US20040009535A1 - Compositions and methods for increasing bone mineralization - Google Patents

Compositions and methods for increasing bone mineralization Download PDF

Info

Publication number
US20040009535A1
US20040009535A1 US10/463,190 US46319003A US2004009535A1 US 20040009535 A1 US20040009535 A1 US 20040009535A1 US 46319003 A US46319003 A US 46319003A US 2004009535 A1 US2004009535 A1 US 2004009535A1
Authority
US
United States
Prior art keywords
seq
antibody
tgf
binding
protein
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
US10/463,190
Inventor
Mary Brunkow
David Galas
Brian Kovacevich
John Mulligan
Bryan Paeper
Jeffrey Van Ness
David Winkler
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.)
UCB Pharma SA
Celltech R&D Inc
Original Assignee
Celltech R&D 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=33563705&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040009535(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/449,218 external-priority patent/US6395511B1/en
Application filed by Celltech R&D Inc filed Critical Celltech R&D Inc
Priority to US10/463,190 priority Critical patent/US20040009535A1/en
Assigned to CELLTECH R&D, INC. reassignment CELLTECH R&D, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRUNKOW, MARY E., GALAS, DAVID J., MULLIGAN, JOHN T., VAN NESS, JEFFREY, KOVACEVICH, BRIAN, PAEPER, BRYAN W., WINKLER, DAVID G.
Publication of US20040009535A1 publication Critical patent/US20040009535A1/en
Priority to SG2008068264A priority patent/SG185824A1/en
Priority to PL04776552T priority patent/PL1638999T3/en
Priority to CA2529578A priority patent/CA2529578C/en
Priority to CN2004800233260A priority patent/CN1835968B/en
Priority to ES04776552T priority patent/ES2428767T3/en
Priority to YU20050932A priority patent/RS52769B/en
Priority to PCT/US2004/018910 priority patent/WO2005003158A2/en
Priority to JP2006517259A priority patent/JP4818107B2/en
Priority to BRPI0411535-0A priority patent/BRPI0411535A/en
Priority to DK04776552.4T priority patent/DK1638999T3/en
Priority to EA200600037A priority patent/EA014525B1/en
Priority to CN2012105588665A priority patent/CN103319596A/en
Priority to KR1020057024173A priority patent/KR101221147B1/en
Priority to MEP-31/08A priority patent/MEP3108A/en
Priority to EP04776552.4A priority patent/EP1638999B1/en
Priority to EP10013086A priority patent/EP2341071A1/en
Priority to PT47765524T priority patent/PT1638999E/en
Priority to AU2004253870A priority patent/AU2004253870B2/en
Priority to MEP-2008-31A priority patent/ME00023B/en
Priority to EA201001528A priority patent/EA022991B1/en
Priority to EP10013087A priority patent/EP2338906A1/en
Priority to NZ544617A priority patent/NZ544617A/en
Priority to SI200432101T priority patent/SI1638999T1/en
Priority to NO20060235A priority patent/NO20060235L/en
Priority to IL172599A priority patent/IL172599A/en
Priority to ZA200600384A priority patent/ZA200600384B/en
Priority to US11/399,210 priority patent/US7578999B2/en
Priority to HK06109124.4A priority patent/HK1088907A1/en
Priority to US12/504,296 priority patent/US7985834B2/en
Assigned to UCB MANUFACTURING, INC. reassignment UCB MANUFACTURING, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: DARWIN MOLECULAR CORPORATION
Assigned to DARWIN MOLECULAR CORPORATION reassignment DARWIN MOLECULAR CORPORATION DISSOLUTION OF CELLTECH R&D, INC. AND SUBSEQUENT DISTRIBUTION OF ASSETS INTO DARWIN MOLECULAR CORPORATION Assignors: CELLTECH R&D, INC.
Priority to JP2011157675A priority patent/JP2011246482A/en
Priority to IL222756A priority patent/IL222756A0/en
Priority to CY20131100883T priority patent/CY1114701T1/en
Priority to HRP20131014TT priority patent/HRP20131014T1/en
Assigned to UCB PHARMA S.A. reassignment UCB PHARMA S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UCB MANUFACTURING, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • C12N5/12Fused cells, e.g. hybridomas
    • 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
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/11Immunoglobulins specific features characterized by their source of isolation or production isolated from eggs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/23Immunoglobulins specific features characterized by taxonomic origin from birds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/495Transforming growth factor [TGF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/108Osteoporosis

Definitions

  • the present invention relates generally to pharmaceutical products and methods and, more specifically, to methods and compositions suitable for increasing the mineral content of bone.
  • Such compositions and methods may be utilized to treat a wide variety of conditions, including for example, osteopenia, osteoporosis, fractures and other disorders in which low bone mineral density are a hallmark of the disease.
  • the first phase occurs in both men and women, and proceeds to attainment of a peak bone mass. This first phase is achieved through linear growth of the endochondral growth plates, and radial growth due to a rate of periosteal apposition.
  • the second phase begins around age 30 for trabecular bone (flat bones such as the vertebrae and pelvis) and about age 40 for cortical bone (e.g., long bones found in the limbs) and continues to old age. This phase is characterized by slow bone loss, and occurs in both men and women.
  • Osteoporosis is a debilitating disease in humans characterized by marked decreases in skeletal bone mass and mineral density, structural deterioration of bone including degradation of bone microarchitecture and corresponding increases in bone fragility and susceptibility to fracture in afflicted individuals. Osteoporosis in humans is preceded by clinical osteopenia (bone mineral density that is greater than one standard deviation but less than 2.5 standard deviations below the mean value for young adult bone), a condition found in approximately 25 million people in the United States.
  • osteoporosis defined as bone mineral content greater than 2.5 standard deviations below that of mature young adult bone. Osteoporosis is one of the most expensive diseases for the health care system, costing tens of billions of dollars annually in the United States. In addition to health care-related costs, long-term residential care and lost working days add to the financial and social costs of this disease. Worldwide approximately 75 million people are at risk for osteoporosis.
  • osteoporosis has been defined as an increase in the risk of fracture due to decreased bone mass
  • none of the presently available treatments for skeletal disorders can substantially increase the bone density of adults.
  • drugs are needed which could increase bone density in adults, particularly in the bones of the wrist, spinal column and hip that are at risk in osteopenia and osteoporosis.
  • estrogen is now being prescribed to retard bone loss. There is, however, some controversy over whether there is any long term benefit to patients and whether there is any effect at all on patients over 75 years old. Moreover, use of estrogen is believed to increase the risk of breast and endometrial cancer.
  • calcitonin include calcitonin, bisphosphonates, anabolic steroids and sodium fluoride.
  • Such therapeutics however, have undesirable side effects (e.g., calcitonin and steroids may cause nausea and provoke an immune reaction, bisphosphonates and sodium fluoride may inhibit repair of fractures, even though bone density increases modestly) that may prevent their usage (see Khosla and Rigss, supra).
  • No currently practiced therapeutic strategy involves a drug that stimulates or enhances the growth of new bone mass.
  • the present invention provides compositions and methods which can be utilized to increase bone mineralization, and thus may be utilized to treat a wide variety of conditions where it is desired to increase bone mass. Further, the present invention provides other, related advantages.
  • the present invention provides a novel class or family of TGF-beta binding-proteins, as well as assays for selecting compounds which increase bone mineral content and bone mineral density, compounds which increase bone mineral content and bone mineral density and methods for utilizing such compounds in the treatment or prevention of a wide variety of conditions.
  • isolated nucleic acid molecules are provided, wherein said nucleic acid molecules are selected from the group consisting of: (a) an isolated nucleic acid molecule comprising sequence ID Nos. 1, 5, 7, 9, 11, 13, or, 15, or complementary sequence thereof; (b) an isolated nucleic acid molecule that specifically hybridizes to the nucleic acid molecule of (a) under conditions of high stringency; and (c) an isolated nucleic acid that encodes a TGF-beta binding-protein according to (a) or (b).
