctggcacc tccctgcctg tccttgggga ttctacttct tcctgtgtgg 2832 ggcccatagc tgttgtctaa caggtaagaa atgaaattga actattgact gggccccaga 2892 aatccataaa atggctgcag acagttgttt ctgtgtcctg ttctaccccc actccagtac 2952 ataactacta tgtactgtgt agagccattc tatatgctga atgttctgct gttgcaaact 3012 tgccagggta ttagccagtg tttgtgccaa gcagttttcg gggacaacag aatgactcag 3072 accaagatgg ataggatggt tagggctttg cttcttgctg tttttctttg aactagtcat 3132 tgtcctgcag gtcccttcat cttccatacc tagcccactc ttttagccct taccttaaat 3192 ctctcagata agttggttca caaagaatgt taagtactga atcatgtgtg actgagacca 3252 gagatggcaa atgaatggca caccatttct ccttctcctg ccccagggca ggtaccactg 3312 atctgcatca gagttgcctg ctattctctg gtgtatcctt cacatctagg tgccctcaag 3372 cagctgtgtg agtgttgaga tctctgccat ctctggctga gatactgctg tcctgtgaag 3432 tgtttcccat gacctttttc ttcccctttg aatccctctt gtctggagta gtccttgcct 3492 tcttcttgct ccagtaggcc ttttccttac cccagccctt gtgccaggct aagctggtac 3552 aagagctgcc aactcacaga gttttgctag gcgagagagg tgcagggaag aggcagaggt 3612 atgcaccttc ccccttgaag agaggggaaa ggcctacagt ggcccacata attgcctgac 3672 tcacacttca gctacctctt aatgcctgtg gagggactgg agctgctgga tcccagtgtg 3732 gtggtgtagg aggccacagt gagcaggtgg ccccagctgg gtttcccagg tcaggaatgt 3792 gggccccagg caaggtgcag cctttgctca cagctccatc catgtctaga ccttcaggcc 3852 agtctgcaga tgaggttccc tacctttttc ttctcttcat tgaccaaatc aaccaatcac 3912 tacagctgct ctgcttctgc tttccaaagt agcccaggtc ctgggccaga tgcaggggag 3972 gtgcctatcc atgagtgaag gccagtgtct tcctcacctg ggtggtccca cacttgtgac 4032 cctcagtttt aggacccaag atctgtgttg gtttcttaga ttgctagctt ttcctccagg 4092 ggaccacagc aggtgaagct caagagcgca tggctctgct aatagtaaat tgttttcagg 4152 gccttgtcca gctgagagct tcatgtccac cagattctga gaggtgtcag cagcactttt 4212 tttttttatt tgttgtttgt tttccatgag gttatcggac catgggctga gctcaggcac 4272 tttctgtagg agactgttat ttctgtaaag atggttattt aaccctcctc caccccatca 4332 cggtggccct gagggctgac ccggaggcca gtggagctgc ctggtgtcca cgggggaggg 4392 ccaaggcctg ctgagctgat tctccagctg ctgccccagc ctttccgcct tgcacagcac 4452 agaggtggtc accccaggga cagccaggca cctgctcctc ttgcccttcc tgggggaaag 4512 gagctgcctt ctgtccctgt aactgctttc cttatggccc aacccggcca ctcagacttg 4572 tttgaagctg cactggcagc ttttttgtct cctttgggta ttcacaacag ccagggactt 4632 gattttgatg tattttaaac cacattaaat aaagagtctg ttgccttaaa aaaaaaaaaa 4692 aaaaa 4697 18 60 DNA Homo sapiens CDS (1)...(60) 18 gtg gac gtg gat gag tac gac gag aac aag ttc gtg gac gag gaa gac 48 Val Asp Val Asp Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp 1 5 10 15 ggc ggc gac ggc 60 Gly Gly Asp Gly 20 19 15 PRT Homo sapiens 19 Glu Glu Glu Glu Asp Asp Asp Glu Asp Glu Asp Glu Glu Asp Asp 1 5 10 15 20 26 PRT Homo sapiens 20 Glu Glu Glu Glu Asp Asp Asp Glu Asp Glu Asp Glu Glu Asp Asp Val 1 5 10 15 Ser Glu Gly Ser Glu Val Pro Glu Ser Asp 20 25 21 11 PRT Homo sapiens 21 Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp 1 5 10 22 10 PRT Homo sapiens 22 Glu Asp Asp Asp Pro Asp Gly Phe Leu Gly 1 5 10 23 30 PRT Oryctolagus cuniculus 23 Val Asp Val Asp Glu Tyr Asp Glu Asn Lys Phe Val Asp Glu Glu Asp 1 5 10 15 Gly Gly Asp Gly Gln Ala Gly Pro Asp Glu Gly Glu Val Asp 20 25 30 24 6 PRT Oryctolagus cuniculus 24 Asp Glu Gly Glu Val Asp 1 5 25 16 PRT Oryctolagus cuniculus 25 Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp Asp Asp Asp 1 5 10 15 26 28 PRT Oryctolagus cuniculus 26 Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp Asp Asp Asp 1 5 10 15 Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp 20 25 27 12 PRT Oryctolagus cuniculus 27 Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp 1 5 10 28 10 PRT Oryctolagus cuniculus 28 Pro Pro Gly Lys Pro Ala Leu Pro Gly Ala 1 5 10 29 15 PRT Oryctolagus cuniculus 29 Glu Asp Gly Val Gln Gly Glu Pro Pro Glu Pro Glu Asp Ala Glu 1 5 10 15 30 45 DNA Homo sapiens 30 gaagaggaag aagatgatga tgaagatgaa gatgaagaag atgat 45 31 78 DNA Homo sapiens 31 gaagaggaag aagatgatga tgaagatgaa gatgaagaag atgatgtgtc agagggctct 60 gaagtgcccg agagtgac 78 32 33 DNA Homo sapiens 32 gtgtcagagg gctctgaagt gcccgagagt gac 33 33 30 DNA Homo sapiens 33 gaggatgatg accccgatgg cttcttaggc 30 34 90 DNA Homo sapiens 34 gtggacgtgg atgaatatga cgagaacaag ttcgtggacg aagaagatgg gggcgacggc 60 caggccgggc ccgacgaggg cgaggtggac 90 35 18 DNA Homo sapiens 35 gacgagggcg aggtggac 18 36 48 DNA Homo sapiens 36 gaggaggagg aggaggagga ggaagacgac gaggacgacg acgacgac 48 37 84 DNA Homo sapiens 37 gaggaggagg aggaggagga ggaagacgac gaggacgacg acgacgacgt cgtgtccgag 60 ggctcggagg tgcccgagag cgat 84 38 36 DNA Homo sapiens 38 gtcgtgtccg agggctcgga ggtgcccgag agcgat 36 39 30 DNA Homo sapiens 39 ccccccggga agccagccct cccaggagcc 30 40 45 DNA Homo sapiens 40 gaggatgggg tccagggtga gccccctgaa cctgaagatg cagag 45 41 7 PRT Homo sapiens 41 Arg Asp Val Ser Glu Glu Leu 1 5 42 21 DNA Homo sapiens 42 cgtgatgtct ctgaggagct g 21 43 538 PRT Homo sapiens 43 Met Ala Gly Pro Pro Ala Leu Pro Pro Pro Glu Thr Ala Ala Ala Ala 1 5 10 15 Thr Thr Ala Ala Ala Ala Ser Ser Ser Ala Ala Ser Pro His Tyr Gln 20 25 30 Glu Trp Ile Leu Asp Thr Ile Asp Ser Leu Arg Ser Arg Lys Ala Arg 35 40 45 Pro Asp Leu Glu Arg Ile Cys Arg Met Val Arg Arg Arg His Gly Pro 50 55 60 Glu Pro Glu Arg Thr Arg Ala Glu Leu Glu Lys Leu Ile Gln Gln Arg 65 70 75 80 Ala Val Leu Arg Val Ser Tyr Lys Gly Ser Ile Ser Tyr Arg Asn Ala 85 90 95 Ala Arg Val Gln Pro Pro Arg Arg Gly Ala Thr Pro Pro Ala Pro Pro 100 105 110 Arg Ala Pro Arg Gly Ala Pro Ala Ala Ala Ala Ala Ala Ala Pro Pro 115 120 125 Pro Thr Pro Ala Pro Pro Pro Pro Pro Ala Pro Val Ala Ala Ala Ala 130 135 140 Pro Ala Arg Ala Pro Arg Ala Ala Ala Ala Ala Ala Thr Ala Pro Pro 145 150 155 160 Ser Pro Gly Pro Ala Gln Pro Gly Pro Arg Ala Gln Arg Ala Ala Pro 165 170 175 Leu Ala Ala Pro Pro Pro Ala Pro Ala Ala Pro Pro Ala Val Ala Pro 180 185 190 Pro Ala Gly Pro Arg Arg Ala Pro Pro Pro Ala Val Ala Ala Arg Glu 195 200 205 Pro Pro Leu Pro Pro Pro Pro Gln Pro Pro Ala Pro Pro Gln Gln Gln 210 215 220 Gln Pro Pro Pro Pro Gln Pro Gln Pro Pro Pro Glu Gly Gly Ala Val 225 230 235 240 Arg Ala Gly Gly Ala Ala Arg Pro Val Ser Leu Arg Glu Val Val Arg 245 250 255 Tyr Leu Gly Gly Ser Gly Gly Ala Gly Gly Arg Leu Thr Arg Gly Arg 260 265 270 Val Gln Gly Leu Leu Glu Glu Glu Ala Ala Ala Arg Gly Arg Leu Glu 275 280 285 Arg Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly 290 295 300 Arg Ala Pro Pro Ala Ala Ser Ala Arg Pro Ser Arg Ser Lys Arg Gly 305 310 315 320 Gly Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Asp Asp Asp Glu 325 330 335 Asp Glu Asp Glu Glu Asp Asp Val Ser Glu Gly Ser Glu Val Pro Glu 340 345 350 Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Glu Arg 355 360 365 Gly Pro Gln Ser Ala Lys Glu Arg Val Lys Glu Trp Thr Pro Cys Gly 370 375 380 Pro His Gln Gly Gln Asp Glu Gly Arg Gly Pro Ala Pro Gly Ser Gly 385 390 395 400 Thr Arg Gln Val Phe Ser Met Ala Ala Met Asn Lys Glu Gly Gly Thr 405 410 415 Ala Ser Val Ala Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro 420 425 430 Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys 435 440 445 Pro Pro Gly Arg Lys Glu Lys Pro Ser Asp Pro Val Glu Trp Thr Val 450 455 460 Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala 465 470 475 480 Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met 485 490 495 Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala 500 505 510 Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe 515 520 525 Glu Asp Asp Asp Pro Asp Gly Phe Leu Gly 530 535 44 546 PRT Homo sapiens 44 Met Lys Asn Gln Asp Lys Lys Asn Gly Ala Ala Lys Gln Ser Asn Pro 1 5 10 15 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Pro Glu Gly Ala Gln Glu 20 25 30 Arg Pro Ser Gln Ala Ala Pro Ala Val Glu Ala Glu Gly Pro Gly Ser 35 40 45 Ser Gln Ala Pro Arg Lys Pro Glu Gly Ala Gln Ala Arg Thr Ala Gln 50 55 60 Ser Gly Ala Leu Arg Asp Val Ser Glu Glu Leu Ser Arg Gln Leu Glu 65 70 75 80 Asp Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Gly Pro Gly 85 90 95 Glu Asp Gly Ala Gln Gly Glu Pro Ala Glu Pro Glu Asp Ala Glu Lys 100 105 110 Ser Arg Thr Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Thr Pro Val 115 120 125 Val Asn Gly Glu Lys Glu Pro Ser Lys Gly Asp Pro Asn Thr Glu Glu 130 135 140 Ile Arg Gln Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro Gln 145 150 155 160 Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu Met 165 170 175 Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala Leu 180 185 190 Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln Lys 195 200 205 Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu Lys 210 215 220 Asp His Leu Arg Gly Glu His Ser Lys Ala Val Leu Ala Arg Ser Lys 225 230 235 240 Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu Lys 245 250 255 Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys Glu 260 265 270 Val Thr Ser His Phe Gln Val Thr Leu Asn Asp Ile Gln Leu Gln Met 275 280 285 Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met Glu 290 295 300 Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg Glu 305 310 315 320 Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln Leu 325 330 335 Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala Glu 340 345 350 Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val Glu 355 360 365 Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu Lys 370 375 380 Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn Thr 385 390 395 400 Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met Glu 405 410 415 Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met Tyr 420 425 430 Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala Glu 435 440 445 Glu Lys Thr Val Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys Ile 450 455 460 Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn Asp 465 470 475 480 Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Ser Leu 485 490 495 Thr Asp Ser Gly Pro Glu Arg Arg Pro Glu Gly Pro Gly Ala Gln Ala 500 505 510 Pro Ser Ser Pro Arg Val Thr Glu Ala Pro Cys Tyr Pro Gly Ala Pro 515 520 525 Ser Thr Glu Ala Ser Gly Gln Thr Gly Pro Gln Glu Pro Thr Ser Ala 530 535 540 Arg Ala 545 45 1614 DNA Homo sapiens CDS (1)...(1614) 45 atg gcg ggg ccc ccg gcc cta ccc ccg ccg gag acg gcg gcg gcc gcc 48 Met Ala Gly Pro Pro Ala Leu Pro Pro Pro Glu Thr Ala Ala Ala Ala 1 5 10 15 acc acg gcg gcc gcc gcc tcg tcg tcc gcc gct tcc ccg cac tac caa 96 Thr Thr Ala Ala Ala Ala Ser Ser Ser Ala Ala Ser Pro His Tyr Gln 20 25 30 gag tgg atc ctg gac acc atc gac tcg ctg cgc tcg cgc aag gcg cgg 144 Glu Trp Ile Leu Asp Thr Ile Asp Ser Leu Arg Ser Arg Lys Ala Arg 35 40 45 ccg gac ctg gag cgc atc tgc cgg atg gtg cgg cgg cgg cac ggc ccg 192 Pro Asp Leu Glu Arg Ile Cys Arg Met Val Arg Arg Arg His Gly Pro 50 55 60 gag ccg gag cgc acg cgc gcc gag ctc gag aaa ctg atc cag cag cgc 240 Glu Pro Glu Arg Thr Arg Ala Glu Leu Glu Lys Leu Ile Gln Gln Arg 65 70 75 80 gcc gtg ctc cgg gtc agc tac aag ggg agc atc tcg tac cgc aac gcg 288 Ala Val Leu Arg Val Ser Tyr Lys Gly Ser Ile Ser Tyr Arg Asn Ala 85 90 95 gcg cgc gtc cag ccg ccc cgg cgc gga gcc acc ccg ccg gcc ccg ccg 336 Ala Arg Val Gln Pro Pro Arg Arg Gly Ala Thr Pro Pro Ala Pro Pro 100 105 110 cgc gcc ccc cgc ggg gcc ccc gcc gcc gcc gcc gcc gcc gcg ccg ccg 384 Arg Ala Pro Arg Gly Ala Pro Ala Ala Ala Ala Ala Ala Ala Pro Pro 115 120 125 ccc acg ccc gcc ccg ccg cca ccg ccc gcg ccc gtc gcc gcc gcc gcc 432 Pro Thr Pro Ala Pro Pro Pro Pro Pro Ala Pro Val Ala Ala Ala Ala 130 135 140 ccg gcc cgg gcg ccc cgc gcg gcc gcc gcc gcc gcc aca gcg ccc ccc 480 Pro Ala Arg Ala Pro Arg Ala Ala Ala Ala Ala Ala Thr Ala Pro Pro 145 150 155 160 tcg cct ggc ccc gcg cag ccg ggc ccc cgc gcg cag cgg gcc gcg ccc 528 Ser Pro Gly Pro Ala Gln Pro Gly Pro Arg Ala Gln Arg Ala Ala Pro 165 170 175 ctg gcc gcg ccg ccg ccc gcg cca gcc gct ccc ccg gcg gtg gcg ccc 576 Leu Ala Ala Pro Pro Pro Ala Pro Ala Ala Pro Pro Ala Val Ala Pro 180 185 190 ccg gcc ggc ccg cgc cgc gcc ccc ccg ccc gcc gtc gcc gcc cgg gag 624 Pro Ala Gly Pro Arg Arg Ala Pro Pro Pro Ala Val Ala Ala Arg Glu 195 200 205 ccg ccg ctg ccg ccg ccg cca cag ccg ccg gcg ccg cca cag cag cag 672 Pro Pro Leu Pro Pro Pro Pro Gln Pro Pro Ala Pro Pro Gln Gln Gln 210 215 220 cag ccg ccg ccg ccg cag cca cag ccg ccg ccg gag ggg ggc gcg gtg 720 Gln Pro Pro Pro Pro Gln Pro Gln Pro Pro Pro Glu Gly Gly Ala Val 225 230 235 240 cgg gcc ggc ggc gcg gcg cgg ccc gtg agc ctg cgg gaa gtc gtg cgc 768 Arg Ala Gly Gly Ala Ala Arg Pro Val Ser Leu Arg Glu Val Val Arg 245 250 255 tac ctc ggg ggc agc ggc ggc gcc ggc ggt cgc cta acc cgc ggc cgc 816 Tyr Leu Gly Gly Ser Gly Gly Ala Gly Gly Arg Leu Thr Arg Gly Arg 260 265 270 gtg cag ggg ctg ctg gag gag gag gcg gcg gct cga ggc cgt ctg gag 864 Val Gln Gly Leu Leu Glu Glu Glu Ala Ala Ala Arg Gly Arg Leu Glu 275 280 285 cgc acc cgt ctc gga gcg ctt gcg ctg ccc cgc ggg gac agg ccc gga 912 Arg Thr Arg Leu Gly Ala Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly 290 295 300 cgg gcg ccg ccg gcc gcc agc gcc cgc ccg tct cgc agc aag aga ggt 960 Arg Ala Pro Pro Ala Ala Ser Ala Arg Pro Ser Arg Ser Lys Arg Gly 305 310 315 320 gga gaa gag cga gta ctt gag aaa gaa gag gaa gaa gat gat gat gaa 1008 Gly Glu Glu Arg Val Leu Glu Lys Glu Glu Glu Glu Asp Asp Asp Glu 325 330 335 gat gaa gat gaa gaa gat gat gtg tca gag ggc tct gaa gtg ccc gag 1056 Asp Glu Asp Glu Glu Asp Asp Val Ser Glu Gly Ser Glu Val Pro Glu 340 345 350 agt gac cgt cct gca ggt gcc cag cac cac cag ctt aac ggc gag cgg 1104 Ser Asp Arg Pro Ala Gly Ala Gln His His Gln Leu Asn Gly Glu Arg 355 360 365 gga cct cag agt gcc aag gag agg gtc aag gag tgg acc ccc tgc gga 1152 Gly Pro Gln Ser Ala Lys Glu Arg Val Lys Glu Trp Thr Pro Cys Gly 370 375 380 ccg cac cag ggc cag gat gaa ggg cgg ggg cca gcc ccg ggc agc ggc 1200 Pro His Gln Gly Gln Asp Glu Gly Arg Gly Pro Ala Pro Gly Ser Gly 385 390 395 400 acc cgc cag gtg ttc tcc atg gca gcc atg aac aag gaa ggg gga aca 1248 Thr Arg Gln Val Phe Ser Met Ala Ala Met Asn Lys Glu Gly Gly Thr 405 410 415 gct tct gtt gcc acc ggg cca gac tcc ccg tcc ccc gtg cct ttg ccc 1296 Ala Ser Val Ala Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro 420 425 430 cca ggc aaa cca gcc cta cct ggg gcc gac ggg acc ccc ttt ggc tgt 1344 Pro Gly Lys Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys 435 440 445 ccg ccc ggg cgc aaa gag aag cca tct gat ccc gtc gag tgg acc gtg 1392 Pro Pro Gly Arg Lys Glu Lys Pro Ser Asp Pro Val Glu Trp Thr Val 450 455 460 atg gat gtc gtc gaa tat ttt act gag gct gga ttc ccg gag cag gcg 1440 Met Asp Val Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala 465 470 475 480 aca gct ttc caa gag cag gaa att gat ggc aaa tct ttg ctg ctc atg 1488 Thr Ala Phe Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met 485 490 495 cag cgc aca gat gtg ctc acc ggc ctg tcc atc cgc ctc ggg cca gcc 1536 Gln Arg Thr Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala 500 505 510 ctg aaa atc tac gag cac cac atc aag gtg ctt cag caa ggc cac ttt 1584 Leu Lys Ile Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe 515 520 525 gag gat gat gac ccc gat ggc ttc tta ggc 1614 Glu Asp Asp Asp Pro Asp Gly Phe Leu Gly 530 535 46 1638 DNA Homo sapiens CDS (1)...(1638) 46 atg aag aac caa gac aaa aag aac ggg gct gcc aaa caa tcc aat cca 48 Met Lys Asn Gln Asp Lys Lys Asn Gly Ala Ala Lys Gln Ser Asn Pro 1 5 10 15 aaa agc agc cca gga caa ccg gaa gca gga ccc gag gga gcc cag gag 96 Lys Ser Ser Pro Gly Gln Pro Glu Ala Gly Pro Glu Gly Ala Gln Glu 20 25 30 cgg ccc agc cag gcg gct cct gca gta gaa gca gaa ggt ccc ggc agc 144 Arg Pro Ser Gln Ala Ala Pro Ala Val Glu Ala Glu Gly Pro Gly Ser 35 40 45 agc cag gct cct cgg aag ccg gag ggt gct caa gcc aga acg gct cag 192 Ser Gln Ala Pro Arg Lys Pro Glu Gly Ala Gln Ala Arg Thr Ala Gln 50 55 60 tct ggg gcc ctt cgt gat gtc tct gag gag ctg agc cgc caa ctg gaa 240 Ser Gly Ala Leu Arg Asp Val Ser Glu Glu Leu Ser Arg Gln Leu Glu 65 70 75 80 gac ata ctg agc aca tac tgt gtg gac aat aac cag ggg ggc ccc ggc 288 Asp Ile Leu Ser Thr Tyr Cys Val Asp Asn Asn Gln Gly Gly Pro Gly 85 90 95 gag gat ggg gca cag ggt gag ccg gct gaa ccc gaa gat gca gag aag 336 Glu Asp Gly Ala Gln Gly Glu Pro Ala Glu Pro Glu Asp Ala Glu Lys 100 105 110 tcc cgg acc tat gtg gca agg aat ggg gag cct gaa cca act cca gta 384 Ser Arg Thr Tyr Val Ala Arg Asn Gly Glu Pro Glu Pro Thr Pro Val 115 120 125 gtc aat gga gag aag gaa ccc tcc aag ggg gat cca aac aca gaa gag 432 Val Asn Gly Glu Lys Glu Pro Ser Lys Gly Asp Pro Asn Thr Glu Glu 130 135 140 atc cgg cag agt gac gag gtc gga gac cga gac cat cga agg cca cag 480 Ile Arg Gln Ser Asp Glu Val Gly Asp Arg Asp His Arg Arg Pro Gln 145 150 155 160 gag aag aaa aaa gcc aag ggt ttg ggt aag gag atc acg ttg ctg atg 528 Glu Lys Lys Lys Ala Lys Gly Leu Gly Lys Glu Ile Thr Leu Leu Met 165 170 175 cag aca ttg aat act ctg agt acc cca gag gag aag ctg gct gct ctg 576 Gln Thr Leu Asn Thr Leu Ser Thr Pro Glu Glu Lys Leu Ala Ala Leu 180 185 190 tgc aag aag tat gct gaa ctg ctg gag gag cac cgg aat tca cag aag 624 Cys Lys Lys Tyr Ala Glu Leu Leu Glu Glu His Arg Asn Ser Gln Lys 195 200 205 cag atg aag ctc cta cag aaa aag cag agc cag ctg gtg caa gag aag 672 Gln Met Lys Leu Leu Gln Lys Lys Gln Ser Gln Leu Val Gln Glu Lys 210 215 220 gac cac ctg cgc ggt gag cac agc aag gcc gtc ctg gcc cgc agc aag 720 Asp His Leu Arg Gly Glu His Ser Lys Ala Val Leu Ala Arg Ser Lys 225 230 235 240 ctt gag agc cta tgc cgt gag ctg cag cgg cac aac cgc tcc ctc aag 768 Leu Glu Ser Leu Cys Arg Glu Leu Gln Arg His Asn Arg Ser Leu Lys 245 250 255 gaa gaa ggt gtg cag cgg gcc cgg gag gag gag gag aag cgc aag gag 816 Glu Glu Gly Val Gln Arg Ala Arg Glu Glu Glu Glu Lys Arg Lys Glu 260 265 270 gtg acc tcg cac ttc cag gtg aca ctg aat gac att cag ctg cag atg 864 Val Thr Ser His Phe Gln Val Thr Leu Asn Asp Ile Gln Leu Gln Met 275 280 285 gaa cag cac aat gag cgc aac tcc aag ctg cgc caa gag aac atg gag 912 Glu Gln His Asn Glu Arg Asn Ser Lys Leu Arg Gln Glu Asn Met Glu 290 295 300 ctg gct gag agg ctc aag aag ctg att gag cag tat gag ctg cgc gag 960 Leu Ala Glu Arg Leu Lys Lys Leu Ile Glu Gln Tyr Glu Leu Arg Glu 305 310 315 320 gag cat atc gac aaa gtc ttc aaa cac aag gac cta caa cag cag ctg 1008 Glu His Ile Asp Lys Val Phe Lys His Lys Asp Leu Gln Gln Gln Leu 325 330 335 gtg gat gcc aag ctc cag cag gcc cag gag atg cta aag gag gca gaa 1056 Val Asp Ala Lys Leu Gln Gln Ala Gln Glu Met Leu Lys Glu Ala Glu 340 345 350 gag cgg cac cag cgg gag aag gat ttt ctc ctg aaa gag gca gta gag 1104 Glu Arg His Gln Arg Glu Lys Asp Phe Leu Leu Lys Glu Ala Val Glu 355 360 365 tcc cag agg atg tgt gag ctg atg aag cag caa gag acc cac ctg aag 1152 Ser Gln Arg Met Cys Glu Leu Met Lys Gln Gln Glu Thr His Leu Lys 370 375 380 caa cag ctt gcc cta tac aca gag aag ttt gag gag ttc cag aac aca 1200 Gln Gln Leu Ala Leu Tyr Thr Glu Lys Phe Glu Glu Phe Gln Asn Thr 385 390 395 400 ctt tcc aaa agc agc gag gta ttc acc aca ttc aag cag gag atg gaa 1248 Leu Ser Lys Ser Ser Glu Val Phe Thr Thr Phe Lys Gln Glu Met Glu 405 410 415 aag atg act aag aag atc aag aag ctg gag aaa gaa acc acc atg tac 1296 Lys Met Thr Lys Lys Ile Lys Lys Leu Glu Lys Glu Thr Thr Met Tyr 420 425 430 cgg tcc cgg tgg gag agc agc aac aag gcc ctg ctt gag atg gct gag 1344 Arg Ser Arg Trp Glu Ser Ser Asn Lys Ala Leu Leu Glu Met Ala Glu 435 440 445 gag aaa aca gtc cgg gat aaa gaa ctg gag ggc ctg cag gta aaa atc 1392 Glu Lys Thr Val Arg Asp Lys Glu Leu Glu Gly Leu Gln Val Lys Ile 450 455 460 caa cgg ctg gag aag ctg tgc cgg gca ctg cag aca gag cgc aat gac 1440 Gln Arg Leu Glu Lys Leu Cys Arg Ala Leu Gln Thr Glu Arg Asn Asp 465 470 475 480 ctg aac aag agg gta cag gac ctg agt gct ggt ggc cag ggc tcc ctc 1488 Leu Asn Lys Arg Val Gln Asp Leu Ser Ala Gly Gly Gln Gly Ser Leu 485 490 495 act gac agt ggc cct gag agg agg cca gag ggg cct ggg gct caa gca 1536 Thr Asp Ser Gly Pro Glu Arg Arg Pro Glu Gly Pro Gly Ala Gln Ala 500 505 510 ccc agc tcc ccc agg gtc aca gaa gcg cct tgc tac cca gga gca ccg 1584 Pro Ser Ser Pro Arg Val Thr Glu Ala Pro Cys Tyr Pro Gly Ala Pro 515 520 525 agc aca gaa gca tca ggc cag act ggg cct caa gag ccc acc tcc gcc 1632 Ser Thr Glu Ala Ser Gly Gln Thr Gly Pro Gln Glu Pro Thr Ser Ala 530 535 540 agg gcc 1638 Arg Ala 545 47 550 PRT Oryctolagus cuniculus 47 Met Ala Gly Pro Pro Ala Leu Pro Pro Pro Glu Thr Ala Ala Ala Ala 1 5 10 15 Thr Thr Ala Ala Ala Ala Ala Ser Ser Ser Ala Ala Ser Pro His Tyr 20 25 30 Gln Glu Trp Ile Leu Asp Thr Ile Asp Ser Leu Arg Ser Arg Lys Ala 35 40 45 Arg Pro Asp Leu Glu Arg Ile Cys Arg Met Val Arg Arg Arg His Gly 50 55 60 Pro Glu Pro Glu Arg Thr Arg Ala Glu Leu Glu Lys Leu Ile Gln Gln 65 70 75 80 Arg Ala Val Leu Arg Val Ser Tyr Lys Gly Ser Ile Ser Tyr Arg Asn 85 90 95 Ala Ala Arg Val Gln Pro Pro Arg Arg Gly Ala Thr Pro Pro Ala Pro 100 105 110 Pro Arg Ala Pro Arg Gly Gly Pro Ala Ala Ala Ala Ala Pro Pro Pro 115 120 125 Thr Pro Ala Pro Pro Pro Pro Pro Ala Pro Val Ala Ala Ala Ala Ala 130 135 140 Pro Ala Arg Ala Pro Arg Ala Ala Ala Ala Ala Ala Ala Ala Thr Ala 145 150 155 160 Pro Pro Ser Pro Gly Pro Ala Gln Pro Gly Pro Arg Ala Gln Arg Ala 165 170 175 Ala Pro Leu Ala Ala Pro Pro Pro Ala Pro Ala Ala Pro Pro Ala Ala 180 185 190 Ala Pro Pro Ala Gly Pro Arg Arg Ala Pro Pro Pro Ala Ala Ala Val 195 200 205 Ala Ala Arg Glu Ser Pro Leu Pro Pro Pro Pro Gln Pro Pro Ala Pro 210 215 220 Pro Gln Gln Gln Gln Gln Pro Pro Pro Pro Pro Pro Pro Gln Gln Pro 225 230 235 240 Gln Pro Pro Pro Glu Gly Gly Ala Ala Arg Ala Gly Gly Pro Ala Arg 245 250 255 Pro Val Ser Leu Arg Glu Val Val Arg Tyr Leu Gly Gly Ser Ser Gly 260 265 270 Ala Gly Gly Arg Leu Thr Arg Gly Arg Val Gln Gly Leu Leu Glu Glu 275 280 285 Glu Ala Ala Ala Arg Gly Arg Leu Glu Arg Thr Arg Leu Gly Ala Leu 290 295 300 Ala Leu Pro Arg Gly Asp Arg Pro Gly Arg Ala Pro Pro Ala Ala Ser 305 310 315 320 Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly Glu Glu Arg Val Leu Glu 325 330 335 Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp Asp Asp 340 345 350 Asp Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp Arg Pro Ala 355 360 365 Gly Ala Gln His His Gln Leu Asn Gly Gly Glu Arg Gly Pro Gln Thr 370 375 380 Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu Cys Gly Pro His Pro Gly 385 390 395 400 Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly Ser Gly Thr Arg Gln Val 405 410 415 Phe Ser Met Ala Ala Leu Ser Lys Glu Gly Gly Ser Ala Ser Ser Thr 420 425 430 Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro Gly Lys Pro 435 440 445 Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro Ala Gly Arg 450 455 460 Lys Glu Lys Pro Ala Asp Pro Val Glu Trp Thr Val Met Asp Val Val 465 470 475 480 Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr Ala Phe Gln 485 490 495 Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln Arg Thr Asp 500 505 510 Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu Lys Ile Tyr 515 520 525 Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu Asp Asp Asp 530 535 540 Pro Glu Gly Phe Leu Gly 545 550 48 2561 DNA Oryctolagus cuniculus CDS (246)...(1895) 48 ggtctgtgtg tgcgtgcgtg cgagtgagtg agtgtgtgca tatttttttt tctcttttct 60 ttctctctct tttttttttt tttgcaaaga aacagcagcg ccgccgccgc tccgccgagg 120 cgctgcgccc cccggggggg ggaggcggag gaggcgggca gcggcggagg gaggggagcc 180 ggggaggggg gcgccgcgct gggagggagg cagcgcgcac ggtgcagccg ggccgggcgg 240 gaggc atg gcg ggg ccc ccg gcc cta ccc ccg ccg gag acg gcg gcg gcc 290 Met Ala Gly Pro Pro Ala Leu Pro Pro Pro Glu Thr Ala Ala Ala 1 5 10 15 gcc acc acg gcc gcg gcc gcc gcc tcg tcg tcc gcc gct tcc ccg cac 338 Ala Thr Thr Ala Ala Ala Ala Ala Ser Ser Ser Ala Ala Ser Pro His 20 25 30 tac caa gag tgg att ctg gac acc atc gac tcg ctg cgc tcg cgc aag 386 Tyr Gln Glu Trp Ile Leu Asp Thr Ile Asp Ser Leu Arg Ser Arg Lys 35 40 45 gcg cgg ccg gac ctg gag cgc atc tgc cgg atg gtg cgg cgg cgg cac 434 Ala Arg Pro Asp Leu Glu Arg Ile Cys Arg Met Val Arg Arg Arg His 50 55 60 ggc ccg gag ccg gag cgc acg cgc gcc gag ctc gag aaa ctg atc cag 482 Gly Pro Glu Pro Glu Arg Thr Arg Ala Glu Leu Glu Lys Leu Ile Gln 65 70 75 cag cgc gcc gtg ctc cgg gtc agc tac aag ggg agc atc tcg tac cgc 530 Gln Arg Ala Val Leu Arg Val Ser Tyr Lys Gly Ser Ile Ser Tyr Arg 80 85 90 95 aac gcg gcg cgc gtc cag ccg ccc cgg cgc gga gcc acc ccg ccg gcc 578 Asn Ala Ala Arg Val Gln Pro Pro Arg Arg Gly Ala Thr Pro Pro Ala 100 105 110 ccg ccg cgc gcc ccc cgc ggg ggc ccc gcc gcc gcc gcc gcg ccg ccg 626 Pro Pro Arg Ala Pro Arg Gly Gly Pro Ala Ala Ala Ala Ala Pro Pro 115 120 125 ccc acg ccc gcc ccg ccg ccg ccg ccc gcg ccc gtc gcc gcc gcc gcc 674 Pro Thr Pro Ala Pro Pro Pro Pro Pro Ala Pro Val Ala Ala Ala Ala 130 135 140 gcc ccg gcc cgg gcg ccc cgc gcg gcc gcc gcc gcc gct gcc gcc aca 722 Ala Pro Ala Arg Ala Pro Arg Ala Ala Ala Ala Ala Ala Ala Ala Thr 145 150 155 gcg ccc ccc tcg ccc ggc ccc gcg cag ccg ggc ccc cgc gcg cag cgg 770 Ala Pro Pro Ser Pro Gly Pro Ala Gln Pro Gly Pro Arg Ala Gln Arg 160 165 170 175 gcc gcg ccc ctg gcc gcg ccg ccg ccc gcg ccc gcc gct ccc ccg gcg 818 Ala Ala Pro Leu Ala Ala Pro Pro Pro Ala Pro Ala Ala Pro Pro Ala 180 185 190 gcg gcg ccc ccg gcc ggc ccg cgc cgc gcc ccc ccg ccc gcc gcc gcc 866 Ala Ala Pro Pro Ala Gly Pro Arg Arg Ala Pro Pro Pro Ala Ala Ala 195 200 205 gtc gcc gcc cgg gag tcg ccg ctg ccg ccg ccg cca cag ccg ccg gcg 914 Val Ala Ala Arg Glu Ser Pro Leu Pro Pro Pro Pro Gln Pro Pro Ala 210 215 220 ccg cca cag cag cag cag cag ccg ccg ccg cca ccg ccg ccg cag cag 962 Pro Pro Gln Gln Gln Gln Gln Pro Pro Pro Pro Pro Pro Pro Gln Gln 225 230 235 cca cag ccg ccg ccg gag ggg ggc gcg gcg cgg gcc ggc ggc ccg gcg 1010 Pro Gln Pro Pro Pro Glu Gly Gly Ala Ala Arg Ala Gly Gly Pro Ala 240 245 250 255 cgg ccc gtg agc ctg cgg gaa gtc gtg cgc tac ctc ggg ggt agc agc 1058 Arg Pro Val Ser Leu Arg Glu Val Val Arg Tyr Leu Gly Gly Ser Ser 260 265 270 ggc gct ggc ggc cgc ctg acc cgc ggc cgc gtg cag ggt ctg ctg gaa 1106 Gly Ala Gly Gly Arg Leu Thr Arg Gly Arg Val Gln Gly Leu Leu Glu 275 280 285 gag gag gcg gcg gcg cgg ggc cgc ctg gag cgc acc cgt ctc gga gcg 1154 Glu Glu Ala Ala Ala Arg Gly Arg Leu Glu Arg Thr Arg Leu Gly Ala 290 295 300 ctt gcg ctg ccc cgc ggg gac agg ccc gga cgg gcg cca ccg gcc gcc 1202 Leu Ala Leu Pro Arg Gly Asp Arg Pro Gly Arg Ala Pro Pro Ala Ala 305 310 315 agc gcc cgc gcg gcg cgg aac aag aga gct ggc gag gag cga gtg ctt 1250 Ser Ala Arg Ala Ala Arg Asn Lys Arg Ala Gly Glu Glu Arg Val Leu 320 325 330 335 gaa aag gag gag gag gag gag gag gag gaa gac gac gag gac gac gac 1298 Glu Lys Glu Glu Glu Glu Glu Glu Glu Glu Asp Asp Glu Asp Asp Asp 340 345 350 gac gac gtc gtg tcc gag ggc tcg gag gtg ccc gag agc gat cgt ccc 1346 Asp Asp Val Val Ser Glu Gly Ser Glu Val Pro Glu Ser Asp Arg Pro 355 360 365 gcg ggt gcg cag cat cac cag ctg aat ggc ggc gag cgc ggc ccg cag 1394 Ala Gly Ala Gln His His Gln Leu Asn Gly Gly Glu Arg Gly Pro Gln 370 375 380 acc gcc aag gag cgg gcc aag gag tgg tcg ctg tgt ggc ccc cac cct 1442 Thr Ala Lys Glu Arg Ala Lys Glu Trp Ser Leu Cys Gly Pro His Pro 385 390 395 ggc cag gag gaa ggg cgg ggg ccg gcc gcg ggc agt ggc acc cgc cag 1490 Gly Gln Glu Glu Gly Arg Gly Pro Ala Ala Gly Ser Gly Thr Arg Gln 400 405 410 415 gtg ttc tcc atg gcg gcc ttg agt aag gag ggg gga tca gcc tct tcg 1538 Val Phe Ser Met Ala Ala Leu Ser Lys Glu Gly Gly Ser Ala Ser Ser 420 425 430 acc acc ggg cct gac tcc ccg tcc ccg gtg cct ttg ccc ccc ggg aag 1586 Thr Thr Gly Pro Asp Ser Pro Ser Pro Val Pro Leu Pro Pro Gly Lys 435 440 445 cca gcc ctc cca gga gcc gat ggg acc ccc ttt ggc tgc cct gcc ggg 1634 Pro Ala Leu Pro Gly Ala Asp Gly Thr Pro Phe Gly Cys Pro Ala Gly 450 455 460 cgc aaa gag aag ccg gca gac ccc gtg gag tgg aca gtc atg gac gtc 1682 Arg Lys Glu Lys Pro Ala Asp Pro Val Glu Trp Thr Val Met Asp Val 465 470 475 gtg gag tac ttc acc gag gcg ggc ttc cct gag caa gcc acg gct ttc 1730 Val Glu Tyr Phe Thr Glu Ala Gly Phe Pro Glu Gln Ala Thr Ala Phe 480 485 490 495 cag gag cag gag atc gac ggc aag tcc ctg ctg ctc atg cag cgc acc 1778 Gln Glu Gln Glu Ile Asp Gly Lys Ser Leu Leu Leu Met Gln Arg Thr 500 505 510 gat gtc ctc acc ggc ctg tcc atc cgc ctg ggg cca gcg ttg aaa atc 1826 Asp Val Leu Thr Gly Leu Ser Ile Arg Leu Gly Pro Ala Leu Lys Ile 515 520 525 tat gag cac cat atc aag gtg ctg cag cag ggt cac ttc gag gac gat 1874 Tyr Glu His His Ile Lys Val Leu Gln Gln Gly His Phe Glu Asp Asp 530 535 540 gac ccg gaa ggc ttc ctg gga tgagcacaga gccgccgcgc cccttgtccc 1925 Asp Pro Glu Gly Phe Leu Gly 545 550 cacccccacc ccgcctggac ccattcctgc ctccatgtca cccaaggtgt cccagaggcc 1985 aggagctgga ctgggcaggc gaggggtgcg gacctaccct gattctggta gggggcgggg 2045 ccttgctgtg ctcattgcta cccccccacc ccgtgtgtgt ctctgcacct gcccccagca 2105 cacccctccc ggagcctgga tgtcgcctgg gactctggcc tgctcatttt gcccccagat 2165 cagccccctc cctccctcct gtcccaggac attttttaaa agaaaaaaag gaaaaaaaaa 2225 aattggggag ggggctggga aggtgcccca agatcctcct cggcccaacc aggtgtttat 2285 tcctatatat atatatatat gttttgttct gcctgttttt