  • isolated nucleic acid molecules are provided based upon hybridization to only a portion of one of the above-identified sequences (e.g., for (a) hybridization may be to a probe of at least 20, 25, 50, or 100 nucleotides selected from nucleotides 156 to 539 or 555 to 687 of Sequence ID No. 1).
  • hybridization may be to a probe of at least 20, 25, 50, or 100 nucleotides selected from nucleotides 156 to 539 or 555 to 687 of Sequence ID No. 1).
  • the necessary stringency to be utilized for hybridization may vary based upon the size of the probe.
  • high stringency conditions could include: 60 mM Tris pH 8.0, 2 mM EDTA, 5 ⁇ Denhardt's, 6 ⁇ SSC, 0.1% (w/v) N-laurylsarcosine, 0.5% (w/v) NP-40 (nonidet P-40) overnight at 45 degrees C., followed by two washes with 0.2 ⁇ SSC/0.1% SDS at 45-50 degrees.
  • suitable conditions might include the following: 5 ⁇ SSPE, 5 ⁇ Denhardt's, and 0.5% SDS overnight at 42-50 degrees, followed by two washes with 2 ⁇ SSPE (or 2 ⁇ SSC)/0.1% SDS at 42-50 degrees.
  • isolated nucleic acid molecules are provided which have homology to Sequence ID Nos. 1, 5, 7, 9, 11, 13, or 15, at a 50%, 60%, 75%, 80%, 90%, 95%, or 98% level of homology utilizing a Wilbur-Lipman algorithm.
  • Representative examples of such isolated molecules include, for example, nucleic acid molecules which encode a protein comprising Sequence ID NOs. 2, 6, 10, 12, 14, or 16, or have homology to these sequences at a level of 50%, 60%, 75%, 80%, 90%, 95%, or 98% level of homology utilizing a Lipman-Pearson algorithm.
  • Isolated nucleic acid molecules are typically less than 100 kb in size, and, within certain embodiments, less than 50 kb, 25 kb, 10 kb, or even 5 kb in size. Further, isolated nucleic acid molecules, within other embodiments, do not exist in a “library” of other unrelated nucleic acid molecules (e.g., a subclone BAC such as described in GenBank Accession No. AC003098 and EMB No. AQ171546). However, isolated nucleic acid molecules can be found in libraries of related molecules (e.g., for shuffling, such as is described in U.S. Pat. Nos. 5,837,458; 5,830,721; and 5,811,238). Finally, isolated nucleic acid molecules as described herein do not include nucleic acid molecules which encode Dan, Cerberus, Gremlin, or SCGF (U.S. Pat. No. 5,780,263).
  • cloning vectors which contain the above-noted nucleic acid molecules, and expression vectors which comprise a promoter (e.g., a regulatory sequence) operably linked to one of the above-noted nucleic acid molecules.
  • suitable promoters include tissue-specific promoters, and viral-based promoters (e.g., CMV-based promoters such as CMV I-E, SV40 early promoter, and MuLV LTR).
  • Expression vectors may also be based upon, or derived from viruses (e.g., a “viral vector”).
  • viral vectors include herpes simplex viral vectors, adenoviral vectors, adenovirus-associated viral vectors and retroviral vectors.
  • host cells containing or comprising any of above-noted vectors (including for example, host cells of human, monkey, dog, rat, or mouse origin).
  • methods of producing TGF-beta binding-proteins comprising the step of culturing the aforementioned host cell containing vector under conditions and for a time sufficient to produce the TGF-beta binding protein.
  • the protein produced by this method may be further purified (e.g., by column chromatography, affinity purification, and the like).
  • isolated proteins which are encoded by the above-noted nucleic acid molecules (e.g., Sequence ID NOs. 2, 4, 6, 8, 10, 12, 14, or 16) may be readily produced given the disclosure of the subject application.
  • fusion proteins comprising a first polypeptide segment comprising a TGF-beta binding-protein encoded by a nucleic acid molecule as described above, or a portion thereof of at least 10, 20, 30, 50, or 100 amino acids in length, and a second polypeptide segment comprising a non-TGF-beta binding-protein.
  • the second polypeptide may be a tag suitable for purification or recognition (e.g., a polypeptide comprising multiple anionic amino acid residues—see U.S. Pat. No. 4,851,341), a marker (e.g., green fluorescent protein, or alkaline phosphatase), or a toxic molecule (e.g., ricin).
  • antibodies are provided which are capable of specifically binding the above-described class of TGF-beta binding proteins (e.g., human BEER).
  • the antibody may be a polyclonal antibody, or a monoclonal antibody (e.g., of human or murine origin).
  • the antibody is a fragment of an antibody which retains the binding characteristics of a whole antibody (e.g., an F(ab′) 2 , F(ab) 2 , Fab′, Fab, or Fv fragment, or even a CDR).
  • hybridomas and other cells which are capable of producing or expressing the aforementioned antibodies.
  • methods are provided detecting a TGF-beta binding protein, comprising the steps of incubating an antibody as described above under conditions and for a time sufficient to permit said antibody to bind to a TGF-beta binding protein, and detecting the binding.
  • the antibody may be bound to a solid support to facilitate washing or separation, and/or labeled. (e.g., with a marker selected from the group consisting of enzymes, fluorescent proteins, and radioisotopes).
  • isolated oligonucleotides are provided which hybridize to a nucleic acid molecule according to Sequence ID NOs. 1, 3, 5, 7, 9, 11, 13, 15, 17, or 18 or the complement thereto, under conditions of high stringency.
  • the oligonucleotide may be found in the sequence which encodes Sequence ID Nos. 2, 4, 6, 8, 10, 12, 14, or 16.
  • the oligonucleotide is at least 15, 20, 30, 50, or 100 nucleotides in length.
  • the oligonucleotide is labeled with another molecule (e.g., an enzyme, fluorescent molecule, or radioisotope).
  • primers which are capable of specifically amplifying all or a portion of the above-mentioned nucleic acid molecules which encode TGF-beta binding-proteins.
  • specifically amplifying should be understood to refer to primers which amplify the aforementioned TGF-beta binding-proteins, and not other TGF-beta binding proteins such as Dan, Cerberus, Gremlin, or SCGF (U.S. Pat. No. 5,780,263).
  • oligonucleotide which encodes a TGF-beta binding protein
  • methods for detecting a nucleic acid molecule which encodes a TGF-beta binding protein, comprising the steps of incubating an oligonucleotide as described above under conditions of high stringency, and detecting hybridization of said oligonucleotide.
  • the oligonucleotide may be labeled and/or bound to a solid support.
  • ribozymes are provided which are capable of cleaving RNA which encodes one of the above-mentioned TGF-beta binding-proteins (e.g., Sequence ID NOs. 2, 6, 8, 10, 12, 14, or 16).
  • Such ribozymes may be composed of DNA, RNA (including 2′-O-methyl ribonucleic acids), nucleic acid analogs (e.g., nucleic acids having phosphorothioate linkages) or mixtures thereof.
  • nucleic acid molecules e.g., DNA or cDNA
  • vectors which are capable of expressing or producing the ribozymes.
  • vectors include plasmids, retrotransposons, cosmids, and viral-based vectors (e.g., viral vectors generated at least in part from a retrovirus, adenovirus, or, adeno-associated virus).
  • viral-based vectors e.g., viral vectors generated at least in part from a retrovirus, adenovirus, or, adeno-associated virus.
  • host cells e.g., human, dog, rat, or mouse cells
  • the host cell may be stably transformed with the vector.
  • ribozymes either synthetically, or by in vitro or in vivo transcription.
  • the ribozymes so produced may be further purified and/or formulated into pharmaceutical compositions (e.g., the ribozyme or nucleic acid molecule encoding the ribozyme along with a pharmaceutically acceptable carrier or diluent).
  • the antisense oligonucleotides and antibodies or other selected molecules described herein may be formulated into pharmaceutical compositions.
  • antisense oligonucleotides comprising a nucleic acid molecule which hybridizes to a nucleic acid molecule according to Sequence ID NOs. 1, 3, 5, 7, 9, 11, 13, or 15, or the complement thereto, and wherein said oligonucleotide inhibits the expression of TGF-beta binding protein as described herein (e.g., human BEER).
  • the oligonucleotide is 15, 20, 25, 30, 35, 40, or 50 nucleotides in length.
  • the oligonucleotide is less than 100, 75, or 60 nucleotides in length.
  • the oligonucleotide may be comprised of one or more nucleic acid analogs, ribonucleic acids, or deoxyribonucleic acids. Further, the oligonucleotide may be modified by one or more linkages, including for example, covalent linkage such as a phosphorothioate linkage, a phosphotriester linkage, a methyl phosphonate linkage, a methylene(methylimino) linkage, a morpholino linkage, an amide linkage, a polyamide linkage, a short chain alkyl intersugar linkage, a cycloalkyl intersugar linkage, a short chain heteroatomic intersugar linkage and a heterocyclic intersugar linkage.
  • linkages including for example, covalent linkage such as a phosphorothioate linkage, a phosphotriester linkage, a methyl phosphonate linkage, a methylene(methylimino) linkage
  • methods for increasing bone mineralization, comprising introducing into a warm-blooded animal an effective amount of the ribozyme as described above.
  • such methods comprise the step of introducing into a patient an effective amount of the nucleic acid molecule or vector as described herein which is capable of producing the desired ribozyme, under conditions favoring transcription of the nucleic acid molecule to produce the ribozyme.
  • transgenic, non-human animals are provided.
  • a transgenic animal is provided whose germ cells and somatic cells contain a nucleic acid molecule encoding a TGF-beta binding-protein as described above which is operably linked to a promoter effective for the expression of the gene, the gene being introduced into the animal, or an ancestor of the animal, at an embryonic stage, with the proviso that said animal is not a human.
  • transgenic knockout animals comprising an animal whose germ cells and somatic cells comprise a disruption of at least one allele of an endogenous nucleic acid molecule which hybridizes to a nucleic acid molecule which encodes a TGF-binding protein as described herein, wherein the disruption prevents transcription of messenger RNA from said allele as compared to an animal without the disruption, with the proviso that the animal is not a human.
  • the disruption is a nucleic acid deletion, substitution, or, insertion.
  • the transgenic animal is a mouse, rat, sheep, pig, or dog.
  • kits for the detection of TGF-beta binding-protein gene expression, comprising a container that comprises a nucleic acid molecule, wherein the nucleic acid molecule is selected from the group consisting of (a) a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS: 1, 3, 5, 7, 9, 1 1, 13, 15, 100, or 101; (b) a nucleic acid molecule comprising the complement of the nucleotide sequence of (a); (c) a nucleic acid molecule that is a fragment of (a) or (b) of at least 15, 20 30, 50, 75, or, 100 nucleotides in length. Also provided are kits for the detection of a TGF-beta binding-protein which comprise a container that comprise one of the TGF-beta binding protein antibodies described herein.
  • methods for determining whether a selected molecule is capable of increasing bone mineral content comprising the steps of (a) mixing one or more candidate molecules with TGF-beta-binding-protein encoded by the nucleic acid molecule according to claim 1 and a selected member of the TGF-beta family of proteins (e.g., BMP 5 or 6), (b) determining whether the candidate molecule alters the signaling of the TGF-beta family member, or alters the binding of the TGF-beta binding-protein to the TGF-beta family member.
  • the molecule alters the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation.
  • the candidate molecule(s) may alter signaling or binding by, for example, either decreasing (e.g., inhibiting), or increasing (e.g., enhancing) signaling or binding.
  • methods for determining whether a selected molecule is capable of increasing bone mineral content, comprising the step of determining whether a selected molecule inhibits the binding of TGF-beta binding-protein to bone, or an analogue thereof.
  • bone or analogues thereof include hydroxyapatite and primary human bone samples obtained via biopsy.
  • the selected molecule is contained within a mixture of molecules and the methods may further comprise the step of isolating one or more molecules which are functional within the assay.
  • TGF-beta family of proteins is bound to a solid support and the binding of TGF-beta binding-protein is measured or TGF-beta binding-protein are bound to a solid support and the binding of TGF-beta proteins are measured.
  • a wide variety of molecules may be assayed for their ability to increase bone mineral content by inhibiting the binding of the TGF-beta binding-protein to the TGF-beta family of proteins.
  • Representative examples of such molecules include proteins or peptides, organic molecules, and nucleic acid molecules.
  • methods are provided for increasing bone mineral content in a warm-blooded animal, comprising the step of administering to a warm-blooded animal a therapeutically effective amount of a molecule identified from the assays recited herein.
  • methods are provided for increasing bone mineral content in a warm-blooded animal, comprising the step of administering to a warm-blooded animal a therapeutically effective amount of a molecule which inhibits the binding of the TGF-beta binding-protein to the TGF-beta super-family of proteins, including bone morphogenic proteins (BMPs).
  • suitable molecules include antisense molecules, ribozymes, ribozyme genes, and antibodies (e.g., a humanized antibody) which specifically recognize and alter the activity of the TGF-beta binding-protein.
  • methods for increasing bone mineral content in a warm-blooded animal, comprising the steps of (a) introducing into cells which home to the bone a vector which directs the expression of a molecule which inhibits the binding of the TGF-beta binding-protein to the TGF-beta family of proteins and bone morphogenic proteins (BMPs), and (b) administering the vector-containing cells to a warm-blooded animal.
  • BMPs bone morphogenic proteins
  • methods further comprise, prior to the step of introducing, isolating cells from the marrow of bone which home to the bone.
  • the cells which home to bone are selected from the group consisting of CD34+ cells and osteoblasts.
  • molecules are provided (preferably isolated) which inhibit the binding of the TGF-beta binding-protein to the TGF-beta super-family of proteins.
  • the molecules may be provided as a composition, and can further comprise an inhibitor of bone resorption.
  • an inhibitor of bone resorption include calcitonin, estrogen, a bisphosphonate, a growth factor having anti-resorptive activity and tamoxifen.
  • molecules which may be utilized in the aforementioned therapeutic contexts include, e.g., ribozymes, ribozyme genes, antisense molecules, and/or antibodies (e.g., humanized antibodies). Such molecules may depending upon their selection, used to alter, antagonize, or agonize the signalling or binding of a TGF-beta binding-protein family member as described herein.
  • the above-described molecules and methods of treatment or prevention may be utilized on conditions such as osteoporosis, osteomalasia, periodontal disease, scurvy, Cushing's Disease, bone fracture and conditions due to limb immobilization and steroid usage.
  • the present invention also provides antibodies that specifically bind to a TGF-beta binding protein, sclerostin (SOST), and provides immunogens comprising sclerostin peptides derived from regions of sclerostin that interact with a member of the TGF-beta superfamily such as a bone morphogenic protein.
  • SOST sclerostin
  • the invention provides an antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide, said sclerostin polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the antibody competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in GenBank Ace. Nos.
  • BMP bone morphogenic protein
  • NM — 004329 SEQ ID NO: 102; D89675 (SEQ ID NO: 103); NM — 001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM — 030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP — 001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); or AAB33865 (SEQ ID NO: 110) and wherein the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. NOS.
  • the invention provides an antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide and that impairs formation of a sclerostin homodimer, wherein the sclerostin polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65.
  • the antibody is a polyclonal antibody.
  • the antibody is a monoclonal antibody, which is a mouse, human, rat, or hamster monoclonal antibody.
  • the invention also provides a hybridoma cell or a host cell that is capable of producing the monoclonal antibody.
  • the antibody is a humanized antibody or a chimeric antibody.
  • the invention further provides a host cell that produces the humanized or chimeric antibody.
  • the antigen-binding fragment of the antibody is a F(ab′) 2 , Fab′, Fab, Fd, or Fv fragment.