cgttttttgg tgcgtggcct 2345 ttcttccctc ccaccaccac tcatggcccc agccctgctc gccctgtcgg cgggagcagc 2405 tgggaatggg aggagggtgg gaccttgggt ctgtctccca ccctctctcc cgttggttct 2465 gttgtcgctc cagctggctg tattgctttt taatattgca ccgaagggtt gttttttttt 2525 ttttaaataa aattttaaaa aaaggaaaaa aaaaaa 2561 49 12619 DNA Homo sapiens 49 aagctttata aagatttaac tacctaataa ggtagagaag taatttatgt gcccactaaa 60 aaatactcaa tttctgaatg ttcgtccaaa attaacttgt cagatcatta aatcattgac 120 tagaaacacg ttgagtacct attatgtact aggcacttag atcattgtga gacaataaaa 180 aatactgcat tagaaaagga catttttcac atcttaaatg caataagcat tatttggctg 240 gcagttaatt acatttaaca cattaaacat atagagcaaa attctgagca atcaaaataa 300 ttataccctt gagcaatcga ttatttaaat ttctttcact attcccttaa gctgatttct 360 actctgggat tctttcatag ttctcaaata agaaaataaa aaatttccta aataaggcaa 420 tacaaaagaa tagaaatgta agagaagaga tatattagct cttgaatccc tgtttccatt 480 tgctgtcaat agtgcctcta atgttcgatt ttctcttcaa agaaaaatct tgatttaaaa 540 ggaagaaaaa gtacaatcac ctttaacagc taaagtatac tgattagcat ctactaaagt 600 tagcaaagac tgaaactgaa aaaaaattgt aaaatcttta ttctaagtta tataacgcca 660 ttcaccatag taatgatttt atactttggt atatggcttt ttaaaataaa tattgccaac 720 aggtaaaaat ttttcctttg ctgtcttaag gcattcctaa gagaattttt accagtgtgt 780 gttcataact tgaatgttaa tttaaacaat gttacttcta tcacctaaat gatatactta 840 tagaagagtg gtttaattgg gaacagaaaa acaccacatt gcttcttccc aagaaaaagg 900 gatgtattcc attctcgagg tctctctccc actctctatt tatatataat atactgcata 960 gataaatata cacacattat atatgtattt ttttgaactt aaagaagact ggacatatgt 1020 atttacatgt atatatccaa caaatattta attttgagat ctctctccct cttctgattt 1080 attattctca gtatgaattc tcaaactgta cggtctttca catttcattc attcatcaag 1140 catgtatcga gtcccttctg catgcttagc tttttgtcat atggaaggaa gatacaaaag 1200 aaaaactgtt tctgcccttc agaatctttc catctcttct aggaaggaga taaaacacca 1260 tatatcatta agaaatttat aagactagtc ccaaaaccaa tggtacaagc aacatgcatt 1320 ttacatttat gtagaatttt agagcttgga aacactttcg tgatatataa tcctaagaac 1380 aatcttgtaa agtgcacatt attagctcca tttcagtgat gaggaatctg agacagaatt 1440 ttaagtgaca tgtctcgttc aaacattatg agtggaagag tcaacactta agcctgagtt 1500 ttctgattct aagcctagtg ctcttttcaa cacagcactg gaaaccaaag attgtggtac 1560 acaacaaggc aacagccagt cttcttgctc gaggtccaac taaactggac ccataccgag 1620 cagtgtccag ccaaatgtcc aaattaattt tatcctgcaa atatttgttc ttcagtgtaa 1680 tacacacagc acaactacca tttccttcgt cttagtgcct ttatctccta cattccagaa 1740 atggggatgt caaatatttt tttaaatctg gcctagatgg aatcatataa atctcaaatc 1800 ataatataaa tcttaaaggt ctggtttcca ccaatccttc cacattttgt tttcccccag 1860 cactagagag cctaacctac cctcacccct ttcgagcatt cttgctccaa acgaccacct 1920 attttaagat gtcaatgacc ctttcccaaa ttctacaaat tcaccccagt tttgccaccc 1980 gaccccagcg cctgcccgga cacgttcccc tccctcccaa tagatttgat accgagttca 2040 ggttctgcag atcccgttgc gatgctgtca cacagcactg acagataaga tttgaccttt 2100 cgactccgtc cttggggact tcccgctggc caagaagggt agttccaatc ccaggaaacg 2160 ggcttcctgc tcaggaacgc agcctctagc agcgcacagt ctgaggcaat gtctccggca 2220 attagaacga tgctgggcgc ccgggtgtgc atcactctgc ctcatactcc taccaactgc 2280 agggcactcg gtccggcagc cagtccatcc cacccacacc caagtcccag ccagccggac 2340 cttacgcagg accccgatga taggtcgttg acggctgcag caaaagccaa ggccacctgc 2400 cgctgctgcc catccccgcc aatctgagac cccctagact ggaccgcaga aaagcgtttc 2460 tatgggaacc cccccaccga gaatcacgtg acgcaatcgg acgaccaatc gcttcttacc 2520 tctgcccgcg gtccagcttt tggccctccc tctcgccccc gcctccttcg cccagccccg 2580 ccccttgcct gcggagagcc cgcgcctgcg cgctgtgtcc tgcgcgctcc ttccctcgcg 2640 cgcgctctcc gtggaagagc aggggcagcg tgggaggcgc caagggagcg cgaacctgag 2700 gaggaagaaa cggggctagc gcgcaggccc agaacggtcc gagccgcggc agtcggcgac 2760 gcctcagagc ggaagaggga agtgaatcag gcgccgggta gtgggttgct gggctgggct 2820 tgctgaggta gaggcagcgc caagaagagg cctttgccgc tggtcgggat tgggatgtcg 2880 aagaacacag tgtcgtcggc ccgcttccgg aaggtggacg tggatgaata tgacgagaac 2940 aagttcgtgg acgaagaaga tgggggcgac ggccaggccg ggcccgacga gggcgaggtg 3000 gactcctgcc tgcggcaata tccttgcatt caccgccctc cccaccccag cccagcccag 3060 cccgcccttc tcctgggacc cgggagcctg caggatccgc ggggcaccgg cgcggagctg 3120 cctctcaacc tgcggcttaa cctgtctctt tgggatcgcc cgctctgaga gggcaagggg 3180 gaagcccccg tttcctaccc agtcggcagg agacgcgagg gtcccactct tggaagcctg 3240 ccctaccccg cgcgccttcc acgcccccag attcctcagg ttgcacccga gtgcctgcct 3300 gcctcgggaa ctggtcccgc cgcccgcgcc ctcgcggcgc tggggaaggc ggccccggct 3360 ggtggggaag gctggtgccg accgccttag tttttcttcc tagaactctg atttcctggg 3420 gtcacattag ctccagaaat ttctgattgt ggggaacctg catctttcct tagtggtttt 3480 gttttttggt tgtgtttttg ttattggtag cgttaaggta gtttattgct taccgggggg 3540 ccgggggaga tgggactgtt cgaaaattga gggtccctgt gctttcagcc cattggcctt 3600 tttaaaaaaa aaaaaaaaag aagaagaaga aggggatttg gcaaaatata cattgtacag 3660 aatttgttaa ctgggggagg ggaatgaata caaaaaatac aaaactccta gaaggaagct 3720 tggagccttt tacctgctaa gaaaaggaca atagaaaaaa caacggggaa tgcgtgtgga 3780 gaatccttgg aaatatttaa aataaacccc aatgaataag atagaagatg agtcattcgt 3840 ataaagcaga atcatttttg taatcctaaa attgtttcca ttttagttaa aatatggcag 3900 tcagttcccg gtttctgttt ttgcatattt gaatattcat aactttggct tcgcatttgc 3960 attacatctt ttttagaaaa atgtaaatgt tgcaaaaaaa ccgaagctgt agttttagaa 4020 aatctcagac actgaatttg tatgcatttc taattcttgg gtgtattcat aaggaagact 4080 ctcaacaatg tcctgttata gtggggaaat atgagagtga aaatatttaa tggcaacaat 4140 atcctttttt aaaggcacct aaatagagca ttagacattt atcaatatat agatagtgct 4200 ttgcccaact ttcacaatta attagctgtt gctcttttgc attatttaaa tacttaagtg 4260 cttggagtta taaaaaatga gctaatctac atcaggcatg cttctctaga aatccctgca 4320 gccttgaaaa taacagcttg tcaaccagag attttgtgta agaacttttt ctttagaaaa 4380 taaatggtga acatgcttcc taaaaacatt atttgtgatg ggataagatg gtgttttatg 4440 aaaccccagt gtattttagg taatttgtgg tgacttttaa aaggtactgc tgtatccata 4500 tcagtggatc tgctttttga tcagttcatc ttaaaatata aagatactgt ctcttcttac 4560 cgttacatac agccaggaaa gacagcccta gtggtggggt actagagttg gaggaacaag 4620 tgaactctgt ggttttcctt ttaggggaat gtttgtacat tctgacagtc tgattggcct 4680 tctgtttctc atgcttgcta actcactagt gctttcaaag agagcctgaa tttaataggt 4740 atggtctaac acagtttgaa taacctttgt gaaatatgag agaaaatatc taaagcaaaa 4800 aattaagctg ccacctaagg gacatatgaa ttattacatc ttctgtgatg cctcttttca 4860 tcaatattga gagattgcta atgtgtatca ttcagattgc taatctgcca gcatgttcta 4920 ccagcatttc agataataca gaatatggtt ctagcaaaag tttggtcttt attttttcaa 4980 ttagaatcac aggaaaagac atattttggt tgataatagg ttatttcatt tgggggacta 5040 ataattctga tatatatttt aggatttctt taacaccact ctaggtaatg tttgcatatg 5100 tatctcactg ggaaatgaaa gactatcaag gtgttcactt gatagttaga accaagggtg 5160 aaacagtctt tgctttatta aaaaaaagtc taatgttcta ttttgctttt gatattttgc 5220 ctttgattaa catcctggaa accaacacat tgaatttcca gtattgaaca tagtgaccaa 5280 agtaattttc tttttatatg taaatcaagt cataaagaac cagtggttat aatgctttct 5340 gggggccatc ctttgctgtt acacccttaa cttccatcac aggaaacatg acagctgccc 5400 tacaggcagc tctgaagaac ccccctatca acaccaagag tcaggcagtg aaggtgagtc 5460 gcagactaca acacagtgat ctctgctgat atcttattct tagtaaaatc cttgcagtgc 5520 aaaaaaaaat caatatttta actgtttgct atctttgaca agaagagttt ataatgtagt 5580 ttgataggta aaaatttcac gtgaaaaaat agccctataa tgtagttatg ataatgctgc 5640 atggtaagat acagtaagtt caaacgatag tgaaatcatt tgtgtgtgtt tttagaggag 5700 accactcagg ctgaatttga gcaaaggttt gaaaaataag ttaaaccttt acaaaaataa 5760 acagattgta attgcttttt aaagattttt taaaaccata caaatactaa atacttatta 5820 tagaaagctc agacatatga gaaggttaaa aagatagtgg tttgtggtcc cagcacccag 5880 agataacagt tactactttg gggccttgct gtattgttac agagttccct tttgtttttt 5940 taagaatgaa tttttaaaac gggctttttc agctatatgc aatggtacat gagctttcct 6000 tccccaataa gttaatagcc ttttttaaca cttgtatatg gataagctcc agtgtataca 6060 taactaatct tttgtttata tttagactga cttttttttt cctattgtaa accactgaaa 6120 tcaatatttt ttggtaaatt tttaattgtt ctctttgagt aaattgctag cagtgaatta 6180 ctggatcaaa gaatgcactt ttttttaagg cttttggtat gcagtattgc caaattgccc 6240 ttcagaacag ttgtgcaact tacattctct gcagtctttt actaattctt aacctattta 6300 cgtatttatt taaaatgatg cccatagcat caaccccgtt gtccatagct attcatacat 6360 cctaggagct tcaagaatct caattgaata gtagtaagta ataacttagg taaatgcata 6420 ataattatct aggtaacata attttttatt ggggaaaatt tctttggttt ttacaagttg 6480 taaagattgt cgttgaaatt tcatttttac cgtggatgca aagatatttt tctaaatctg 6540 gtaattgcag tctttaaacc aaagataaca gtaggtggta gaaacattct gtgaaatcct 6600 gaccagtagg aatgctggag gtatcacttt gtgttgaatg gaaggagaaa cgaattgttg 6660 aaaaggtcag ttaagtgttt cctttgcttg gccggatggg taagaaaata actgcttttg 6720 aagcaggctt ttgccaaaga aaaaagatca ttattaatga acatcactat atttcatatc 6780 tacagtcaat tcatataaat tacagtcaat tttcttttaa gacagcttgg tttattaaaa 6840 tttttaaata aaaaagtttt taagaaaaaa ttacttctga aggataattc aaggtgaaac 6900 tgcaaatctg cctccttgtt ttgttgggaa tttttttttt tttttttttt ttttgagacg 6960 gagtctcact ctatcaccca ggttggagtg cagtggtgca atctcaactc actgcaccct 7020 ccgcctcccg ggtttaagca atcctcctgc ttcagcctcc cgagtagctg ggatcacagg 7080 cacacaccac catgcctgga taatttctgt atttttagaa gaaaacaggg ttttaccatt 7140 ttggccaggc tggtctcgaa ctcctgacct caggtgatct gcccatctcg gcctcccaaa 7200 gtgctgggat tacagctgtg ggccaccaca cccggccgtt ttgttgggat tttttttttt 7260 taagatcaag acataaattt aaatgttgtt ttaataaatt gttaaattat cacattgatc 7320 tgttagcaaa tcctctcagc tctgccttca attatgttaa tagtctgtca agtttcttac 7380 cacctccact gctactatgc ttaccacatc cagcctgtat tattgcaatt gcctcctaat 7440 tgctctccct gcttctacct tatcccctac tcccacagct tattttctgt aacatagatg 7500 ccaaagcaat cctgttaaaa tgtgagtcag attatggcac tgctcttaaa accttccaat 7560 gtcttctcat ttctctcagt aaaagccaaa ctccttacaa tgcctgtagg ccttacacga 7620 tctgtcctcc cataacctct gacttactca cgtgcttttc tcccaccaat ccactccaac 7680 cacattgggt ttttttctgt tcctggaaca cactgaacac acactaatag cactgttctt 7740 tcctctgtct gaaacacttt cctcagttat cccaagcctt ctttcacgtc cttcaggtcc 7800 ttactcaaat gtcacattca tagtgtagac tttctgaaat tctaaaccct cctcatacag 7860 atatgtctaa atgttctgtt atttattgac ccaccaggac cgggcaggca gcattgtctt 7920 gaaggtgctc atctctttta aagctaatga tatagaaaag gcagttcaat ctctggacaa 7980 gaatggtgtg gatctcctaa tgaagtatat ttataaagga tttgagagcc cgtctgacaa 8040 tagcagtgct atgttactgc aatggcatga aaaggtaagt tatgaattat aaatctatat 8100 gactggttct tttacaatag ggaatgacaa tgacaacctc tctcacctaa ataaccattt 8160 tgatttgttg tacatttttg ttattacaaa taaaatgcat gaaaaggata gttcatattt 8220 atgtttacta gccttggtct taagagattc tgattccaac acttgtgttt attcaacaat 8280 gattattagt aattaaacat aatcttgaac tctgaattaa atcaaaactt tgtaaaagaa 8340 aataagcaat acaaatcaag aattctttca cagtgaccaa aaggtgaaaa caacacaagg 8400 atcgaatatg attcaaccat taaaaggaat gacattctga cacatgctat aacattaata 8460 aaccttgaaa acataccaag tgaaatgagc caaacacaaa agaactaata ttttataatt 8520 ttacttatat gaaataatct aggataggca aacacaaagg gacagaaagt ccttagaggt 8580 tactaggaag tagggaaagc aaggaatagg gagttagtgc ttaataggta cagagttcct 8640 ccttggagtg gtaaaaaagt tttggaaaca gatagtggtg atggctacag tacattgtga 8700 atataattaa tgccaatgga ttttacactt aaagatggtt aaaatggcaa attttgtgtt 8760 agatatttta caactttttt aaagaattag gagtttggag gatcaagaat tcttaaatca 8820 tgtttttcta ttttcatgtg tatattttgc aatgtaagta gatgctggta catcatctgt 8880 caaaagagta taagtgattt tgagctttgg gtaaaaaact ggataacatg taaatagaac 8940 cagtcataaa aatattgagt gtttgaagtg tatctgagtg aaaacacaaa cataagaaaa 9000 aagcacatag taaaacaata gttccccctt ttactctaaa atgcaccaat ttgggtagta 9060 atttatatgg caccctattc atggaacact ttctgttgcc aggtaccata ctattaatgt 9120 tttatttaac ctttacaaca accctgtgga agtatataaa tatctttatc atcctcaatt 9180 tacagatgaa aagctagctt taaaacccaa gccagcgtag ttctagcata gcctcaagat 9240 tgcagtgaac attgattact tattatattc cacatattct tcaaaggact ttataaatat 9300 taactcattt aatcctcata aaaatggagg gaaatgcttg ctattattcc tcttttgtca 9360 ctgaggaaac tgaggcatgt gtgaagtctt catttcttcc aaatgtcagt caccagtttt 9420 taccaatctt cgaagtattt ctgaaatcta tctgttcaag cgtatctaat gcagctgttc 9480 acagcatctc tcccagtctg ttgccatagc ttcctgactg gtttcccagt taacagtttt 9540 gcctccttca aatctgttct ccacccagcc atcaaaatga tatctttaaa atcaaaattg 9600 cccttgtcag tcacctgcag ggataaagtc aaagttccca agtctagctt catcttccat 9660 gtcattcttc ccctcaggct atagcaatgc cagccttttt cctgaatgca ccatattgtt 9720 tcacacctcc atacatttgc tcatgatttt ctggtgttag cctgtcacct actcattctt 9780 ttaatgtgtc atttcctcca tgaagcctta gctgaaacat tcctctatac tgttaatctg 9840 ggtataagcc tctccctggt gctttaatag cacctgcagc acaactctca tttcatacat 9900 tagattaaaa ttacctgttt atatgtctgt ctcctcatgc tagaccagaa aatgctgtat 9960 ttgttcactt ttgtatcccc agcatctagc acagtactca gtatacaaag gtattccata 10020 aatatttttt gaacagaaag aaaccagagc tcagattcct aatacttgat cattactctc 10080 tatttttcaa attagagtca gagttaaagt ttctaagttc ttagctatta aacaatacct 10140 tctttctttg ggagaaaaaa aatctgacaa aggctgacta atcgaagtgg aagttgggat 10200 ggttgatccc agtttgaatt ttcttctgac tatgtggtga gaatgagaaa tgcagaatgt 10260 ccacctgttt tgagcaggaa cactatgctg cagatttttt tttttttttt tttttttttt 10320 ttttgagacg gagtcttgct ctgtcgccca ggctggagtg cagtggcgca atctcggctc 10380 actgcaagct ccgcctcctg ggttcacacc attgtcctgc ctcagcctcc cgagtagctg 10440 ggactacagg cacccgccac cacgcccggc taattttttg tatttttagt agagacgggg 10500 tttcaccatg ttagccagga tggtcttgat ctcctgacct cgtgatccgc cggcctcggc 10560 ctcccaaagt gctgggatta caggcgtgag ccaccgcgcc cggcctatgc tgcagatttt 10620 ttaaaacatt atttagaatt aatgtactaa aatgtaaact agtatctcac tagaatgtaa 10680 cttcatgagg gcagggactt tcaaggtttt gtttattact gtaacctcag tgccaagaac 10740 agtacctggt gcataattgg tgctcaagaa tttattattt gttaactaat aaattcaggg 10800 tctatagcag tgcccattcc ttctttaaga aaaatgtttt accaaatatg agaattgacc 10860 ttttattatt ctgtcaacat ttacatcctg gtttgttttt aggcacttgc tgctggagga 10920 gtagggtcca ttgttcgtgt cttgactgca agaaaaactg tgtagtctgg caggaagtgg 10980 attatctgcc tcgggagtgg gaattgctgg tacaaagacc aaaacaacca aatgccaccg 11040 ctgccctgtg ggtagcatct gtttctctca gctttgcctt cttgcttttt catatctgta 11100 aagaaaaaaa ttacatatca gttgtccttt aatgaaaatt gggataatat agaagaaatt 11160 gtgttaaaat agaagtgttt catcctttca aaaccatttc agtgatgttt ataccaatct 11220 gtatatagta taatttacat tcaagtttaa ttgtgcaact tttaacccct gttggctggt 11280 tttttgttct gttttgtttt gtattatttt taactaatac tgagagattt ggtcagaatt 11340 tgaggccagt ttcctagctc attgctagtc agggaaatga tatttataaa aaatatgaga 11400 gactggcagc tattaacatt gcaaaactgg accatatttc ccttatttaa taagcaaaat 11460 atgtttttgg aataagtggt gggtgaatac cactgctaag ttatagcttt gtttttgctt 11520 gcctcctgat tatctgtact gtgggtttaa gtatgctact ttctctcagc atccaataat 11580 catggcccct caatttattt gtggtcaccc agggttcaga gcaagaagtc ttgctttata 11640 caaatgtatc cataaaatat cagagcttgt tgggcatgaa catcaaactt ttgttccact 11700 aatatggctc tgtttggaaa