  • the invention also provides an antibody that is a single chain antibody and provides a host cell that is capable of expressing the single chain antibody.
  • the invention provides a composition comprising such antibodies and a physiologically acceptable carrier.
  • the invention provides an immunogen comprising a peptide comprising at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in GenBank Ace.
  • BMP bone morphogenic protein
  • NM — 004329 SEQ ID NO: 102
  • D89675 SEQ ID NO: 103
  • NM — 001203 SEQ ID NO: 104
  • S75359 SEQ ID NO: 105
  • NM — 030849 SEQ ID NO: 106
  • D38082 SEQ ID NO: 107
  • NP — 001194 SEQ ID NO: 108
  • BAA19765 SEQ ID NO: 109
  • AAB33865 SEQ ID NO: 110
  • the invention also provides an immunogen comprising a peptide that comprises at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that impairs formation of a SOST homodimer.
  • the subject invention immunogens are associated with a carrier molecule.
  • the carrier molecule is a carrier polypeptide, and in particular embodiments, the carrier polypeptide is keyhole limpet hemocyanin.
  • the invention also provides a method for producing an anti-body that specifically binds to a SOST polypeptide, comprising immunizing a non-human animal with an immunogen comprising a peptide comprising at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, wherein (a) the SOST polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65; (b) the antibody competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site; (c) the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace.
  • BMP bone morphogenic protein
  • NM — 004329 SEQ ID NO: 102
  • D89675 SEQ ID NO: 103
  • NM — 001203 SEQ ID NO: 104
  • S75359 SEQ ID NO: 105
  • NM — 030849 SEQ ID NO: 106
  • D38082 SEQ ID NO: 107
  • NP — 001194 SEQ ID NO: 108
  • BAA19765 SEQ ID NO: 109
  • AAB33865 SEQ ID NO: 110
  • the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. NOs.
  • the invention provides a method for producing an antibody that specifically binds to a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, comprising immunizing a non-human animal with an immunogen comprising a peptide that comprises at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the antibody impairs formation of a SOST homodimer.
  • FIG. 1 is a schematic illustration comparing the amino acid sequence of Human Dan; Human Gremlin; Human Cerberus and Human Beer. Arrows indicate the Cysteine backbone.
  • FIG. 2 summarizes the results obtained from surveying a variety of human tissues for the expression of a TGF-beta binding-protein gene, specifically, the Human Beer gene.
  • a semi-quantitative Reverse Transcription-Polymerase Chain Reaction (RT-PCR) procedure was used to amplify a portion of the gene from first-strand cDNA synthesized from total RNA (described in more detail in EXAMPLE 2A).
  • FIGS. 3 A- 3 D summarize the results obtained from RNA in situ hybridization of mouse embryo sections, using a cRNA probe that is complementary to the mouse Beer transcript (described in more detail in EXAMPLE 2B).
  • Panel 3A is a transverse section of 10.5 dpc embryo.
  • Panel 3B is a sagittal section of 12.5 dpc embryo and panels 3C and 3D are sagittal sections of 15.5 dpc embryos.
  • FIGS. 4 A- 4 C illustrate, by western blot analysis, the specificity of three different polyclonal antibodies for their respective antigens (described in more detail in EXAMPLE 4).
  • FIG. 4A shows specific reactivity of an anti-H. Beer antibody for H. Beer antigen, but not H. Dan or H. Gremlin.
  • FIG. 4B shows reactivity of an anti-H. Gremlin antibody for H. Gremlin antigen, but not H. Beer or H. Dan.
  • FIG. 4C shows reactivity of an anti-H. Dan antibody for H. Dan, but not H. Beer or H. Gremlin.
  • FIG. 5 illustrates, by western blot analysis, the selectivity of the TGF-beta binding-protein, Beer, for BMP-5 and BMP-6, but not BMP-4 (described in more detail in EXAMPLE 5).
  • FIG. 6 demonstrates that the ionic interaction between the TGF-beta binding-protein, Beer, and BMP-5 has a dissociation constant in the 15-30 nM range.
  • FIG. 7 presents an alignment of the region containing the characteristic cystine-knot of a SOST (sclerostin) polypeptide and its closest homologues.
  • SOST serostin
  • Three disulphide bonds that form the cystine-knot are illustrated as solid lines.
  • An extra disulphide bond, shown by a dotted line, is unique to this family, which connects two ⁇ -hairpin tips in the 3D structure.
  • the polypeptides depicted are SOST: sclerostin (SEQ ID NO: 126); CGHB: Human Chorionic Gonadotropin ⁇ (SEQ ID NO: 127); FSHB: follicle-stimulating hormone beta subunit (SEQ ID NO: 128); TSHB: thyrotropin beta chain precursor (SEQ ID NO: 129); VWF: Von Willebrand factor (SEQ ID NO: 130); MUC2: human mucin 2 precursor (SEQ ID NO: 131); CER1: Cerberus 1 ( Xenopus laevis homolog) (SEQ ID NO: 132); DRM: gremlin (SEQ ID NO: 133); DAN: (SEQ ID NO: 134); CTGF: connective tissue growth factor precursor (SEQ ID NO: 135); NOV: NovH (nephroblastoma overexpressed gene protein homolog) (SEQ ID NO: 136); CYR6: (SEQ ID NO: 137).
  • FIG. 8 illustrates a 3D model of the core region of SOST (SOST_Core).
  • FIG. 9 presents a 3D model of the core region of SOST homodimer.
  • FIGS. 10A and 10B provide an amino acid sequence alignment of Noggin from five different animals: human (NOGG_HUMAN (SEQ ID NO: 138); chicken (NOGG_CHICK, SEQ ID NO: 139); African clawed frog (NOGG_XENLA, SEQ ID NO: 140); NOGG_FUGRU, SEQ ID NO: 141); and zebrafish (NOGG_ZEBRA, SEQ ID NO: 142); and SOST from human (SOST_HUMAN, SEQ ID NO: 46), rat (SOST_RAT, SEQ ID NO: 65), and mouse (SOST Mouse, SEQ ID NO: 143).
  • FIG. 11 illustrates the Noggin/BMP-7 complex structure.
  • the BMP homodimer is shown on the bottom portion of the figure in surface mode.
  • the Noggin homodimer is shown on top of the BMP dimer in cartoon mode.
  • the circles outline the N-terminal binding region, the core region, and the linker between the N-terminal and core regions.
  • FIG. 12 depicts a 3D model of the potential BMP-binding fragment located at the SOST N-terminal region.
  • a BMP dimer is shown in surface mode, and the potential BMP-binding fragment is shown in stick mode.
  • a phenylalanine residue fitting into a hydrophobic pocket on the BMP surface is noted.
  • Microlecule should be understood to include proteins or peptides (e.g., antibodies, recombinant binding partners, peptides with a desired binding affinity), nucleic acids (e.g., DNA, RNA, chimeric nucleic acid molecules, and nucleic acid analogues such as PNA); and organic or inorganic compounds.
  • proteins or peptides e.g., antibodies, recombinant binding partners, peptides with a desired binding affinity
  • nucleic acids e.g., DNA, RNA, chimeric nucleic acid molecules, and nucleic acid analogues such as PNA
  • organic or inorganic compounds e.g., organic or inorganic compounds.
  • TGF-beta should be understood to include any known or novel member of the TGF-beta super-family, which also includes bone morphogenic proteins (BMPs).
  • BMPs bone morphogenic proteins
  • TGF-beta receptor should be understood to refer to the receptor specific for a particular member of the TGF-beta super-family (including bone morphogenic proteins (BMPs)).
  • BMPs bone morphogenic proteins
  • TGF-beta binding-protein should be understood to refer to a protein with specific binding affinity for a particular member or subset of members of the TGF-beta super-family (including bone morphogenic proteins (BMPs)).
  • BMPs bone morphogenic proteins
  • Specific examples of TGF-beta binding-proteins include proteins encoded by Sequence ID Nos. 1, 5, 7, 9, 11, 13, 15, 100, and 101.
  • Inhibiting the “binding of the TGF-beta binding-protein to the TGF-beta family of proteins and bone morphogenic proteins (BMPs)” should be understood to refer to molecules which allow the activation of TGF-beta or bone morphogenic proteins (BMPs), or allow the binding of TGF-beta family members including bone morphogenic proteins (BMPs) to their respective receptors, by removing or preventing TGF-beta from binding to TGF-binding-protein. Such inhibition may be accomplished, for example, by molecules which inhibit the binding of the TGF-beta binding-protein to specific members of the TGF-beta super-family.
  • Vector refers to an assembly that is capable of directing the expression of desired protein.
  • the vector must include transcriptional promoter elements that are operably linked to the gene(s) of interest.
  • the vector may be composed of deoxyribonucleic acids (“DNA”), ribonucleic acids (“RNA”), or a combination of the two (e.g., a DNA-RNA chimeric).
  • the vector may include a polyadenylation sequence, one or more restriction sites, as well as one or more selectable markers such as neomycin phosphotransferase or hygromycin phosphotransferase.
  • other genetic elements such as an origin of replication, additional nucleic acid restriction sites, enhancers, sequences conferring inducibility of transcription, and selectable markers, may also be incorporated into the vectors described herein.
  • An “isolated nucleic acid molecule” is a nucleic acid molecule that is not integrated in the genomic DNA of an organism.
  • a DNA molecule that encodes a TGF-binding protein that has been separated from the genomic DNA of a eukaryotic cell is an isolated DNA molecule.
  • Another example of an isolated nucleic acid molecule is a chemically-synthesized nucleic acid molecule that is not integrated in the genome of an organism.
  • the isolated nucleic acid molecule may be genomic DNA, cDNA, RNA, or composed at least in part of nucleic acid analogs.
  • isolated polypeptide is a polypeptide that is essentially free from contaminating cellular components, such as carbohydrate, lipid, or other proteinaceous impurities associated with the polypeptide in nature.
  • such isolated polypeptides are at least about 90% pure, more preferably at least about 95% pure, and most preferably at least about 99% pure.
  • a particular protein preparation contains an isolated polypeptide if it appears nominally as a single band on SDS-PAGE gel with Coomassie Blue staining.
  • isolated when referring to organic molecules (e.g., organic small molecules) means that the compounds are greater than 90% pure utilizing methods which are well known in the art (e.g., NMR, melting point).
  • “Sclerosteosis” is a term that was applied by Hansen (1967) (Hansen, H. G., Sklerosteose. in: Opitz, H.; Schmid, F., Handbuch der Kinderheil ambience. Berlin: Springer (pub.) 6 1967. Pp. 351-355) to a disorder similar to van Buchem hyperostosis corticalis generalisata but possibly differing in radiologic appearance of the bone changes and in the presence of asymmetric cutaneous syndactyly of the index and middle fingers in many cases. The jaw has an unusually square appearance in this condition.
  • Humanized antibodies are recombinant proteins in which murine or other non-human animal complementary determining regions of monoclonal antibodies have been transferred from heavy and light variable chains of the murine or other non-human animal immunoglobulin into a human variable domain.
  • an “antibody fragment” is a portion of an antibody such as F(ab′) 2 , F(ab) 2 , Fab′, Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-TGF-beta binding-protein monoclonal antibody fragment binds to an epitope of TGF-beta binding-protein.
  • antibody fragment or antigen-binding fragment also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex.
  • antibody fragments include isolated fragments consisting of the light chain variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (“sFv proteins”), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • a “detectable label” is a molecule or atom that can be conjugated to a polypeptide moiety such as an antibody moiety or a nucleic acid moiety to produce a molecule useful for diagnosis.
  • detectable labels include chelators, photoactive agents, radioisotopes, fluorescent agents, paramagnetic ions, enzymes, and other marker moieties.
  • an “immunoconjugate” is a molecule comprising an anti-TGF-beta binding-protein antibody, or an antibody fragment, and a detectable label or an effector molecule.
  • an immunoconjugate has roughly the same, or only slightly reduced, ability to bind TGF-beta binding-protein after conjugation as before conjugation.
  • TGF-beta “Transforming Growth Factor-beta”
  • TGF-bBP “Transforming Growth Factor-beta binding-protein” (one representative TGF-bBP is designated “H. Beer”)
  • BMP “bone morphogenic protein”
  • PCR “polymerase chain reaction”
  • RT-PCR PCR process in which RNA is first transcribed into DNA using reverse transcriptase (RT);
  • cDNA any DNA made by copying an RNA sequence into DNA form.
  • the present invention provides a novel class of TGF-beta binding-proteins, as well as methods and compositions for increasing bone mineral content in warn-blooded animals.
  • the present inventions are based upon the unexpected discovery that a mutation in the gene which encodes a novel member of the TGF-beta binding-protein family results in a rare condition (sclerosteosis) characterized by bone mineral contents which are one- to four-fold higher than in normal individuals.
  • Sclerosteosis is a disease related to abnormal bone mineral density in humans. Sclerosteosis is a term that was applied by Hansen (1967) (Hansen, H. G., Sklerosteose. In: Opitz, H.; Schmid, F., Handbuch der Kinderheil ambience. Berlin: Springer (pub.) 6 1967. Pp. 351-355) to a disorder similar to van Buchem hyperostosis corticalis generalisata but possibly differing in radiologic appearance of the bone changes and differing in the presence of asymmetric cutaneous syndactyly of the index and middle fingers in many cases.
  • Sclerosteosis is now known to be an autosomal semi-dominant disorder that is characterized by widely disseminated sclerotic lesions of the bone in the adult. The condition is progressive. Sclerosteosis also has a developmental aspect that is associated with syndactyly (two or more fingers are fused together). The Sclerosteosis Syndrome is associated with large stature and many affected individuals attain a height of six feet or more. The bone mineral content of homozygotes can be 1 to 6 fold greater than observed in normal individuals, and bone mineral density can be 1 to 4 fold above normal values (e.g., from unaffected siblings).
  • Sclerosteosis is characterized by the continual deposition of bone throughout the skeleton during the lifetime of the affected individuals. In homozygotes the continual deposition of bone mineral leads to an overgrowth of bone in areas of the skeleton where there is an absence of mechanoreceptors (skull, jaw, cranium). In homozygotes with Sclerosteosis, the overgrowth of the bones of the skull leads to cranial compression and eventually to death due to excessive hydrostatic pressure on the brain stem. In all other parts of the skeleton there is a generalized and diffuse sclerosis. Cortical areas of the long bones are greatly thickened resulting in a substantial increase in bone strength. Trabecular connections are increased in thickness which in turn increases the strength of the trabecular bone. Sclerotic bones appear unusually opaque to x-rays.
  • the rare genetic mutation that is responsible for the Sclerosteosis syndrome has been localized to the region of human chromosome 17 that encodes a novel member of the TGF-beta binding-protein family (one representative example of which is designated “H. Beer”).
  • H. Beer a novel member of the TGF-beta binding-protein family
  • the mechanism of bone mineralization is more fully understood, allowing the development of assays for molecules that increase bone mineralization, and use of such molecules to increase bone mineral content, and in the treatment or prevention of a wide number of diseases.
  • TGF-beta The Transforming Growth Factor-beta (TGF-beta) super-family contains a variety of growth factors that share common sequence elements and structural motifs (at both the secondary and tertiary levels). This protein family is known to exert a wide spectrum of biological responses that affect a large variety of cell types. Many of the TGF-beta family members have important functions during the embryonal development in pattern formation and tissue specification; in adults the family members are involved, e.g., in wound healing and bone repair and bone remodeling, and in the modulation of the immune system.
  • the super-family includes the Bone Morphogenic Proteins (BMPs), Activins, Inhibins, Growth and Differentiation Factors (GDFs), and Glial-Derived Neurotrophic Factors (GDNFs).