aaactgcaaa tcagaaagaa tgatttgcag aaagaaagaa 11760 aaactatggt gtaatttaaa ctctgggcag cctctgaatg aaatgctact ttctttagaa 11820 atataatagc tgccttagac attatgaggt atacaactag tatttaagat accatttaat 11880 atgccccgta aatgtcttca gtgttcttca gggtagttgg gatctcaaaa gatttggttc 11940 agatccaaac aaatacacat tctgtgtttt agctcagtgt tttctaaaaa aagaaactgc 12000 cacacagcaa aaaattgttt actttgttgg acaaaccaaa tcagttctca aaaaatgacc 12060 ggtgcttata aaaagttata aatatcgagt agctctaaaa caaaccacct gaccaagagg 12120 gaagtgagct tgtgcttagt atttacattg gatgccagtt ttgtaatcac tgacttatgt 12180 gcaaactggt gcagaaattc tataaactct ttgctgtttt tgatacctgc tttttgtttc 12240 attttgtttt gttttgtaaa aatgataaaa cttcagaaaa taaaatgtca gtgttgaata 12300 atttattttt ctctgacact ttaacaatta tgaatgtatg gttaattaag aggaaaggtt 12360 ttctgcttct accaccaagt actgtactct taacaagaac agtttggtag ggtttttata 12420 agactatata gatataagat gatagagaag agagtcatga atgatgtcag agcactactg 12480 aagcctttgg agtgattcca tagccttctg gatggcagct gaatacctat atgtagtatc 12540 actgcccaaa gacctagact agaaagtgca aagtagctta gcagctgcag tcattcactc 12600 ccagcctcca aaattctct 12619 50 12425 DNA Homo sapiens 50 gatccctctc caggtggaag ctcccttcat accaaagttt aaaggccctg gggatacgag 60 taactttgac gactatgagg aagaagaaat ccgggtctcc atcaatgaga agtgtggcaa 120 ggagttttct gagttttagg ggcatgcctg tgcccccatg ggttttcttt tttctttttt 180 cttttttttg gtcggggggg tgggagggtt ggattgaaca gccagagggc cccagagttc 240 cttgcatcta atttcacccc caccccaccc tccagggtta gggggagcag gaagcccaga 300 taatcagagg gacagaaaca ccagctgctc cccctcatcc ccttcaccct cctgccccct 360 ctcccacttt tcccttcctc tttccccaca gccccccagc ccctcagccc tcccagccca 420 cttctgcctg ttttaaacga gtttctcaac tccagtcaga ccaggtcttg ctggtgtatc 480 cagggacagg gtatggaaag aggggctcac gcttaactcc agcccccacc cacaccccca 540 tcccacccaa ccacaggccc cacttgctaa gggcaaatga acgaagcgcc aaccttcctt 600 tcggagtaat cctgcctggg aaggagagat ttttagtgac atgttcagtg ggttgcttgc 660 tagaattttt ttaaaaaaac aacaatttaa aatcttattt aagttccacc agtgcctccc 720 tccctccttc ctctactccc acccctccca tgtcccccca ttcctcaaat ccattttaaa 780 gagaagcaga ctgactttgg aaagggaggc gctggggttt gaacctcccc gctgctaatc 840 tcccctgggc ccctccccgg ggaatcctct ctgccaatcc tgcgagggtc taggcccctt 900 taggaagcct ccgctctctt tttccccaac agacctgtct tcacccttgg gctttgaaag 960 ccagacaaag cagctgcccc tctccctgcc aaagaggagt catcccccaa aaagacagag 1020 ggggagcccc aagcccaagt ctttcctccc agcagcgttt ccccccaact ccttaatttt 1080 attctccgct agattttaac gtccagcctt ccctcagctg agtggggagg gcatccctgc 1140 aaaagggaac agaagaggcc aagtcccccc aagccacggc ccggggttca aggctagagc 1200 tgctggggag gggctgcctg ttttactcac ccaccagctt ccgcctcccc catcctgggc 1260 gcccctcctc cagcttagct gtcagctgtc catcacctct cccccacttt ctcatttgtg 1320 cttttttctc tcgtaataga aaagtgggga gccgctgggg agccacccca ttcatccccg 1380 tatttccccc tctcataact tctccccatc ccaggaggag ttctcaggcc tggggtgggg 1440 ccccgggtgg gtgcgggggc gattcaacct gtgtgctgcg aaggacgaga cttcctcttg 1500 aacagtgtgc tgttgtaaac atatttgaaa actattacca ataaagtttt gtttaaaaaa 1560 aaagtgtcgc tggtgttctc gacttcgatc acccacccac acacccccag ggggttggaa 1620 agggaatttc ggaccccagc gtgcaggccg atcaggtcct ggcttgaagt ccttgtaacc 1680 agggtttagc tgaaattccg gcactccttc ggccccgcag gagaaacgag cgtcaaactg 1740 ccctttgacc ccagattcgg ggtccccaaa tctgcggcgc gccccctcgg cgtccagccc 1800 gggaccgaga gggcgctcta gggaggcgct ggggctggcg cgccaggagg ccgagcggcg 1860 gcgggggcgg ccctggcagg gggagtagaa gggggagagg gtgcgcgccc cccttcccgc 1920 atcctcagcg ccgggccagg cgcgcctgag ggacgcgggg gcggcggcag caggagggtc 1980 cccgcagcac cctgcgagcg cggcagcccc ggcccgcggg cggcgagttc ccggtaagtg 2040 cggtcccgag agcggagcgc gctggagagg cgtggagagg ggggctgggc gccggggacg 2100 tctgggtccc gcgcccaatg gctggagggc ggccgagcgc cgcccgcccg ccctgcccgc 2160 cccctctccc ctccccccgg cactcccctc cccctccccc gcccgccgct ttcccccgcc 2220 cccgccccgg cgccaactcc gcggcgcctc cttaaaaagc gcgcgggagt tgtaaggggg 2280 ggccggagcg agccggagtg agcgagagcg cagggtaaag ggggcgggcg gggggcccgg 2340 gctccacctt aaaagcgggc gcgtgggggt gggagggagg aaggcgggcg gcggggagga 2400 gggagggagg gaaggaaggg gggccggagt gtcccgggcg cagggcgcgc gtgcggcggc 2460 ggcggcggcg gggaggggcc ggccgcgccg cgctcccctc ctccccctcg catccccggc 2520 cccgcgcgcg cccagcagaa gcgggtctgt gtgtgcgtgc gtgcgagtga gtgagtgtgt 2580 gcatattttt ttctctcttt tctttctctc tcactgtttt ttcctctctc tctctctccc 2640 tctctctctc tttttttttt tttttttttt gcaaagaaac agcagcgccg ccgccgctcc 2700 gccgaggcgc tgcgcccccc ggggggggag gcggaggagg cgggcagcgg cggagggagg 2760 ggagccgggg aggggggcgc cgcgctggga gggaggcagc gcgcacggtg cagccgggcc 2820 gggcgggagg catggcgggg cccccggccc tacccccgcc ggagacggcg gcggccgcca 2880 ccacggcggc cgccgcctcg tcgtccgccg cttccccgca ctaccaagag tggatcctgg 2940 acaccatcga ctcgctgcgc tcgcgcaagg cgcggccgga cctggagcgc atctgccgga 3000 tggtgcggcg gcggcacggc ccggagccgg agcgcacgcg cgccgagctc gagaaactga 3060 tccagcagcg cgccgtgctc cgggtcagct acaaggggag catctcgtac cgcaacgcgg 3120 cgcgcgtcca gccgccccgg cgcggagcca ccccgccggc cccgccgcgc gccccccgcg 3180 gggcccccgc cgccgccgcc gccgccgcgc cgccgcccac gcccgccccg ccgccaccgc 3240 ccgcgcccgt cgccgccgcc gccccggccc gggcgccccg cgcggccgcc gccgccgcca 3300 cagcgccccc ctcgcctggc cccgcgcagc cgggcccccg cgcgcagcgg gccgcgcccc 3360 tggccgcgcc gccgcccgcg ccagccgctc ccccggcggt ggcgcccccg gccggcccgc 3420 gccgcgcccc cccgcccgcc gtcgccgccc gggagccgcc gctgccgccg ccgccacagc 3480 cgccggcgcc gccacagcag cagcagccgc cgccgccgca gccacagccg ccgccggagg 3540 ggggcgcggt gcgggccggc ggcgcggcgc ggcccgtgag cctgcgggaa gtcgtgcgct 3600 acctcggggg cagcggcggc gccggcggtc gcctaacccg cggccgcgtg caggggctgc 3660 tggaggagga ggcggcggct cgaggccgtc tggagcgcac ccgtctcgga gcgcttgcgc 3720 tgccccgcgg ggacaggccc ggacgggcgc cgccggccgc cagcgcccgc ccgtctcgca 3780 gcaaggtgag cgcgccgggg agcgggggcg ccgcgcggtg ggcaggtgcg ggcgaagttg 3840 gtggcggggg cgcgagtccc gggaggaact gggtggcggg tggctggggc tttgcgcgcg 3900 tttcctgcgg gctcggtgcg tggtgacctt ggcaagtgat tgaatctccc ggagcctcag 3960 tttcctccgc tgtaaacgcg gtttaataac agtagcgacc ccttggggtt gttgagcgag 4020 tttagtaaga tttggttgtc gagggcttta gttaacacag agcctggcac ggagtgaatg 4080 cgtaaaagtt agtccgtatt gttcttaaag gtggaatcgg ttcctcctcc ccaccgcccg 4140 gacgccacag tcagggtctg ggattagaac agctactaat tttgcatgct tctctcctcg 4200 gctccagaga ggtggagaag agcgagtact tgagaaagaa gaggaagaag atgatgatga 4260 agatgaagat gaagaagatg atgtgtcaga gggctctgaa gtgcccgaga gtgaccgtcc 4320 tgcaggtgcc cagcaccacc agcttaacgg cgagcgggga cctcagagtg ccaaggagag 4380 ggtcaaggag tggaccccct gcggaccgca ccagggccag gatgaagggc gggggccagc 4440 cccgggcagc ggcacccgcc aggtgttctc catggcagcc atgaacaagg aagggggaac 4500 aggtaaggat ccctctgggt ggggaagagt gctaggtgga gaggaactca gcccgaagac 4560 aaagccaaag acaggtgttt ttttccttcc cagcttctgt tgccaccggg ccagactccc 4620 cgtcccccgt gcctttgccc ccaggcaaac cagccctacc tggggccgac gggaccccct 4680 ttggctgtcc gtaagttggg gtattggaga catgggggtg ctgctcaggt gtgtggtaca 4740 gccagagaga catccgtgtt cactggtgtc tgtttgtttt gatgcagtcc cgggcgcaaa 4800 gagaagccat ctgatcccgt cgagtggacc gtgatggatg tcgtcgaata ttttactgag 4860 gctggattcc cggagcaggc gacagctttc caagagcagg tgagtttcca gcccaggact 4920 acacactgac agacacagag ggcctccctg ggatgtgccc tgatcccggc tttctctgtt 4980 cctgtcccac ccaggaaatt gatggcaaat ctttgctgct catgcagcgc acagatgtgc 5040 tcaccggcct gtccatccgc ctcgggccag ccctgaaaat ctacgagcac cacatcaagg 5100 tgcttcagca aggccacttt gaggatgatg accccgatgg cttcttaggc tgagcgccca 5160 gcctcacccc tgccccagcc cattccggcc cccatctcac ccaagatccc ccagagtcca 5220 ggagctggac ggggacaccc tcagccctca taacagattc caaggagagg gcaccctctt 5280 gtccttatct ttgccccttg tgtctgtctc acacacatct gctcctcagc acgtcggtgt 5340 ggggagggga ttgctcctta aaccccaggt ggctgaccct ccccacccag tccaggacat 5400 tttaggaaaa aaaaaatgaa atgtgggggg cttctcatct ccccaagatc ctcttccgtt 5460 cagccagatg tttcctgtat aaatgtttgg atctgcctgt ttattttggt gggtggtctt 5520 tcctccctcc cctaccaccc atgcccccct tctcagtctg cccctggcct ccagccccta 5580 ggggactagc tgggttgggg ttcctcgggc cttttctctc ctcccttttt ctttctgttg 5640 attgtcgctc cagctggctg tattgctttt taatattgca ccgaaggttt tttaaataaa 5700 attttaaaaa aagaaaaagg gaaaaaaaag ccacggagtc cattttatga atggggtggg 5760 gagagggcac taaagagcct cctaagagag cctcaggtta ggacagaatt gtttggggag 5820 ggagaaaaac agaaacaatg aattatagct gcctcacagc catgtataac aataattgct 5880 ccaggaaggt gggaatattt gctttttttt cttctgtaat ctcaccgtgt ccgtgtccag 5940 aacagagcta ggcacacagc aggtgctcaa tttttgtttt tcgtttagac aggtttcatt 6000 ctttcaccca ggctggagtg cagtggtgct atcatagctc attgtagcct caaactcctg 6060 ggctgaagtg atcctcccac ctcagcctcc tgagtagctg ggactacagg tgcactctgc 6120 catgccgggc taacttttaa aaatttttgt ccgggcacag tggctcatgc ctgtaatccc 6180 agcactttgg gaggccgagg tgggtggatc atgaggtcag gagttcaaga tcagcctggc 6240 caagatgatg aaaccctgtc tctactaaaa atataaaaaa aaattagctg ggcgtggtgg 6300 tgggtgcctg taatcctagc tattcaggag gctgaggcag aggattgctt acacctggga 6360 ggcggagggt gcagtgagcc aagatcgtgc cactgcactc cagcctgggt gacaaagtga 6420 gactctgtct caaaaaaaaa tctttgtgtg tgtgtggaga tgagggtatg cactttgttg 6480 gccaggttgg cctcgaactc ccagccaagc aattctgcct gggattacaa gcgtgagcca 6540 ccatgcctgg cctcaaatat tgttgaatgg ctagcagtta agtccttggg tttataagca 6600 tttcctcaac tgtcctccca agtccccata agacaaaaaa ctcataaaat cccaccttac 6660 agaagaggca gctggcccgg cacagagatg ctgtctgccc cgggtcacac agggtggcat 6720 ctgacaccct gtctgagttc ttcactcaga gtctttaaat ataattagcg tatttgacat 6780 aatgtacatt aaaaactata aacctgtcag cctttgtcta ctgcaaagaa tccactacaa 6840 atattggggc agggatctgt tcttggacca tagtagtgtc tccagacctc atggtcctct 6900 tcattaaaac aacagaaaat tccttctggg ccatcagatg agaccatgag atagaagatt 6960 tccaagtgaa gattttgttt caagacagag tcttgctctg tcactcaggc tagagtgtac 7020 tggtgcaatc ataactgtgg tgacagcctc gaacttttgg gtacaagtga ttctcatgcc 7080 tcagacaaca cccaactaat attttggttt ttgtatagac agggtcttgc tatgtggctt 7140 aggctggtct tgaactcctg gcctcaagca gtcctcccgc ttcagcctcc taaagtgtca 7200 ggattacaga catgagccac caagtccagc ctgaagattt ttaaaaatta ttgttagtag 7260 tagtcgccag agttactaca tccaaagtcc ctactaagtt ctaagtagtc cctactaagt 7320 tctaaggcag tttctcaact cattagagtt gttttttgtt tttaaagaaa aaaagaggct 7380 gggcacttta ggagaccgac acgggaggat cgcttgagtc caggagtttg agaccaacct 7440 gggcaacatg ggcccccatc tctaaaaatt ttaaattaaa aaaatgtttt aacaacaaaa 7500 agcgttctgg gagtgagggg ctggggcctg ggcggcctca ttccatatac ctgtgccggg 7560 ttgaggggtt ggagacacgt ttagagaccc ctccactcta ggaatccacc tcgagagata 7620 aaggtcccgg ccctagccac acccccagga cacggccaga ggccacctcc ctaggcgggt 7680 ccctccccac cgccaggttc ctggagcgcg tgcggcgcgt gtgcaggggt agggggccgc 7740 aggcgcgcgg actggagagg cgcgcccctc ccgcgtgttg aaattcaaaa gaggcgaacg 7800 gcccccggcg cggcggcgcg gctccggtgg agaggtcaag gcaggggcca gtcggaggct 7860 cccggggcgg ggtcgaaccc gcggccaacc tgagcagcag cggaagctta aagagctcag 7920 gttcccgccc cccggcccta ccatggctac agagcagtgg ttcgaggggt cgctccccct 7980 ggaccctgga gaaacaccgc ctccagacgc cttggaacct gggacgccgc cctgcggaga 8040 cccctccagg tcgacgcccc ctggcaggcc tgggaaccca tctgagccgg atcctgaaga 8100 tgccgagggg cggctggctg aggcccgggc ctccacgtct tcccccaaac ctctggtccc 8160 ccggcctggg ccagcacctc cccgcctatc cctggacact ttgttcagcc ccatcaccca 8220 acagctgcgc tacctactga agaaggcaga tgatttccag agctacttgc tctacaggtg 8280 atgctggaca gggtcccagg tccccatggg taaggagact tggaggggag gcgacaggat 8340 gggtgacaca caccagggtc gcaaaattac aagcgctagg agccagaggg agacagtgga 8400 agaagctagc atattagaat ccagtttaag agaatgagga agactgtaga attgcgggta 8460 ggggatggct gctattactg tcgtggcagg gtgggcctgg ggttgtcaag tctctaggac 8520 tttttctccc agtttttaag tgctgtctta cattttgagc cctgtgctgg ctaaacaaga 8580 cccacctgag ccaaacttgg cctgcaggac atcagtttga gactccaaag gataatgtga 8640 ttcccagacc aggtttccct gtgactctca atttcagtgt ccattggaat ttcctaggag 8700 gctgggttgg gtttgtttgc gtgtttgttt ttgagatgga gtctcactct gtcgcccagg 8760 ctggagtgca gtggtgcaat ctcagctcac tgcaacctcc gcctcccgga ttgaagcaat 8820 tctctgcctc agcctcccga gtagctggga ttacaggcgc ccaccaacat gtgttgcccg 8880 gctaattttt ttcttttctt agtagagaca gagtttcacc atcttggcca gactggtctt 8940 gagctcctga cctcatgatc cacccgcctt ggcctcccaa agtgctggaa ttacagacgt 9000 gagccaccgc gcctacccga ggctgggttt ttttgttttg ttttgttgtt atgtgttttt 9060 ttgaaatgga gtcttgctct gtcacctagg ctggagtgca gtggggcgaa ctcagctcac 9120 tgcaacctcc gcctcccagg ttcgagggat tctcatgagg ctgttttttt ttttttaatg 9180 agacagggtc tcgctctgtc acccaagctg gagtgcaagt ggggcagtca tagctcactg 9240 caccctcgaa ctcctggtct caagcaatct tccacctccc ctcctgggta actgggacta 9300 caggtgccac catgcccagc taattatttt tgtgtagaga tgggttcttg ctatgttgcc 9360 taggcttgtc tggaactcct ggcctcaagc aatcctccag cctcagcctc ccaaaactct 9420 aggattgcag gcgtgagcca ctgtgcccag accctgcagg aagctctggg tcctaagtgt 9480 tgtgacactc aggtgtcagc actttaacaa gtgttccaaa tgggtttgat gcaggtaaac 9540 cagaaagatg ttcagaaaag acctgaaact gggggctttt ctaatgggtc aaagccaggg 9600 atacaggttg ggattgagta gaatggggaa aactgcgggg tggggagggg ttgtgaggga 9660 ttccaggcaa aggccccctt cttccttcag cagagaccaa gtacagaagg agcagctggc 9720 caaggccatg cccaccttct tacagatgtg tgagccctac ttcctgtacc tggaggcagc 9780 cgcgagaagc atacccccca tctatggacc cctgcaggag ctggtccgaa agggggtgtg 9840 tggaggtttc ttagacccca cgcccctttc ttctcgcagc tctgagcctg tggggatggt 9900 ggagggggag gcccactcct cgcaggccag ctgatctcac tgtacccccc tcttgtatgc 9960 agctgttaga gatctcccaa cagctgaccc tgcgcctgga acagctggtc ctcatgtacg 10020 cttcctttgg gttcgtggac ctggaggaga tgaaccccct taggtaaaat ggtaggagac 10080 tcagatgggg ggatgaagga gtccaaggcc cagcctcacc cctccattct ctcatgtctc 10140 gccagcatct cctgtttctt ttgcgggagg ttctccatca gcctgtccca tgaggtctcc 10200 atcttcagat actgtgcccc aaccgcctac actgccagcc gcttcccccg ctacctctat 10260 aagaagatgc gctggcacct ggaagccacc ccagaggccc ctggtcgggg acaagattcc 10320 cttgtggatt agtaagtcct cttacccaaa tcaaagtcct cccctttcta tgatgaatgc 10380 caatatgacc ctccaaaccg tcaccagcaa agtgaaaagt gagccagggc ccgaggcagt 10440 ggctcacgcc tgtaatccca acactttggg aggccgaggc aggaggatca cttgagctca 10500 agagtttgag atcagcctgg gcaagatggc aagaccctgt ctcaacaaca aagaaattcg 10560 ccaggcgtga tggctggcac ctgtagtccc agctacttgg gaggcttagg caggaggagc 10620 acttgagccc aggaatcaag gctacggtga gctgtgattg tgccactgca ctccaccctg 10680 agtggaagca ataatctgtc tcttaaaaaa aaaaaaaagt gaaccaggaa actaaaggct 10740 tttgaaaggc tacctctatt ttcttaaaac ccaccctccc accaaaataa aagttctcat 10800 cttaaaagta ggctggcagg gagaaaaggc cttggagtca cattcctacc tgagaacttc 10860 agggcaactt ctgatgagtt cccacctcaa ctccaaaatt aaagccctca acagaagtag 10920 ctaggaagct gatcacttct aattacagct ccctcccctc ctagctactt tctgtgctat 10980 cgagatactt gggaagacac aggccagagt ccagccaatt cgtgcccaca gatccagaag 11040 ctgtggtcca