  • BMPs Bone Morphogenic Proteins
  • GDFs Growth and Differentiation Factors
  • GDNFs Glial-Derived Neurotrophic Factors
  • Primary classification is established through general sequence features that bin a specific protein into a general sub-family. Additional stratification within the sub-family is possible due to stricter sequence conservation between members of the smaller group.
  • the amino acid identity can be as high as 75% among members of the smaller group. This level of identity enables a single representative sequence to illustrate the key biochemical elements of the sub-group that separates it from other members of the larger family.
  • TGF-beta2 The crystal structure of TGF-beta2 has been determined.
  • the general fold of the TGF-beta2 monomer contains a stable, compact, cysteine knotlike structure formed by three disulphide bridges. Dimerization, stabilized by one disulfide bridge, is antiparallel.
  • TGF-beta signals by inducing the formation of hetero-oligomeric complexes of type I and type II receptors. Transduction of TGF-beta signals involves these two distinct type I and type II subfamilies of transmembrane serine/threonine kinase receptors. At least seven type I receptors and five type II receptors have been identified (see Kawabata et al., Cytokine Growth Factor Rev. 9:49-61 (1998); Miyazono et al., Adv. Immunol. 75:115-57 (2000). TGF-beta family members initiate their cellular action by binding to receptors with intrinsic serine/threonine kinase activity.
  • TGF-beta family binds to a characteristic combination of type I and type II receptors, both of which are needed for signaling.
  • a TGF-beta ligand first binds to the type II receptor (TbR-II), which occurs in the cell membrane in an oligomeric form with activated kinase. Thereafter, the type I receptor (TbR-I), which cannot bind ligand in the absence of TbR-II, is recruited into the complex to form a ligand/type II/type I ternary complex.
  • TbR-II type II receptor
  • TbR-II then phosphorylates TbR-I predominantly in a domain rich in glycine and serine residues (GS domain) in the juxtamembrane region, and thereby activates TbR-I.
  • the activated type I receptor kinase then phosphorylates particular members of the Smad family of proteins that translocate to the nucleus where they modulate transcription of specific genes.
  • BMPS Bone Morphogenic Proteins
  • BMPs bone morphogenic proteins
  • OPs osteogenic proteins
  • the BMPs/OPs (BMP 2-14, and osteogenic protein 1 and ⁇ 2, OP-1 and OP-2) see, e.g., GenBank P12643 (BMP-2); GenBank P12645 (BMP3); GenBank P55107 (BMP-3b, Growth/differentiation factor 10) (GDF-10)); GenBank P12644 (BMP4); GenBank P22003 (BMP5); GenBank P22004 (BMP6); GenBank P18075 (BMP7); GenBank P34820 (BMP8); GenBank Q9UK05 (BMP9); GenBank O95393 (BM10); GenBank O95390 (BMP11, Growth/differentiation factor 11 precursor (GDF-11)); GenBank O95972 (BM15)) are members of the TGF-beta super-family.
  • BMP/OP sub-family The striking evolutionary conservation between members the BMP/OP sub-family suggests that they are critical in the normal development and function of animals. Moreover, the presence of multiple forms of BMPs/OPs raises an important question about the biological relevance of this apparent redundancy. In addition to postfetal chondrogenesis and osteogenesis, the BMPs/OPs play multiple roles in skeletogenesis (including the development of craniofacial and dental tissues) and in embryonic development and organogenesis of parenchymatous organs, including the kidney. It is now understood that nature relies on common (and few) molecular mechanisms tailored to provide the emergence of specialized tissues and organs. The BMP/OP super-family is an elegant example of nature parsimony in programming multiple specialized functions deploying molecular isoforms with minor variation in amino acid motifs within highly conserved carboxy-terminal regions.
  • BMPs are synthesized as large precursor proteins. Upon dimerization, the BMPs are proteolyically cleaved within the cell to yield carboxy-terminal mature proteins that are then secreted from the cell. BMPs, like other TGF-beta family members, initiate signal transduction by binding cooperatively to both type I and type II serine/threonine kinase receptors. Type I receptors for which BMPs may act as ligands include BMPR-IA (also known as ALK-3), BMPR-IB (also known as ALK-6), ALK-1, and ALK-2 (also known as ActR-1).
  • BMPs bind to BMP type II receptor (BMPR-II), Activin type II (ActR-II), and Activin type IIB (ActR-IIB).
  • BMPR-II BMP type II receptor
  • ActR-II Activin type II
  • ActR-IIB Activin type IIB
  • GenBank NM — 004329 GenBank NM — 004329
  • D89675 SEQ ID NO: 103 encoded by SEQ ID NO: 117
  • NM — 001203 SEQ ID NO: 104 encoded by SEQ ID NO: 118
  • S75359 SEQ ID NO: 105 encoded by SEQ ID NO: 119
  • NM — 030849 SEQ ID NO: 106 encoded by SEQ ID NO: 120
  • D38082 SEQ ID NO: 107 encoded by SEQ ID NO: 121).
  • polypeptide sequences of type I receptors are provided in the GenBank database, for example, NP — 001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); and AAB33865 (SEQ ID NO: 110).
  • Polynucleotide sequences and the encoded amino acid sequence of BMP type II receptor polypeptides are provided in the GenBank database and include, for example, U25110 (SEQ ID NO: 111 encoded by SEQ ID NO: 122); NM — 033346 (SEQ ID NO: 112 encoded by SEQ ID NO: 123); NM — 001204 (SEQ ID NO: 113 encoded by SEQ ID NO: 124); and Z48923 (SEQ ID NO: 114 encoded by SEQ ID NO: 125). Additional polypeptide sequences of type II receptors are also provided in the GenBank database, for example, CAA88759 (SEQ ID NO: 115).
  • BMPs similar to other cystine-knot proteins, form a homodimer structure (Scheufler et al., J. Mol. Biol. 287:103-15 (1999)).
  • BMP type I receptor binding site and type II receptor binding site were mapped to the surface of the BMP structure (Innis et al., Protein Eng. 13:839-47 (2000)).
  • the location of the type I receptor binding site on BMP was later confirmed by the x-ray structure of BMP-2/BMP Receptor IA complex (Nickel et al., J. Joint Surg. Am.
  • the BMP and Activin sub-families are subject to significant post-translational regulation, such as by TGF-beta binding proteins.
  • a number of these natural antagonists have been identified, and on the basis of sequence divergence, the antagonists appear to have evolved independently due to the lack of primary sequence conservation. Earlier studies of these antagonists highlighted a distinct preference for interacting and neutralizing BMP-2 and BMP-4.
  • antagonists include noggin, chordin, chordin-like, follistatin, FSRP, the DAN/Cerberus protein family, and sclerostin (SOST) (see Balemans et al., supra, and references cited therein).
  • SOST sclerostin
  • noggin binds to BMPs with high affinity (Zimmerman et al., 1996).
  • a study of the noggin/BMP-7 complex structure revealed the binding interactions between the two proteins (Groppe et al., Nature 420:636-42 (2002)).
  • Superposition of the noggin-BMP-7 structure onto a model of the BMP signaling complex showed that noggin binding effectively masks both pairs of binding epitopes (i.e., BMP Type I and Type II receptor binding sites) on BMP-7.
  • the cysteine-rich scaffold sequence of noggin is preceded by an N-terminal segment of about 20 amino acid residues that are referred to as the “clip” (residues 28-48).
  • the type I receptor-binding site is occluded by the N-terminal portion of the clip domain of Noggin, and the type II receptor binding site is occluded by the carboxy terminal portion of the clip domain.
  • Two ⁇ -strands in the core region near the C-terminus of noggin also contact BMP-7 at the type II receptor binding site. This binding mode enables a noggin dimer to efficiently block all the receptor binding sites (two type I and two type II receptor binding sites) on a BMP dimer.