tcggccgatg ggtgccccta ggaccagccg aggatgacct ttattcatgg 11100 taggagctag ggcaatagca acgtgggcct gggagctgga gggggaggca gaaccccacc 11160 aaagacaatc caccttccca aacactttgc ttcccttagt agtgatagca ttttattgtg 11220 ccctgaaaag cacttcatgc agaccccagt aacaacccat ggagatctat gctattggcc 11280 ccatttaaca aagaaaacag ggtgctcaga gaagttgtta cctgcccaag gacacacagc 11340 tagcagagcg aatggacagg tcaggaccag ttattcagcc tctaggagcc attactaagt 11400 ctctgatcaa caaggaaaca agtttccccc gggggttttt cccacccgca gctgaaacaa 11460 agcctctttc acctgagcct ctcactcaaa gggagggact cccgaggggc agggggcact 11520 caagtccagg cctgtctatc cctggccccc ccaccccagg attttgtgcc cgcaccgctt 11580 ggggactacc agcagctgct gaccatcggc ttcgaggagc ccacgcccac gctggccacc 11640 gacctgctgg tgcagatcct cacgggccag gcaggccagg cccggcctcc gagcgcagcc 11700 gggcctgcgg ggtgggcagc gcaggggtct tgaacctggg gaagagggta ggagctggaa 11760 cttgacagtt ccaaactcca gaataggggg caggggaggg gctcactcgt tctcgcagtg 11820 cagccgggcc tcgccttcca aagggccagg ccgagctgac ctgtctgcac cgagtccggc 11880 ttggccgtgg ggccctgaat gcggacacgt cagttttgtg ttaaataaaa gaaagaaaga 11940 ggtcacaggc tcagcgtccg ctgcgaatgc cgcgcccctc ccccggggga ttgccccacc 12000 cactcgcgtg gccttctggg aaatgtagtc ttttgaaaga agcctggaat tcgccaatag 12060 gcggacgaga gtttggcgca tgcgcatagg cgcacatgaa gcaaaaaggg aagtggtgcc 12120 cgtcaacacc ggaacccaga aaactgcaag tttagggtac cggggaaatt caacgtccac 12180 tggaggaaga gacttaaggc tacgcccact cccatatttt gacccggaag ttatttattt 12240 tagcgtagaa gactactttt cccgacgcgc cccaggaaag tgccctcgat cagtttccta 12300 agggcccgag ttagactttt tttttctctt ccagcttttg ggacttgggg gccggacagg 12360 tcgtcgtctt tcttggggta tccggggtgc ggacaaggtg ggagagccct acggtatcca 12420 agctt 12425 51 22255 DNA Homo sapiens 51 caacatgctt gggaccagaa gtgtttccaa tttgggattt tctcaaattt taccggttga 60 gcttccccaa tctgaaaatc tgaaatccaa catgcacggc tctgaagtct ttcactgagc 120 ctttggggga aatatttaac atcctaacag ccctaaacca acgctcaatt agcacaacag 180 tttacaatct tctctaccca cagcctgatg cgaggctctg ggactagact atttagccaa 240 cagttcttgc aaaattaact gacttataag taaatagtaa tttcaacacc tcactgctaa 300 tgctgtaaca actctgcaga cctagggagc aagtacggtt tgcagagcac tgggaaggct 360 ctgaagtgac ctttgaactg ggcctcaaaa aattttgggt ttggcaaaag tcaaatctct 420 taggcttcaa attccaggca caaggattgt tgggtttgat ttcattatcc agaagcaatg 480 gggatacaga attgtgatct catgtgtagg gaactgtggg ggttttttct actttaaccc 540 cagtgagact ttgtagagtg tggggtagag aaaaggctca tgaatatgcc tgaagcctaa 600 ctcagcacct ttctgaggaa ctgactgcca aaatggtaat ggagagggga aaatatgacc 660 tactttcaca agttaccttg actgcctcag ggaaacctgc tgtggtagtg tttcttctgg 720 gtgaaagacc aggtaattac ctgggtgctg gtctcagact taccagtttt gaatccctgt 780 tttaaccact cactatcgat atgaccttgg ataagttacc taacctttct cttactgtcc 840 ttttccgtaa aatggggata acagatagta gttatttcta tgagtggtta tgagaaccaa 900 gctattagat agcgggaaag cacacagtaa gcgttcaagg aactgctatt gttattaaaa 960 gcctcctttg gaagaaggac attgaggccc agagagagaa cagaacgtcc agccacacag 1020 caaatccgtg atgaagttgg gactggagta tgggtctcct gagtctcagc ccaggactct 1080 atccctcttc ccgagtcctc ggagttcccg gatggagtca catttgttca cggccaggga 1140 ggaaggtttg atggaggcct gcaggaaaca acagccaggc gcaaggcttt gggagttgaa 1200 gcatagcttc tgcgagatag aaacaaggtt gacatgggca ctcgtgcaga atgacgggct 1260 ccttttggac tcccaggact acagtccctt atgcaccttg ggatctgcgg ctagcccctg 1320 cgtaaagagg gacgcgtagt cttttccctg ccccgccctg ccggggcgcc cgcctccgag 1380 gccgccctcg cttcgtcctt cccagcaagc tccgcgccgg cgccggctat tgattggctg 1440 aggcgggagc aggcggctgg ccggcagcag ttactcgggg tttccggtgc gaggccagag 1500 gtggggaagc catcggacgt cggcggtgag gtacgtgcag cggcggccgg tgggcgagac 1560 tatttgagag tgtgcgggcc gggatgttct cggcctgtgg ggaaatcacg ccaactcccc 1620 gcgtgggccg ggggctgtct ggggatatgc gcatgcgcgg gcgtgcctcg cggcttgagg 1680 gcgcgcgggg cgtgggtggc tgcgcgcgcg gggggcgcac gtggggcctg aggggcgggg 1740 gcggtgccgg gagtcccgcc acgtcagtct ccggccctga gccaatcccg cgcccggcct 1800 gccgcgaggg ggccggttgt gccgggaagt ggctccaggg agaagaggcc tcttccctca 1860 cccgctgtgg gagctgcgcc ccgaaagcct gccccggcac gtcgggctct cctgacccgc 1920 caagaccaga gagccgttgg cgccctccgc ccgggcctgc cggtccgttt attttaagaa 1980 gctttgtgcg cctgctgtgg ggatttctga tccaggctgc gaagaatttc gaagtctgga 2040 aaatagcaac tgtgtttgtt tctaaaggat cttctcctga cccagcatcg ctcatcacaa 2100 tgaagaacca agacaaaaag aacggggctg ccaaacaatc caatccaaaa agcagcccag 2160 gacaaccgga agcaggaccc gagggagccc aggagcggcc cagccaggcg gctcctgcag 2220 tagaagcaga aggtcccggc agcagccagg ctcctcggaa gccggagggt gtgtgccagc 2280 tctgcgttgc cagcgggcag ggggaggagc tgtggggtcg gcctcgcttc tggacttaca 2340 ggccgaggcc aggttgtccg ggaggaggag atgtagaatg agaggacagt gctgggggcc 2400 gcggtccccc ctgcgctctg gcgagttggc ggagctgccc cctctaagca caggaacaga 2460 gttctggaga gaagctccga cgggattaag tcaggtggca gccaaacgag gcacccagtc 2520 aggaaatcca ggtcccgtta gaaacacctc agccaccagc agctaactgc ccttcctgtt 2580 tgaggcattt ctagaatgat ctgaatggca agaaatgggt tttgtggggg ggaaggagat 2640 ggactagaag ttgctccgtg ccatccctgt gtgctgatgc tttacatact tttatgatct 2700 aacaaatatg ttcgggtggt agtgagaaat agttgtgtca ttttacaagt aaacagactt 2760 aaagaagtta ggcaacgatt actataattt cttgatttaa aagatgtttc gaatctaaat 2820 tctgacagga actagatttg ctgaatgata ctccattctt gcttctcagt ttccataaaa 2880 aaaaaagtta ggcaacattt aactcaaact gatgagtttg gctgggcctg aaaaatccca 2940 accagtggta taatcgtctt ctttctcact ctacccctca tcctctcctg ctgtaggggc 3000 tcaagccaga acggctcagt ctggggccct tcgtgatgtc tctgaggagc tgagccgcca 3060 actggaagac atactgagca catactgtgt ggacaataac caggggggcc ccggcgagga 3120 tggggcacag ggtgagccgg ctgaacccga agatgcagag aagtcccgga cctatgtggc 3180 aaggaatggg gagcctgaac caactccagt agtcaatgga gagaaggaac cctccaaggg 3240 ggatccaaac acagaagaga tccggcagag tgacgaggtc ggagaccgag accatcgaag 3300 gccacaggag aagaaaaaag ccaagggttt gggtgagcag agggcggctc tttgtgaagc 3360 tggtgaggag agggagtttg gacttgacgt tctctgggcc agtctgttct gccaggattc 3420 aaaggaaaac ggtacttctc agagcagcaa gtcactctag tctaatcaaa gccagggatg 3480 tgggggccac ggcatagaga gatgcaggag ttaccagcac aaagccttct gggttttgga 3540 gcaactggag cttggcatgg gacctgttct ctctttgaga aaatggagac gggaggctag 3600 ggtaggctcc tgtgccagcc agtactacct gctgtgtgac cttgggtgtg tcccttctcc 3660 tctctgggtc ttagtttata tttctcttta cagtaagaaa attagactag gccagagttg 3720 aaaacccaaa tatctgcata agctgggctt ggccatgggg ccacctgaag atggaggctt 3780 tactgcttcc ctgattagtt gctctcacta gccaactgag agcaggcaaa actacaggct 3840 gggtgcagtc aggctttttt tttttttttt tttttttaaa taaagaaaag ccagaaatct 3900 agagttatgt gagaactcta gattttttca tagttagcag ctaaaatggt aagagccaaa 3960 caaaacccat ccgtgggttg gatttggcac acatgcctgc gaattgcagt ctccatgctg 4020 atctcttggg cccttctggg gaggcagagg gaaggctccc tgactcagtc acaggcaatg 4080 gggaataggc agtgacagtc attttacagc agggtatgta tgtttaagag tctaggccgg 4140 ggtgtggtgg ctcacgcctg taattgcagc actttgggag gccgaggcgg gtggatcacc 4200 tgagggtcag gagttcgaga acagcctggc caacatgatg aaatcccgtc tctactaaaa 4260 atacaaaaat tagctggaca tgctggcaca cgcctgtaat cccagctact tgggaggctg 4320 aggcaggaga atggcttgaa cccgggaggc agaggttgca gtgaactgag attgtgccac 4380 tacatccagc ctgggtgaca agagtgaaac tctgtctcaa aaaaaaaaaa aaagaatcta 4440 gaatctaagt cgagtgtcat tatatccatg ttttattcct attccctttt ccccttatgt 4500 atcctcttac tttaaagagg aactttaaaa aatcttaggg acgactaggc agagtggctc 4560 acacctgtaa ctccagcact ttgggaggcc aaggcaggca gattatgagg tcaggagttc 4620 gagaccagcc tggccaacat ggtgaaaccc cagttctact aaagatacaa aaaatcagcc 4680 gggcgtggtg gcacgtgcct ataatcccag atactcggga ggctgaggca ggagaatcac 4740 ttgaacccgt gaggcaaagt tttcagtgag ctgagatcat gccattgcac tccacctggg 4800 tgacagggtg agactccatc tcaaaaaaag aaaaaggaaa aaatcttaac gtcacataca 4860 tggaaagatc atctttttca ccccccaccc ccaactgaga tggagttttg ctcttgtcac 4920 ccaagctgga gtgcactggc gcgatctagc tccctgcaag ctccgcctcc cgggttcaca 4980 ccattctccc tgcctcagcc tcccgagtag ctgggactac aggctcctgc taccatgccc 5040 ggctaatttt tttgtatttt ttttagtaga gacggggttt catctgtgtt agccaggatg 5100 gttttgatct cctgacctcg tgatccgccc gcctcagcct cccaaagtgc tgggattaca 5160 ggcgtaagcc actgcacccc gccttttttt tttaattaat taattttttt agacagagtc 5220 tcgctctgtc ccaagctgga gtgcagtggc gcgatctggg ctcactgcaa cctccgcctc 5280 ctgggttcac ggcgattctc ctgcctcagc ctcccgagta gctgggacta caggctcctg 5340 ctaccatgcc cggctaattt ttttgtattt tttttagtag agacggggtt tcactgtgtt 5400 agccaggatg gttttgatct cctgacctcg tgatccgccc gcctcagcct cccaaagtcc 5460 gcctcagcct cccaaagtgc tgggattaca ggcgtaagcc actgtaccct gccttttttt 5520 tttaattaat taattttttt agacagagtc tcgctctgtc accaagctgg agtgcagtgg 5580 cgcgatttgg gctcactgca acctccgctt cttgggttca agcgattttc ctacctcagc 5640 ctccggagta actgggacta caggcgcgtg ccaccacacc aagctaattt ttttgtgtat 5700 gtctttagta gagatggggt ttcaccatgt taggatggtc tcgatctctt gacctcgtga 5760 tccgcctgcc tcggcctccc aaagtgctgg gattacaggc atgagccacc ttgcctggcc 5820 gaaagtatct tcattttaaa gttcactgtt tggctactct gttgacaaga gtttagtatt 5880 tctcaaggag gctaagatac ctattccttt ttggatccta cctctatcag gagggtgggc 5940 cttccttgca ttgaaacagt atgaaaacag tagccctgaa ttcataagtg ggacaccttt 6000 cttctattgg tagagcaggc agtttttttc tcctgccaat ggtgcctact aaggagattt 6060 cactagggta cagtcgttca tttgataagc atttgttgag catatcctct gtgatggtac 6120 tatggacagt actggggcta tagtgagggc aggattgagt tggtccttat ggcaaggaag 6180 gcagctaatc aacaagcaaa atataaagta tgatggggag ggctgtcttc agcactcatg 6240 agtgtgagcc caggcctgga ggggacacct ggagaagagg gtgcatgtct ttgctcctgt 6300 gcttttcagg gaaggagatc acgttgctga tgcagacatt gaatactctg agtaccccag 6360 aggagaagct ggctgctctg tgcaagaagt atgctgaact ggtcagttcc cccctccgcg 6420 ggcaccttcc ctgcgttggg aaaatcagca tgccacctgg tgtaaggttg ggggtgcaga 6480 gtcaagtagg tggcttaatt cctgttcagc ttttctctga actatctgtt aaatggggaa 6540 tcacttccag ccagcctctt cagggctgtg cagcaagagg agaaactgca tattccttga 6600 aagaaatttc tcaaagaatg attccaaggt ggtagagccc ttgttcctgg cctgagtcca 6660 agacaccttg tgatcttgat gcttcttcct caaatacaga tgcatagagc cattatcaca 6720 gttaataaaa ctaacactag tcacttgata ctttttcctt ttactccaga gcagtcttct 6780 tgtcactgcc tcctcatatt ccccatgaca ttgactttta acagaaacta gactagctgt 6840 cttgtaggat gcccccttct agctttgtca tctctgtggt atcattttac ttctttacct 6900 cctggtacat gtaagtgaag tagaagttag ctctaaagct tgatccaatt cagcttcaac 6960 tttttgacaa gaattcttca taagtacttc atgttccatc acaataaatg caaagcatgc 7020 tcttcccact ttgttgtaac attgttcagt gggttggggg tggggcagcc agattcttcc 7080 atcatcaggt cccttgtcag aatttgaact aacagattta tccattgatg gtcacagcct 7140 gtgtatgtat gtatgtatgt atgtatgtat gtatttattt atttatttat tttttgagac 7200 ggggtcttgc tctgtcgccc aggctggggt gcagtggcac gatctcggct cgctgcaagc 7260 tccgccttct gggttcatgc cattctcctg cctcagcctc ccgagtagct gggtctacag 7320 gcgcccgcca ccatgctagg ctattttttt tttttttttt ttttttagta gagacggggt 7380 ttcaccgtgt tagccaggat ggtctcgatc tcttgacctc gtgatccgcc cgcctcggcc 7440 tcccaaagtg ctgggattac aggcttgagc caccacgcct ggcctattta tttatttatt 7500 cagagtcaga gtctcgctct gtcaccaggc tggagtgcag tggcgcgatc tcggctcatt 7560 gcaacctcca cctcccaggt tcaagcgagt ctcctgcctc agcctcccga gtagctggga 7620 ttacaggtgc atgtcaccat gcctggctaa attttgtatg ttttagtaga gacagagttt 7680 cagtatgttg gccaggatgg tcttgatctc ttggcctcgt gatccgcccg tctcagcctc 7740 ccaaagtgct gggattacag gtgtgagcca ctgtgcctgg cctctaagta tttattttaa 7800 aattaattca ttccacacac atttattaat attttcctgt aaggaacttt actcatcttt 7860 aaaatgggga atgtcatacc tgcctaatga cattcttgta aggattaaat aaaaggtata 7920 aggaagataa gcaccctttt ggagtgatcc agccagggga aaattgctga tgcaagagag 7980 gaaatgagtt gctagagtgg tgttgtgagt agaggagggg agctgaggcc tgcccaagaa 8040 gggggcttgg ctgtggtaac cacatggcta ggtctgtgtg actggaggag aggacggggc 8100 aggtggactg gtagatgtgc agcttgtgcc cctgattctc tagtttcttc tgtgttttga 8160 gatttgatga gaacgatgaa atagttgtct ggaaggagag gagtgtgaat agcatatgca 8220 ttgtattggg attgctggtc ttcctgaaat tggtggccat gaatttaaag tgagactctt 8280 caagtagggt tgttatagta ctggtgtaaa gcaggaaggt gctttactag ggttgcagta 8340 ctactgggga agggccaaga gagttgaggg tgtaagaaat ccaagccagg taatgtagtt 8400 attttaaagg agagtggaag gatggttgag tcaatggatt ggaggtccta tagggtaaga 8460 gactttctga ggatcacaga tactgattgg aatgagctaa aaagataggt gatggtagtc 8520 ctggactggg atgctggaaa ttgagatagt gggtgtgctc tctggtagtg acaaatctag 8580 atctgcgctg tccaagataa attcgtctct agctaattga catgtggcca gtttgaattt 8640 gaacatgcta taaatgtaag atacacatca gcttttgaag acttaagcaa aaacaaagaa 8700 tataaaacat ctttttgtga gagagtgtct cagtcaccca ggctggagtg cagtggcgtg 8760 atgtcctgct tccaggttca aacgattctc ctgcctcaca gcctcctgga gtaactgaga 8820 ttacaggcgc atgccaccaa actggctact tttttgtatt ttttttttag tagaaacggt 8880 ttcaccatgt tggccaggct ggtcttgaac tcctgacctc aagtgatctg cctgcctcag 8940 cctcccaaag tgctgggatt acaggcatga gccaccactc ccggcctcac ttttttacat 9000 tgattccgtg ttgaaattgt aatgttttgg atattaggtt aaatacatat attactaaaa 9060 ttaatttcac ctgtttttta cttttttagt gcggccagta gaatattttt aattacttat 9120 gtggtttgca ttatatttct gttgtacagg cctggatagg gtcatgggag gggaactgag 9180 ctggggaaag gagtgggttt gtggaagagg tgatggactg tgaggccagg gagttagaag 9240 gattatctgt tgatactgaa gtggccacaa atgagaaaag taattgtgtt ggggagagcg 9300 ctgatgaacg cagcgctaac gttttgaagg aatgcgaggg agcgatgggg gtctgtctgt 9360 taataggcac aaggtacggt agcaggtggt ctcatcctcg ggcatgagtg tccagcaagt 9420 tggggaaatg caacagcttg aagtggctct agtggcccag agtcagagct ggaataggaa 9480 ttggcatctg ctggctgtgt ggcccctgct tgccctagtg agttaccatt tctctgtccc 9540 tacggtggag cctttggggt tattgtgagt tcatgggagg agcgtgtaag caccggcaca 9600 gcatcagccc atgagagtgc tcctggcctg agagggtaag ggtcagggca gctcaggaga 9660 ccctagacct gcatagtgat ccccccacca ggaaggcccc acaagatgct cacctgccct 9720 ccctatccct gtccccagct ggaggagcac cggaattcac agaagcagat gaagctccta 9780 cagaaaaagc agagccagct ggtgcaagag aaggaccacc tgcgcggtga gcacagcaag 9840 gccgtcctgg cccgcagcaa gcttgagagc ctatgccgtg agctgcagcg gcacaaccgc 9900 tccctcaagg taggcctggg ccccctggaa caggtgactc tggtttcctt gacttccact 9960 taatgtttct ttcatgggct ttcctcttaa aaagtagtgc aggctagggc caggcgcagt 10020 ggcacacata agtgattaaa aatcttctgg ccactaaaaa acagaaatta attttagtaa 10080 tatacttaac ccaatatcca aaacattaca atttcaacat gaaatcagtg taaaaaagca 10140 aggctgggtg tggtggctca cacctgtaat cccaacactt tgggaggctg aggtggatgg 10200 atcacttgag gccaggagtt tgagaccaac ctggtcaacg cagtgaaacc ccattctact 10260 aaaaatacaa aaattagccg agtgtgctgg caaatgccta taatcccagc tactcaggtg 10320 gctcaggcat gagaattgct tgcacctggg aggctgaggt tgcagtgagc cgagattgca 10380 tcactgcatt acagcctggg caacagagtg agactcagtg tccaaaaaaa aaaaaaagta 10440 gtgcaggctt gtggcataga aatacacttt ctcaataatg ccttacgtta agagagtact 10500 gcttgtaatc atttgacatg tattagataa ggtgaaggat aaagtactaa gagaatccat 10560 aatgcactgg cgttagtatt tctcaatgaa atgacagtcc cctggtaagc ggaggcctgg 10620 ctctgacaag cagctcttgt cccagacgtt ggtcagtcag gaacctgggt ccttcccatg 10680 ttctgctgct tctatggtga ggtcagtctg tggttacacc aagtttaaat acagcctttt 10740 aactttcttt tttatatgta aaatcttaca tgtagttttt agaatgaaat tattatacat 10800 gtaccatttc atatcctgtg cctttttttc actttacata acatttttcc ctatcagtat 10860 gtgtagggct atcttctcat tatatggata tattatatca gtgccctagt taaagcattt 10920 tgggggttgt ttacaatttt tcattattac atatagaact atagtgaaaa ttcttgttat 10980 atttatcact ggtcagttat atagaactta tctgtaggat aagtcatgga attgaaatgg 11040 ctaggtcaca gtatatgcag atttttcatt ttaatagatt ttgctggatt gccttccagt 11100 gagggggcag tgtgccttcc ccatcaaaag tgttgagtgc ctaattctgc acaactttgc 11160 aaaccctggg tgttactaaa ttttaacagc ttggtctctg ggggtacaga ggggacaaat 11220 gcacattaat ctgaaatctg gaagaatagg ccttaggaga tccgacttgc ttcagaatgg 11280 cacttagcac ttacatgtgt gcatgtgtgc ctgcattttt tcttcctttt tttttttttg 11340 gggacggagt cttgctctgt ggcccatcgc ccaggctgga gtgcagtggc gcgatcatag 11400 ctcaccacaa cctccgcctc ccaggttcaa atgactcctc tgcctcagcc tcccaagcag 11460 ctgggaccac aggtgcacac catcacgccg gctaattttt gtattttagt agaaacgggg 11520 tttcaccata ttggccaggc tggtctcaaa ctcctgacct cgtgatccgc ccacctcagc 11580 ctcccaaagt gctgggatta caggcgtgag ccaccgcgcc tgccatgtgc ctgcattttt 11640 ctagggggag aatctcactt gatgtcacct gatatacaga ggggcccatt ggaacccgca 11700 ttgcacaaca tcctggagtc tggctactcc acgctttggg agcagggagg gctgttggca 11760 gagaccatct gtggactagc tgggggaccc ttgtgaggta gcagtggatg atggctctcg 11820 ggctgacttc tttgcccagg aagaaggtgt gcagcgggcc cgggaggagg aggagaagcg 11880 caaggaggtg acctcgcact tccaggtgac actgaatgac attcagctgc agatggaaca 11940 gcacaatgag cgcaactcca agctgcgcca agagaacatg gagctggctg agaggctcaa 12000 gaagctgatt gagcagtatg agctgcgcga ggaggtaagg gtatcacgga cagcagtcat 12060 ggcccagaaa ttgtgaggtt ttgagtgtgt gctaggcact gggacagtac cttttcaggc 12120 ttcatcccat tctccctttc ttcctcctcc tcctccttgg gaggagagta atgttattcc 12180 tcatagataa aaaacaggtg tggagaagag actcacttac agccacacag ccccaggtcc 12240 acagtgcctt gtcccaaatg actgggccag gcatcttttg gaattagaac tatccacatt 12300 ttagaatgga ggtacatgta tggactgtgt gttatatagc accctcagca gggccttggg 12360 gaagccagac acattaatgt atttatgcag tagaacttcc aaatactcac ctacattatg 12420 ggcttacaat gatgcaggtc aagtctggct gccagcttat gacaatttcc attttcagaa 12480 ctttgtagaa tttggaattg caggggaggg gtgtacctgt gatcagtgat ggactccaga 12540 gactgtgtcc actgattcct tgctgctcct gccactcaaa aggcagaatt tatcaggctg 12600 ggcgtggtgg ctcatgcctg taatcccaac actttgggag gccaaagcgg gcggatcacc 12660 tgaggtcagg agttcaagac cagcctggcc aacatggtga aaccctgtct ctactaaaaa 12720 tacaaaaaat tagccaggtg tggtggtgca cggctgtagt cccagctact caggaggctg 12780 aggcaggaga attgcttgaa cccaggaggc agaggttgca atgagccaag attgtgctac 12840 tgcactctag cctgggtgat ataccgagac tccatctcaa aaaaaaaaaa aaaaaaaagc 12900 aggatgtcac tccctttgtc actgcgttgg ctgccacccc aggcacttga atctttggat 12960 cttccctgcc agtcacctgg ctgttctggg cgcgttctca tcatgagaag ggagacctgc 13020 agccccctta cagggctggc agaggacctg ctctggatta ggccctttcc tagcccctgg 13080 ggtgtggcag tgggtgagac cgggaagatc tgccctctta ggttcatagg ccaaagtgat 13140 gatcgtgtgt gcaggaccta gagggcgctc ccctgaccca cccctttcct tgccatactt 13200 catcctctgg gaacaaagct gcttgtttgg tttgagggga gttggtttgg ttcttatccc 13260 tcagcgctga gacatagagg cttcctgggc cactacagtg agacacgaac ttcaagaatc 13320 tgaatacccc cgttttctct ccccgccaag gcaaaaaagg acttagtact acctgtggag 13380 aaggaggtgc aggactacca ggccctgctg ctttgcattt acagccctcc ccagacagac 13440 acaggcaccc tcatcatacc caaactggac ttacctgcta ggcaccttcc cttccccatc 13500 caaaaaaatg gagttatttt cccttatttc agcaagtcca gttgatttta cctttgaagt 13560 agcacctgag tccttcacct tctctccatc ccttctctct cacctgacac aggtctgcag 13620 cgctcctcta gtaggcagga cagccattcc ttggggatgc acatgtctag tctttgccta 13680 gatatggcaa gtctttgcca actgagctag gctgttatgt tcttagaggc attgtttttg 13740 cccattcttc ccatttacaa gagaatcagg gacacagaag tgagggcttc cagccccata 13800 ggtgatcaat cctggggtca gagatttgag tgtgtttatt gcttgccttc ttgggagcag 13860 attccatcca taaaccatgt gcttaccaag gtctgactca ctgggagaga aacgacgtga 13920 ggttggaaag ctgaccttcc agagacttgg ggcccatgtt gtgtggtaca catgggagtc 13980 catcatatca gattgagatg gggggctggg caaagtgccc tggtctgtgg ctgtggggct 14040 accctgagaa agggagcgcc tgacaagccg actgctccca ccatctttgt tgcagcatat 14100 cgacaaagtc ttcaaacaca aggacctaca acagcagctg gtggatgcca agctccagca 14160 ggcccaggag atgctaaagg aggcagaaga gcggcaccag cgggagaagg attttgtgag 14220 gctcaggccc cagggttggg gtgggggtgt gggaggagac aggctgggct ctggctcagc 14280 tcatagccgg gttatatggg agaagtctgg ccagaccagg cacagattcc ttgagtacca 14340 gtctgagagc aggaagcctc agtgggtctg gtgcttgtgg ctaaaaacca aacatagccc 14400 ctgggggctt ctgacaggat ctggggttct gtcttggaaa tagctcctga aagaggcagt 14460 agagtcccag aggatgtgtg agctgatgaa gcagcaagag acccacctga agcaacaggt 14520 gagagcatat aacctgaccc tgtgccttca agtttccctc actgggcccc atcctggggg 14580 tagtgaaatg ggaccctcat tctaggactg gctgtgtcct ggctgctatg acgccttggt 14640 tgagcttagg tgggctcaga ggacttcatt tgtagctcag aaatgtattg cttttgagga 14700 ggtaggaaca gaagagtttg aaaatcaaca taaaggcaaa ataaaagtca ccctaagtct 14760 cctactttcc aggcttagca ttttggatta tatccttcca aatatatagc tttgctttgt 14820 tttaaggaaa aatagtatct caatagaatt actggtcaga gagtcaagga cgggtctgag 14880 tgtgttgacc agagtgcctc ccagagaaac ccagtcttat ctgtgggctg ctttctcccc 14940 acagcttgcc ctatacacag agaagtttga ggagttccag aacacacttt ccaaaagcag 15000 cgaggtattc accacattca agcaggagat ggaaaaggta actgtggtcc aggccaggca 15060 tggctgctgg ggcataagct gcttcattca aaattgttgg gcctgccttc aggaagctcc 15120 catctggggt gtctcaaggg cagggctgtt aggaaggttc acagcctttc ccctcttgag 15180 gcagtatcag tggtatgtat acactccagg ttgtcccagg gaatggggca gtcttttctg 15240 tttgtttggt ttttttgggg ggtttgttgt tgttgttgtt gttgttgttg ttgtttgaga 15300 tggagactca cctattgccc aggctggagt gcagtggcat gatctcagct cattgcagcc 15360 tttgcccccc gggttcaagt gattctcctg cctcagcctc ctgactagct ggaattacag 15420 gcgcgtgcca ccatgcctgg ctaatttttt ctttcttttt tttttgtatt tttagtagag 15480 acggggtttc accatgttgg ccaggctggt ctcgaactct tggcctcaag tgatctgccc 15540 gccttggcct cccaaagtgc tgggattata ggcgtgagcc accatgcctg gccccttacc 15600 attccttgtt attggtggtg gacacctctg acttcctggt ggtgaggtgg cacagagggc 15660 attgactgca tcctgtaatg ccttgcgcct tgggatcaat cattccccac cttggagaca 15720 caggtgcagt ccccaccttg gagacacaga ccttggagag gccagctctg accatttcct 15780 tctgtctgtc acataaccta gatgactaag aagatcaaga agctggagaa agaaaccacc 15840 atgtaccggt cccggtggga gagcagcaac aaggccctgc ttgagatggc tgaggaggtg 15900 ggctgtctgt gatctgcagc cagggtgggg gtgtgcactt agcgcatatc aggccctttc 15960 ctgtatgttc tacccatcag tgacacagct agcatgaggt agaggtgaga tttgcacaca 16020 atgtccaagt ccaaagttaa tgctgttctc tccccatggg aggtggtgag cccagtggta 16080 ggtctccagt gggagtgaag ggagcaaatg gaagaaagga ataaaagagc agaaaaaaac 16140 gggtgccagt gatgtgcctg gtttacatgt aaagcagccc aggtagtttg tgatttcaca 16200 gcttgtaatg tagaagaaag gaactaacga tggagcagca actgcaagcc agaccttgct 16260 gaaagttttt gggttttttt tgtctttttt gctgctgaat gtttttaggt acgttgttca 16320 ttgaaccttc tcttgagctc tgaggatggt attagtagtc ctgttttata gatgagacag 16380 gctcaaaagt caagtccttt gccaaggtca cgtggtagat aaatggagga atacgttatc 16440 tccaagccgt gccccttttc tgcaccatgc tgccccacct gacagcctag tcatggcttc 16500 aactaggact gtttcctaaa gggggccagc tttggactcg gtctgctctc agccttgtta 16560 aagtgtttgc cgccaagtgg tgatggtaag tgggaggttg atggggcacg gcactgaagg 16620 tctcatttct ttccctagaa aacagtccgg gataaagaac tggagggcct gcaggtaaaa 16680 atccaacggc tggagaagct gtgccgggca ctgcagacag agcgcaatga cctgaacaag 16740 agggtacagg acctgagtgc tggtggccag ggctccctca ctgacagtgg ccctgagagg 16800 aggccagagg ggcctggggc tcaagcaccc agctccccca gggtcacaga agcgccttgc 16860 tacccaggag caccgagcac agaagcatca ggccagactg ggcctcaaga gcccacctcc 16920 gccagggcct agagagcctg gtgttgggtc atgctgggaa gggagcggca gcccagccag 16980 gcctggccca taaaaggctc ccatgctgag cagcccattg ctgaagccag gatgttctga 17040 cctggctggc atctggcact tgcaattttg gattttgtgg gtcagtttta cgtacatagg 17100 gcattttgca aggccttgca aatgcattta tacctgtaag tgtacagtgg gcttgcattg 17160 gggatggggg tgtgtacaga tgaagtcagt ggcttgtctg tgagctgaag agtcttgaga 17220 ggggctgtca tctgtagctg ccatcacagt gagttggcag aagtgacttg agcatttctc 17280 tgtctgattt gaggctcaga cccctccctg cccttcagag ctcaagacaa gtaatacacc 17340 caggtcttga ctgcatttgt cttgtgagca gggcttgctt ggtcagctca ggccctccta 17400 gctgctctgg aggctccttt gattctctag acctggaaaa ggtgtcccta ggcagagccc 17460 tggcagggcg ctcagagctg gggatttgct gcctggaaca agggacctgg agaatgtttt 17520 tgcgtgggat gatgtgctgg tcaggagccc cttgggcatc gcttcccctg ccctttggta 17580 gtgccaggac caggccaatg atgcttctca gtagccttat cattcacagg tgcctctcta 17640 gcctgcacaa atgattgaca agagatcacc caaaggatta tttctgaagg tgtttttttc 17700 tttatttctt tttctttttt tttttttttc tttttctttt ttttttgcac atgacagtgt 17760 ttgtattgag gaccttccaa ggaagaggga tgctgtagca gtggtgcctg ggtgcctggc 17820 ctccagtgtc ccacctcctt caccacccca cttggctcct ttgccatctt gatgctgagg 17880 tttcctgttt ggtgagatca ggttgtttgt ggtaaaagaa aggaaagggc ttctgatggc 17940 tttgccacaa gcttacctgt gggtttcagt cctgagaggc caccaccagt tcccatcagc 18000 actgtctcca tgcagcagtt gctgggtccc atgtccagct gcctctttgg cttcatgggt 18060 ttttctgctt cctgccccca cccccacatg tgcaatcctc aagatttgtc ctgattctat 18120 ttcctggcac ctccctgcct gtccttgggg attctacttc ttcctgtgtg ggagcccata 18180 gctgttgtct aacaggtaag aaatgaaatt gaactattga ctgggcccca gaaatccata 18240 aaatggctgc agacagttgt ttctgtgtcc tgttctaccc ccactccagt acataactac 18300 tatgtactgt gtagagccat tctatatgct gaatgttctg ctgttgcaaa cttgccaggg 18360 tattagccag tgtttgtgcc aagcagtttt ctgggacaac agaatgactc agaccaagat 18420 ggataggatg gttagggctt tgcttcttgc tgtttttctt tgaagctagt tcattgtcct 18480 gcaggtccct tcatcttcca tacctagccc actcttttag cccttacctt aaatctctca 18540 gataagttgg ttcacaaaga atgttaagta ctgaatcatg tgtgactgag accagagatg 18600 gcaaatgaat ggcacaccat ttctccttct cctgccccag ggcaggtacc actgatctgc 18660 atcagagttg cctgctattc tctggtgtat ccttcacatc taggtgccct caagcagctg 18720 tgtgagtgtt gagatctctg ccatctctgg ctgagatact gctgtcctgt gaagtgtttc 18780 ccatgacctt tttcttcccc tttgaatccc tctgtctgga gtagtccttg cctcttcctg 18840 ctccagtagg gccttttccc taccccagcc cctgtgccag gctaagctgg tacaagagct 18900 gccaacctca cagagtgttt gctaggcgag agaggtgcag ggaagaggca gaggtatgca 18960 ccttccccct tgaagagagg ggaaaggcct acagtggccc acataattgc ctgactcaca 19020 cttcagctac ctcttaatgc ctgtggaggg actggagctg ctggatccca gtgtggtggt 19080 gtaggaggcc acagtgagca ggtggcccca gctgggtttc ccaggtcagg aatgtgggcc 19140 ccaggcaagg tgcagccttt gctcacagct ccatccatgt ctagaccttc aggccagtct 19200 gcagatgagg ttccctacct ttttcttctc ttcattgacc aaatcaacca atcactacag 19260 ctgctctgct tctgctttcc aaagtagccc aggtcctggg ccagatgcag gggaggtgcc 19320 tatccatgag tgaaggccag tgtcttcctc acctgggtgg gtcccacact tgtgacctca 19380 gttttaggac caagatctgt gttggtttct tagattgcta gcttttcctc caggggacca 19440 cagcaggtga agctcaagag cgcatggctc tgctaatagt aaattgtttt cagggccttg 19500 tccagctgag agcttcatgt ccaccagatt ctgagaggtg tcagcagcac tttttttttt 19560 tatttgttgt ttgttttcca tgaggttatc ggaccatggg ctgagctcag gcactttctg 19620 taggagactg ttatttctgt aaagatggtt atttaaccct tctcacccca tcacggtggc 19680 cctgagggct gacccggagg ccagtggagc tgcctggtgt ccacggggga gggccaaggc 19740 ctgctgagct gattctccag ctgctgcccc agcctttccg ccttgcacag cacagaggtg 19800 gtcaccccag ggacagccag gcacctgctc ctcttgccct tcctggggga agggagctgc 19860 cttctgtccc tgtaactgct ttccttatgg cccagcccgg ccactcagac ttgtttgaag 19920 ctgcactggc agcttttttg tctcctttgg gtattcacaa cagccaggga cttgattttg 19980 atgtatttta aaccacatta aataaagagt ctgttgcctt acttgtttct ctcctgacct 20040 gtgtattcct ttgtttctgg atctgatcca ttcagcccct tccatcatca ctgacttgtt 20100 caggtctgct gcagagcgcc catggtggtt ccctggtatc ttacatattc cacagtgtct 20160 ttgagcagtc gccacagcct caggatgctg gcatattcac ttgagctgcc tgagtggagc 20220 ccttggcaaa gttggcaaga cccttgcctc agagaggatc acacacacac aaaaaagttt 20280 tccctgacct gggggctcac aggctagtga agggaaaagg tacttttagc tatagacagg 20340 tcaatggtgc tgagagcaga gaggaggccc ctgccccctt cagcaaggtg agggggtgat 20400 acctggaatg gccttctgaa ccacagggca ggtagaagat gaacgtcatt tagtgattaa 20460 atggtacagc tgggaagcag gtccatggga ctgggagagg gggtgaggct gggcccagag 20520 tctgggtacc aggttaagga atgtgggcta gatccagagg gcaggggggg caactgaagg 20580 tgtttcaata ggaaattgat aggctccagc agtaaggcaa aaggcatgga gccaggcata 20640 ggccatttga ggcccaggtt aagaggggtg gacactcatc actgctattt gggtctgagc 20700 tgtgggtagg ctcctatagc cctggcctgc ccaagggaat tcacaggggc ctctaattgt 20760 atgcattcct taaggagagc acattctctg ttcagttttt acacccccca tttacccacc 20820 tcaagcatgg gactcctata tgggagacat gctgctggtg gcctcaccca gcaccctgtt 20880 ctctctgggt cctgggttgg tcaggcacaa aggatgatat gtgctgaatg cccaggaaat 20940 ggcagagaca acccacctgc ccttccctcc aggcctccac aaatagatgt gcccacaatg 21000 actgtgacag tcccagcaga gcctctgacc cttctagctg ggtcctgata catgttttcc 21060 atgctggcca tgttatttct agtcgcagat cctctggagg gtgtgggggg ggtgccgccc 21120 caactcttgg agattccaag caaagcagct ctgagaataa tgaggtttct gaccccccag 21180 tgaagcagct gaggatggga accacagggg tgctccctct gtcagcagca ttaccactgt 21240 ctactctagc agctccggtg gggaaggaga gggatttctg ttgtccccag tctgggcccc 21300 tggttattga aaaagttcgg aattactctt tacccttgtg gagtgttctg agtgttggaa 21360 gtacccagga agaagccctg agcaggtgcc ctcaggagca gtgcccatgg ctccccacat 21420 cagccaagag gcccaacccc aggaagccac tcctgcccgg ggatggggaa ggtgggctgg 21480 gtggctgtgt gcactgccct gggccagctc acttgagcct gctgagccgc ctggccaaac 21540 atgagcctct ctcctgttgt atcagatgct gttctgggga cctgcgccag gagcctctgc 21600 cagggcttta aatagctgcc cccattgatc tggctgcagg cagcagcagt cacactgggt 21660 cagcctccat caggtgctca ggtttccctg aggactggag tcaggtgcca gggaatcgcg 21720 tggtctacct tatgacctgg tgctccccac acctgtctcc taggcctggg gggtggggag 21780 gactcctgtc acttcatctg cggcaaaata cagcccccac cacttaccag agaaaactgt 21840 ctggcattgt agagagaggg gttttgccct caaaagactg ttgcttactt tcagtagaat 21900 ggggaatgac actggtatct tccttaaggg ttgttatggg gatgaaatgt atgtaaagtg 21960 ctcaataggg cactggactc actccattga tggctgtctt tgctcgaagt gtcttcctga 22020 tgctgctgct gttgctgctt gtgcttcttc tgtgcttaca ttctctctct ctcactcact 22080 cactctgtct ctcctctccc ccgccccacc ccctttctga caaagccacc accattttgt 22140 aaggaactgt agcttctctc tgaaactgcc gggaaaggga aaatcttttt aaaatagaca 22200 tcacacaacc aacagggtcc cctaggttca ggcggggagg tgaggtcgag tgaga 22255 52 7 PRT Artificial Sequence Polycatonic peptide 52 Arg Arg Arg Arg Arg Arg Arg 1 5 53 7 PRT Artificial Sequence Polycatonic peptide 53 Lys Lys Leu Lys Leu Xaa Xaa 1 5