  • the present invention provides a novel class of TGF-beta binding-proteins that possess a nearly identical cysteine (disulfide) scaffold when compared to Human DAN, Human Gremlin, and Human Cerberus, and SCGF (U.S. Pat. No. 5,780,263) but almost no homology at the nucleotide level.
  • cysteine disulfide
  • SCGF SCGF
  • TGF-beta binding-proteins are disclosed in SEQ ID NOS: 1, 5, 7, 9, 11, 13, 15, 100, and 101.
  • the polynucleotides disclosed herein encode a polypeptide called Beer, which is also referred to herein as sclerostin or SOST.
  • Representative members of this class of binding proteins should also be understood to include variants of the TGF-beta binding-protein (e.g., SEQ ID NOS: 5 and 7).
  • a “TGF-beta binding-protein variant gene” refers to nucleic acid molecules that encode a polypeptide having an amino acid sequence that is a modification of SEQ ID NOS: 2, 10, 12, 14, 16, 46, or 65.
  • Such variants include naturally-occurring polymorphisms or allelic variants of TGF-beta binding-protein genes, as well as synthetic genes that contain conservative amino acid substitutions of these amino acid sequences. A variety of criteria known to those skilled in the art indicate whether amino acids at a particular position in a peptide or polypeptide are similar.
  • a similar amino acid or a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain, which include amino acids with basic side chains (e.g., lysine, arginine, histidine); acidic side chains (e.g., aspartic acid, glutamic acid); uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine); nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); beta-branched side chains (e.g., threonine, valine, isoleucine), and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan).
  • basic side chains e.g., lysine, arginine
  • Proline which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., Leu, Val, Ile, and Ala).
  • substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively.
  • TGF-beta binding-protein gene is nucleic acid molecules that contain insertions or deletions of the nucleotide sequences described herein.
  • TGF-beta binding-protein variant genes can be identified by determining whether the genes hybridize with a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 1, 5, 7, 9, 11, 13, 15, 100, or 101 under stringent conditions.
  • TGF-beta binding-protein variant genes should encode a protein having a cysteine backbone.
  • TGF-beta binding-protein variant genes can be identified by sequence comparison.
  • two amino acid sequences have “100% amino acid sequence identity” if the amino acid residues of the two amino acid sequences are the same when aligned for maximal correspondence.
  • two nucleotide sequences have “100% nucleotide sequence identity” if the nucleotide residues of the two nucleotide sequences are the same when aligned for maximal correspondence.
  • Sequence comparisons can be performed using standard software programs such as those included in the LASERGENE bioinformatics computing suite, which is produced by DNASTAR (Madison, Wis.).
  • a variant TGF-beta binding-protein should have at least a 50% amino acid sequence identity to SEQ ID NOS: 2, 6, 10, 12, 14, 16, 46, or 65 and preferably, greater than 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity.
  • TGF-beta binding-protein variants can be identified by having at least a 70% nucleotide sequence identity to SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101.
  • the present invention contemplates TGF-beta binding-protein gene variants having greater than 75%, 80%, 85%, 90%, or 95% identity to SEQ ID NO: 1 or SEQ ID NO: 100.
  • a variant TGF-beta binding-protein or a polypeptide encoded by a variant TGF-beta binding-protein gene can be functionally characterized by, for example, its ability to bind to and/or inhibit the signaling of a selected member of the TGF-beta family of proteins, or by its ability to bind specifically to an anti-TGF-beta binding-protein antibody.
  • the present invention includes functional fragments of TGF-beta binding-protein genes.
  • a “functional fragment” of a TGF-beta binding-protein gene refers to a nucleic acid molecule that encodes a portion of a TGF-beta binding-protein polypeptide which either (1) possesses the above-noted function activity, or (2) specifically binds with an anti-TGF-beta binding-protein antibody.
  • a functional fragment of a TGF-beta binding-protein gene described herein comprises a portion of the nucleotide sequence of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101.
  • DNA molecules encoding a TGF-beta binding-protein can be obtained by screening a human cDNA or genomic library using polynucleotide probes based upon, for example, SEQ ID NO: 1.
  • the first step in the preparation of a cDNA library is to isolate RNA using methods well-known to those of skill in the art.
  • RNA isolation techniques provide a method for breaking cells, a means of inhibiting RNase-directed degradation of RNA, and a method of separating RNA from DNA, protein, and polysaccharide contaminants.
  • total RNA can be isolated by freezing tissue in liquid nitrogen, grinding the frozen tissue with a mortar and pestle to lyse the cells, extracting the ground tissue with a solution of phenol/chloroform to remove proteins, and separating RNA from the remaining impurities by selective precipitation with lithium chloride (see, for example, Ausubel et al. (eds.), Short Protocols in Molecular Biology, 3rd Edition, pages 4-1 to 4-6 (John Wiley & Sons 1995) [“Ausubel (1995)”]; Wu et al., Methods in Gene Biotechnology, pages 33-41 (CRC Press, Inc. 1997) [“Wu (1997)”]).
  • total RNA can be isolated by extracting ground tissue with guanidinium isothiocyanate, extracting with organic solvents, and separating RNA from contaminants using differential centrifugation (see, for example, Ausubel (1995) at pages 4-1 to 4-6; Wu (1997) at pages 33-41).
  • poly(A) + RNA is preferably isolated from a total RNA preparation.
  • Poly(A) + RNA can be isolated from total RNA by using the standard technique of oligo(dT)-cellulose chromatography (see, for example, Ausubel (1995) at pages 4-11 to 4-12).
  • Double-stranded cDNA molecules may be synthesized from poly(A) + RNA using techniques well-known to those in the art. (see, for example, Wu (1997) at pages 41-46).
  • commercially available kits can be used to synthesize double-stranded cDNA molecules (for example, Life Technologies, Inc. (Gaithersburg, Md.); CLONTECH Laboratories, Inc. (Palo Alto, Calif.); Promega Corporation (Madison, Wis.); and Stratagene Cloning Systems (La Jolla, Calif.)).
  • TGF-beta binding-protein cDNA clones can be modified by constructing a subtracted cDNA library that is enriched in TGF-binding-protein-specific cDNA molecules.
  • Techniques for constructing subtracted libraries are well-known to those of skill in the art (see, for example, Sargent, “Isolation of Differentially Expressed Genes,” in Meth. Enzymol. 152:423, 1987; and Wu et al. (eds.), “Construction and Screening of Subtracted and Complete Expression cDNA Libraries,” in Methods in Gene Biotechnology, pages 29-65 (CRC Press, Inc. 1997)).
  • a cDNA library can be prepared in a vector derived from bacteriophage, such as a ⁇ gt10 vector (see, for example, Huynh et al., “Constructing and Screening cDNA Libraries in ⁇ gt10 and ⁇ gt11, ” in DNA Cloning: A Practical Approach Vol. I, Glover (ed.), page 49 (IRL Press, 1985); Wu (1997) at pages 47-52).
  • double-stranded cDNA molecules can be inserted into a plasmid vector, such as a pBluescript vector (Stratagene Cloning Systems; La Jolla, Calif.), a LambdaGEM-4 (Promega Corp.; Madison, Wis.) or other commercially available vectors.
  • a plasmid vector such as a pBluescript vector (Stratagene Cloning Systems; La Jolla, Calif.), a LambdaGEM-4 (Promega Corp.; Madison, Wis.) or other commercially available vectors.
  • Suitable cloning vectors also can be obtained from the American Type Culture Collection (Rockville, Md.).
  • the cDNA library is inserted into a prokaryotic host, using standard techniques.
  • a cDNA library can be introduced into competent E. coli DH5 cells, which can be obtained from Life Technologies, Inc. (Gaithersburg, Md.).
  • a human genomic DNA library can be prepared by means well-known in the art (see, for example, Ausubel (1995) at pages 5-1 to 5-6; Wu (1997) at pages 307-327). Genomic DNA can be isolated by lysing tissue with the detergent Sarkosyl, digesting the lysate with proteinase K, clearing insoluble debris from the lysate by centrifugation, precipitating nucleic acid from the lysate using isopropanol, and purifying resuspended DNA on a cesium chloride density gradient.