Claims (37)

What is claimed is:
1. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 1;
(b) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 2;
(c) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 3;
(d) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 4;
(e) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 5;
(f) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 6;
(g) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 7;
(h) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 8;
(i) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 9;
(j) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 43;
(k) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 44;
(l) a polynucleotide encoding the polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 47;
(m) a polynucleotide capable of hybridizing to and which is at least about 95% identical to the polynucleotide of (a)-(k) or (l) wherein the encoded polypeptide is capable of binding to LDL; and
(n) a biologically active fragment of polynucleotide (a)-(l) or (m) wherein the encoded polypeptide is capable of binding to LDL.
2. The isolated polynucleotide of claim 1 wherein said member is selected from the group consisting of:
(a) a polynucleotide encoding the polypeptide comprising the amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) of the amino acid sequence as set forth in SEQ ID NO: 43;
(b) a polynucleotide encoding the polypeptide comprising the amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) of the amino acid sequence as set forth in SEQ ID NO: 1 and SEQ ID NO: 6;
(c) a polynucleotide encoding the polypeptide comprising the amino acid residues 338-353 (SEQ ID NO: 25), 338-365 (SEQ ID NO: 26), 354-365 (SEQ ID NO: 27) or 444-453 (SEQ ID NO: 28) of the amino acid sequence as set forth in SEQ ID NO: 47;
(d) a polynucleotide encoding the polypeptide comprising the amino acid residues 96-110 (SEQ ID NO: 29) of the amino acid sequence as set forth in SEQ ID NO: 5;
(e) a polynucleotide encoding the polypeptide comprising the amino acid residues 69-75 (SEQ ID NO: 41) of the amino acid sequence as set forth in SEQ ID NO: 44;
(f) a polynucleotide capable of hybridizing to and which is at least about 95%identical to the polynucleotide of (a)-(d) or (e) wherein the encoded polypeptide is capable of binding to LDL; and
(g) a biologically active fragment of polynucleotide (a)-(e) or (f) wherein the encoded polypeptide is capable of binding to LDL.
3. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 10.
4. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 48.
5. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 14.
6. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 15.
7. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 45.
8. The polynucleotide of claim 1 wherein said polynucleotide comprises the nucleic acid as set forth in SEQ ID NO: 46.
9. The polynucleotide of claim 1 wherein said polynucleotide is genomic DNA.
10. A recombinant vector comprising said polynucleotide of claim 1.
11. A cell comprising said recombinant vector of claim 10.
12. A method for producing an LDL binding protein comprising culturing a cell of claim 11 under conditions that permit expression of said LDL binding protein.
13. An isolated polypeptide comprising a member selected from the group consisting of:
(a) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 1;
(b) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 2;
(c) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 3;
(d) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 4;
(e) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 5;
(f) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 6;
(g) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 7;
(h) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 8;
(i) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 9;
(j) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 43;
(k) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 44;
(1) a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 47;
(m) a polypeptide which is at least about 95% identical to the polypeptide of (a)-(k) or (l) wherein said polypeptide is capable of binding to LDL; and
(n) a biologically active fragment of polypeptide (a)-(l) or (m) wherein said fragment is capable of binding to LDL.
14. The isolated polypeptide of claim 13 wherein said member is selected from the group consisting of:
(a) a polypeptide having the amino acid residues 329-343 (SEQ ID NO: 19), 329-354 (SEQ ID NO: 20), 344-354 (SEQ ID NO: 21) or 529-538 (SEQ ID NO: 22) of the amino acid sequence as set forth in SEQ ID NO: 43;
(b) a polypeptide having the amino acid residues 14-43 (SEQ ID NO: 23) or 38-43 (SEQ ID NO: 24) of the amino acid sequence as set forth in SEQ ID NO: 1 and SEQ ID NO: 6;
(c) a polypeptide having the amino acid residues 338-353 (SEQ ID NO: 25), 338-365 (SEQ ID NO: 26), 354-365 (SEQ ID NO: 27) or 444-453 (SEQ ID NO: 28) of the amino acid sequence as set forth in SEQ ID NO: 47;
(d) a polypeptide having the amino acid residues 96-110 (SEQ ID NO: 29) of the amino acid sequence as set forth in SEQ ID NO: 5;
(e) a polypeptide having the amino acid residues 69-75 (SEQ ID NO: 41) of the amino acid sequence as set forth in SEQ ID NO: 44;
(f) a polypeptide which is at least about 95% identical to the polypeptide of (a)-(d) or (e) wherein said polypeptide is capable of binding to LDL; and
(g) a biologically active fragment of polypeptide (a)-(e) or (f) wherein said fragment is capable of binding to LDL.
15. A method for determining if an animal is at risk for atherosclerosis, comprising:
providing an animal; and
evaluating an aspect of LBP metabolism or structure in said animal, an abnormality in said aspect of LBP metabolism or structure being diagnostic of being at risk for atherosclerosis.
16. The method of claim 15 wherein said LBP is selected from the group consisting of LBP-1, LBP-2 and LBP-3.
17. A method for evaluating an agent for use in treating atherosclerosis, comprising:
providing a test cell, cell-free system or animal;
providing an agent;
administering said agent to said test cell, cell-free system or animal in a therapeutically effective amount; and
evaluating the effect of said agent on an aspect of LBP metabolism or structure, a change in said aspect of LBP metabolism or structure being indicative of the usefulness of said agent in treating atherosclerosis.
18. The method of claim 17 wherein said LBP is selected from the group consisting of LBP-1, LBP-2 and LBP-3.
19. The agent identified in claim 17.
20. A method for evaluating an agent for the ability to alter the binding of LBP polypeptide to a binding molecule, comprising:
providing an agent;
providing LBP polypeptide;
providing a binding molecule;
combining said agent, said LBP polypeptide and said binding molecule; and
detecting the formation of a complex comprising said LBP polypeptide and said binding molecule, an alteration in the formation of said complex in the presence of said agent as compared to in the absence of said agent being indicative of said agent altering the binding of said LBP binding molecule.
21. The method of claim 20 wherein said LBP polypeptide is selected from the group consisting of LBP-1, LBP-2 and LBP-3 polypeptide.
22. A method for evaluating an agent for the ability to bind to an LBP polypeptide, comprising:
providing an agent;
providing an LBP polypeptide;
contacting said agent with said LBP polypeptide; and
evaluating the ability of said agent to bind to said LBP polypeptide.
23. The agent identified in claim 22.
24. A method for evaluating an agent for the ability to bind to a nucleic acid encoding an LBP regulatory sequence, comprising:
providing an agent;
providing a nucleic acid encoding an LBP regulatory sequence;
contacting said agent with said nucleic acid; and
evaluating the ability of said agent to bind to said nucleic acid.
25. The agent identified in claim 24.
26. A method for treating atherosclerosis in an animal, comprising:
providing an animal in need of treatment for atherosclerosis;
providing an agent capable of altering an aspect of LBP structure or metabolism;
administering said agent to said animal in a therapeutically effective amount such that treatment of said atherosclerosis occurs.
27. The method of claim 26 wherein said LBP is LBP-1, LBP-2 or LBP-3 polypeptide or a biologically active fragment or analog thereof.
28. A method for treating an animal at risk for atherosclerosis, comprising:
providing an animal at risk for atherosclerosis;
providing an agent capable of altering an aspect of LBP structure or metabolism; and
administering said agent to said animal in a therapeutically effective amount such that treatment of said animal occurs.
29. A method for treating a cell having an abnormality in structure or metabolism of LBP, comprising:
providing a cell having an abnormality in structure or metabolism of LBP;
providing an agent capable of altering an aspect of LBP structure or metabolism; and
administering said agent to said cell in a therapeutically effective amount such that treatment of said cell occurs.
30. A pharmaceutical composition for treating atherosclerosis in an animal, comprising:
a therapeutically effective amount of an agent, said agent being capable of altering an aspect of LBP metabolism or structure in said animal so as to result in treatment of said atherosclerosis; and
a pharmaceutically acceptable carrier.
31. The pharmaceutical composition of claim 30 wherein said agent is an LBP polypeptide or nucleic acid, or active fragment or analog thereof.
32. A vaccine composition for treating atherosclerosis in an animal, comprising:
a therapeutically effective amount of an agent, said agent being capable of altering an aspect of LBP metabolism or structure in said animal so as to result in treatment of said atherosclerosis; and
a pharmaceutically acceptable carrier.
33. A method for diagnosing atherosclerotic lesions in an animal, comprising:
providing an animal;
providing a labeled agent capable of binding to LBP present in atherosclerotic lesions;
administering said labeled agent to said animal under conditions which allow said labeled agent to interact with said LBP so as to result in labeled LBP; and
determining the localization or quantification of said labeled LBP by imaging so as to diagnose the presence of atherosclerotic lesions in said animal.
34. A method for immunizing an animal against an LBP or fragment or analog thereof, comprising:
providing an animal having LDL;
providing an LBP or fragment or analog thereof;
administering said LBP or fragment or analog thereof to said animal so as to stimulate antibody production by said animal to said LBP or fragment or analog thereof such that binding of said LBP to said LDL is altered.
35. A method of making a fragment or analog of LBP polypeptide, said fragment or analog having the ability to bind to modified LDL and native LDL, comprising:
providing an LBP polypeptide;
altering the sequence of said LBP polypeptide; and
testing said altered LBP polypeptide for the ability to bind to modified LDL and native LDL.
36. A method of treating a subject at risk for atherosclerosis, comprising providing a subject at risk for atherosclerosis and administering to the subject one or more of the following:
an LBP protein or fragment or analog thereof and an adjuvant;
a nucleic acid encoding an LBP protein;
a virus or bacteria comprising a nucleic acid encoding an LBP protein; and
an edible plant comprising a nucleic acid encoding an LBP protein.
37. A method of treating a subject at risk for atherosclerosis, comprising:
providing a subject at risk for atherosclerosis;
identifying one or more autologous LBP proteins produced by the subject;
administering to the subject an non-autologous LBP protein, wherein the non-autologous LBP protein induces an immune response against one or more autologous LBP proteins when administered to the subject.
US10/023,529 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis Abandoned US20020129388A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/023,529 US20020129388A1 (en) 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US3193096P 1996-11-27 1996-11-27
US4854797P 1997-06-03 1997-06-03
US08/979,608 US6355451B1 (en) 1996-11-27 1997-11-26 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US09/517,849 US6605588B1 (en) 1996-11-27 2000-03-02 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US09/616,289 US6632923B1 (en) 1996-11-27 2000-07-14 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/023,529 US20020129388A1 (en) 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/616,289 Division US6632923B1 (en) 1996-11-27 2000-07-14 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Publications (1)

Publication Number Publication Date
US20020129388A1 true US20020129388A1 (en) 2002-09-12

Family

ID=27059254

Family Applications (7)

Application Number Title Priority Date Filing Date
US09/616,289 Expired - Fee Related US6632923B1 (en) 1996-11-27 2000-07-14 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US09/976,740 Expired - Fee Related US6878817B2 (en) 1996-11-27 2001-10-12 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/023,529 Abandoned US20020129388A1 (en) 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/023,523 Abandoned US20020152485A1 (en) 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/616,187 Expired - Fee Related US7244410B2 (en) 1996-11-27 2003-07-09 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/671,242 Expired - Fee Related US7402395B2 (en) 1996-11-27 2003-09-24 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US12/137,832 Expired - Fee Related US7807384B2 (en) 1996-11-27 2008-06-12 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/616,289 Expired - Fee Related US6632923B1 (en) 1996-11-27 2000-07-14 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US09/976,740 Expired - Fee Related US6878817B2 (en) 1996-11-27 2001-10-12 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Family Applications After (4)

Application Number Title Priority Date Filing Date
US10/023,523 Abandoned US20020152485A1 (en) 1996-11-27 2001-12-17 Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/616,187 Expired - Fee Related US7244410B2 (en) 1996-11-27 2003-07-09 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US10/671,242 Expired - Fee Related US7402395B2 (en) 1996-11-27 2003-09-24 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US12/137,832 Expired - Fee Related US7807384B2 (en) 1996-11-27 2008-06-12 Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis

Country Status (7)

Country Link
US (7) US6632923B1 (en)
EP (1) EP1263954B1 (en)
JP (1) JP2003525608A (en)
AT (1) ATE458051T1 (en)
AU (1) AU2001243322A1 (en)
DE (1) DE60141323D1 (en)
WO (1) WO2001064874A2 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020137890A1 (en) * 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20040052763A1 (en) * 2000-06-07 2004-03-18 Mond James J. Immunostimulatory RNA/DNA hybrid molecules
WO2002068694A1 (en) * 2001-02-23 2002-09-06 The Johns Hopkins University School Of Medicine Differentially methylated sequences in pancreatic cancer
JP2003308030A (en) 2002-02-18 2003-10-31 Sanyo Electric Co Ltd Display device
US20110177610A1 (en) 2007-06-18 2011-07-21 Sekisui Medical Co., Ltd. Novel marker for arteriosclerotic disease
US7985728B1 (en) 2007-09-20 2011-07-26 Abbott Cardiovascular Systems Inc. Sustained release of Apo A-I mimetic peptides and methods of treatment
US9173890B2 (en) * 2007-09-20 2015-11-03 Abbott Cardiovascular Systems Inc. Sustained release of Apo A-I mimetic peptides and methods of treatment
US7985727B1 (en) 2007-09-20 2011-07-26 Abbott Cardiovascular Systems Inc. Apo A-I mimetic peptides and methods of treatment
US8101565B2 (en) * 2007-09-20 2012-01-24 Abbott Cardiovascular Systems Inc. Sustained release of Apo A-I mimetic peptides and methods of treatment
US8044021B2 (en) * 2007-09-20 2011-10-25 Abbott Cardiovascular Systems Inc. Sustained release of apo A-I mimetic peptides and methods of treatment
US7678444B2 (en) 2007-12-17 2010-03-16 International Paper Company Thermoformed article made from renewable polymer and heat-resistant polymer
CA2725222A1 (en) * 2008-05-05 2009-11-12 Wei Li Thermoformed article made from bio-based biodegradable polymer composition
WO2011071666A1 (en) 2009-12-08 2011-06-16 International Paper Company Thermoformed articles made from reactive extrusion products of biobased materials

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4660563A (en) * 1984-12-31 1987-04-28 Massachusetts Institute Of Technology Method and means for detection of arterial lesions
US4877599A (en) * 1986-11-10 1989-10-31 New England Deaconess Hospital Corporation Detection of vascular disease with labelled antibodies
US5196324A (en) * 1989-12-15 1993-03-23 Eli Lilly And Company Monoclonal antibodies reactive with a human atheroma associated antigen
US5665872A (en) * 1995-10-09 1997-09-09 Kowa Co., Ltd. LDL receptor analog protein and DNA coding therefor
US5726153A (en) * 1988-05-02 1998-03-10 New England Deaconess Hospital Corporation Synthetic peptides for arterial imaging

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59206046A (en) 1983-05-11 1984-11-21 Asahi Chem Ind Co Ltd Material for adsorbing lipoprotein of low specific weight
ZA893740B (en) * 1988-06-03 1990-02-28 Hoffmann La Roche Recombinant coccidiosis vaccines
US5135848A (en) 1989-10-23 1992-08-04 Lehigh University And Northeast Benjamin Franklin Technology Center Of Pa Methods for evaluating cholesterol metabolism and reagents therefor
AU663291B2 (en) 1990-05-03 1995-10-05 New England Deaconess Hospital Corporation Synthetic peptides for arterial imaging
US5399338A (en) * 1991-05-01 1995-03-21 University Of New Mexico Enhancement of abnormal tissue uptake of antibodies, tumor-specific agents or conjugates thereof for diagnostic imaging or therapy
DE4222385A1 (en) * 1992-07-08 1994-01-13 Boehringer Ingelheim Int Isolating substances which inhibit binding of ligands to low-density lipoprotein receptors - comprises incubating LDL receptors with labelled form of ligand in presence of test substance and measuring deg. of binding
EP0586094A1 (en) 1992-08-03 1994-03-09 Wisconsin Alumni Research Foundation Soluble LDL receptor and gene
AU6234194A (en) 1993-01-15 1994-08-15 Board Of Regents, The University Of Texas System High molecular weight b-cell growth factor: interleukin-14
US5698686A (en) 1994-10-20 1997-12-16 Arch Development Corporation Yeast telomerase compositions
US6177614B1 (en) * 1995-03-16 2001-01-23 Cold Spring Harbor Laboratory Control of floral induction in plants and uses therefor
WO1998023282A1 (en) 1996-11-27 1998-06-04 Boston Heart Foundation, Inc. Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US9612811B2 (en) * 2014-01-21 2017-04-04 Nvidia Corporation Confluence analysis and loop fast-forwarding for improving SIMD execution efficiency

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4660563A (en) * 1984-12-31 1987-04-28 Massachusetts Institute Of Technology Method and means for detection of arterial lesions
US4877599A (en) * 1986-11-10 1989-10-31 New England Deaconess Hospital Corporation Detection of vascular disease with labelled antibodies
US5726153A (en) * 1988-05-02 1998-03-10 New England Deaconess Hospital Corporation Synthetic peptides for arterial imaging
US5196324A (en) * 1989-12-15 1993-03-23 Eli Lilly And Company Monoclonal antibodies reactive with a human atheroma associated antigen
US5665872A (en) * 1995-10-09 1997-09-09 Kowa Co., Ltd. LDL receptor analog protein and DNA coding therefor

Also Published As

Publication number Publication date
US7402395B2 (en) 2008-07-22
EP1263954B1 (en) 2010-02-17
US6878817B2 (en) 2005-04-12
US7807384B2 (en) 2010-10-05
US20020194633A1 (en) 2002-12-19
US7244410B2 (en) 2007-07-17
JP2003525608A (en) 2003-09-02
AU2001243322A1 (en) 2001-09-12
EP1263954A2 (en) 2002-12-11
WO2001064874A2 (en) 2001-09-07
US20040013668A1 (en) 2004-01-22
US20100028327A1 (en) 2010-02-04
US20020152485A1 (en) 2002-10-17
WO2001064874A3 (en) 2002-02-14
US20040040049A1 (en) 2004-02-26
US6632923B1 (en) 2003-10-14
ATE458051T1 (en) 2010-03-15
WO2001064874A9 (en) 2003-02-06
DE60141323D1 (en) 2010-04-01

Similar Documents

Publication Publication Date Title
US6605588B1 (en) Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
US7807384B2 (en) Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
Kuusi et al. Apoprotein E polymorphism and coronary artery disease. Increased prevalence of apolipoprotein E-4 in angiographically verified coronary patients.
Yamashita et al. Molecular biology and pathophysiological aspects of plasma cholesteryl ester transfer protein
Schaefer et al. Familial apolipoprotein E deficiency.
Rader et al. In vivo metabolism of a mutant apolipoprotein, apoA-IIowa, associated with hypoalphalipoproteinemia and hereditary systemic amyloidosis.
Morton Cholesteryl ester transfer protein and its plasma regulator: lipid transfer inhibitor protein
Fielding Lipoprotein receptors, plasma cholesterol metabolism, and the regulation of cellular free cholesterol concentration
Farese Jr et al. Apolipoprotein B gene mutations affecting cholesterol levels
Auwerx et al. Coexistence of abnormalities of hepatic lipase and lipoprotein lipase in a large family.
Ahsan et al. Role of lecithin: cholesterol acyltransferase in HDL metabolism and atherosclerosis
Nakazato et al. Expression of very low density lipoprotein receptor mRNA in rabbit atherosclerotic lesions.
US6355451B1 (en) Low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
CA2410253C (en) Regulating lipid levels via the zmax1 or hbm gene
US20040176582A1 (en) High bone mass gene of 11q13.3
MIIDA Apolipoprotein E phenotypes in patients with coronary artery disease
AU769970B2 (en) Novel low density lipoprotein binding proteins and their use in diagnosing and treating atherosclerosis
Murdoch et al. LDL composition in E2/2 subjects and LDL distribution by Apo E genotype in type 1 diabetes
Levy et al. Association of an exon 3 mutation (Trp66→ Gly) of the LDL receptor with variable expression of familial hypercholesterolemia in a French Canadian family
Metzger et al. Co‐regulation of apo A‐I, apo C‐III and apo A‐IV gene expression in human intestinal biopsies
Gotto Jr et al. Plasma lipoproteins: Recent developments
Hobbs ApoE from A to Z: atherosclerosis to alzheimer's
Zhou et al. Adipose tissue-specific CETP expression in mice: impact on plasma lipoprotein metabolism.
Baggio et al. The plasma apolipoproteins: Physiological properties and possible role in lipid disorders and atherogenesis

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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