  • DNA fragments that are suitable for the production of a genomic library can be obtained by the random shearing of genomic DNA or by the partial digestion of genomic DNA with restriction endonucleases.
  • Genomic DNA fragments can be inserted into a vector, such as a bacteriophage or cosmid vector, in accordance with conventional techniques, such as the use of restriction enzyme digestion to provide appropriate termini, the use of alkaline phosphatase treatment to avoid undesirable joining of DNA molecules, and ligation with appropriate ligases. Techniques for such manipulation are well-known in the art (see, for example, Ausubel (1995) at pages 5-1 to 5-6; Wu (1997) at pages 307-327).
  • Nucleic acid molecules that encode a TGF-beta binding-protein can also be obtained using the polymerase chain reaction (PCR) with oligonucleotide primers having nucleotide sequences that are based upon the nucleotide sequences of the human TGF-beta binding-protein gene, as described herein.
  • PCR polymerase chain reaction
  • General methods for screening libraries with PCR are provided by, for example, Yu et al., “Use of the Polymerase Chain Reaction to Screen Phage Libraries,” in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, White (ed.), pages 211 -215 (Humana Press, Inc. 1993).
  • human genomic libraries can be obtained from commercial sources such as Research Genetics (Huntsville, Ala.) and the American Type Culture Collection (Rockville, Md.).
  • a library containing cDNA or genomic clones can be screened with one or more polynucleotide probes based upon SEQ ID NO: 1, using standard methods as described herein and known in the art (see, for example, Ausubel (1995) at pages 6-1 to 6-11).
  • Anti-TGF-beta binding-protein antibodies produced as described herein, can also be used to isolate DNA sequences that encode a TGF-beta binding-protein from cDNA libraries.
  • the antibodies can be used to screen ⁇ gt11 expression libraries, or the antibodies can be used for immunoscreening following hybrid selection and translation (see, for example, Ausubel (1995) at pages 6-12 to 6-16; Margolis et al., “Screening ⁇ expression libraries with antibody and protein probes,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), pages 1- 14 (Oxford University Press 1995)).
  • TGF-beta binding-protein cDNA or TGF-beta binding-protein genomic fragment can be determined using standard methods. Moreover, the identification of genomic fragments containing a TGF-beta binding-protein promoter or regulatory element can be achieved using well-established techniques, such as deletion analysis (see generally Ausubel (1995), supra).
  • a TGF-beta binding-protein gene can be obtained by synthesizing DNA molecules using mutually priming long oligonucleotides and the nucleotide sequences described herein (see, for example, Ausubel (1995) at pages 8-8 to 8-9).
  • Established techniques using the polymerase chain reaction provide the ability to synthesize DNA molecules at least two kilobases in length (Adang et al., Plant Molec. Biol. 21:1131, 1993; Bambot et al., PCR Methods and Applications 2:266, 1993; Dillon et al., “Use of the Polymerase Chain Reaction for the Rapid Construction of Synthetic Genes,” in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, White (ed.), pages 263-268, (Humana Press, Inc. 1993); Holowachuk et al., PCR Methods Appl. 4:299, 1995).
  • Nucleic acid molecules encoding variant TGF-beta binding-protein genes can be obtained by screening various cDNA or genomic libraries with polynucleotide probes having nucleotide sequences based upon SEQ ID NO: 1, 5, 9, 11, 13, 15, 100, or 101 using procedures described herein.
  • TGF-beta binding-protein gene variants can also be constructed synthetically.
  • a nucleic acid molecule can be devised that encodes a polypeptide having a conservative amino acid change, compared with the amino acid sequence of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65.
  • variants can be obtained that contain one or more amino acid substitutions of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65, in which an alkyl amino acid is substituted for an alkyl amino acid in a TGF-beta binding-protein amino acid sequence, an aromatic amino acid is substituted for an aromatic amino acid in a TGF-beta binding-protein amino acid sequence, a sulfur-containing amino acid is substituted for a sulfur-containing amino acid in a TGF-beta binding-protein amino acid sequence, a hydroxy-containing amino acid is substituted for a hydroxy-containing amino acid in a TGF-beta binding-protein amino acid sequence, an acidic amino acid is substituted for an acidic amino acid in a TGF-beta binding-protein amino acid sequence, a basic amino acid is substituted for a basic amino acid in a TGF-beta binding-protein amino acid sequence, or a dibasic monocarboxylic amino acid is substituted for a dibasic monocarboxylic
  • a “conservative amino acid substitution” is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine.
  • substitutions it is important, when possible, to maintain the cysteine backbone outlined in FIG. 1.
  • Conservative amino acid changes in a TGF-beta binding-protein gene can be introduced by substituting nucleotides for the nucleotides recited in SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101.
  • Such “conservative amino acid” variants can be obtained, for example, by oligonucleotide-directed mutagenesis, linker-scanning mutagenesis, mutagenesis using the polymerase chain reaction, and the like (see Ausubel (1995) at pages 8-10 to 8-22; McPherson (ed.), Directed Mutagenesis: A Practical Approach (IRL Press 1991)).
  • the functional ability of such variants can be determined using a standard method, such as the assay described herein.
  • a variant TGF-beta binding-protein polypeptide can be identified by the ability to specifically bind anti-TGF-beta binding-protein antibodies.
  • Routine deletion analyses of nucleic acid molecules can be performed to obtain “functional fragments” of a nucleic acid molecule that encodes a TGF-beta binding-protein polypeptide.
  • DNA molecules having the nucleotide sequence of SEQ ID NO: 1 can be digested with Bal31 nuclease to obtain a series of nested deletions. The fragments are then inserted into expression vectors in proper reading frame, and the expressed polypeptides are isolated and tested for activity, or for the ability to bind anti-TGF-beta binding-protein antibodies.
  • exonuclease digestion is to use oligonucleotide-directed mutagenesis to introduce deletions or stop codons to specify production of a desired fragment.
  • particular fragments of a TGF-beta binding-protein gene can be synthesized using the polymerase chain reaction.
  • the present invention also contemplates functional fragments of a TGF-beta binding-protein gene that have conservative amino acid changes.
  • a TGF-beta binding-protein variant gene can be identified on the basis of structure by determining the level of identity with nucleotide and amino acid sequences of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101 and 2, 6, 10, 12, 14, 16, 46, or 65 as discussed above.
  • An alternative approach to identifying a variant gene on the basis of structure is to determine whether a nucleic acid molecule encoding a potential variant TGF-beta binding-protein gene can hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101, or a portion thereof of at least 15 or 20 nucleotides in length.
  • variant TGF-beta binding-protein genes encode a polypeptide that can be characterized by its functional activity, or by the ability to bind specifically to an anti-TGF-beta binding-protein antibody. More specifically, variant TGF-beta binding-protein genes encode polypeptides which exhibit at least 50%, and preferably, greater than 60, 70, 80 or 90%, of the activity of polypeptides encoded by the human TGF-beta binding-protein gene described herein.
  • a nucleic acid molecule encoding the polypeptide must be operably linked to regulatory sequences that control transcriptional expression in an expression vector and then introduced into a host cell.
  • regulatory sequences such as promoters and enhancers
  • expression vectors can include translational regulatory sequences and a marker gene that is suitable for selection of cells that carry the expression vector.
  • Expression vectors that are suitable for production of a foreign protein in eukaryotic cells typically contain (1) prokaryotic DNA elements coding for a bacterial replication origin and an antibiotic resistance marker to provide for the growth and selection of the expression vector in a bacterial host; (2) eukaryotic DNA elements that control initiation of transcription, such as a promoter; and (3) DNA elements that control the processing of transcripts, such as a transcription termination/polyadenylation sequence.
  • TGF-beta binding-proteins of the present invention are preferably expressed in mammalian cells.
  • mammalian host cells include African green monkey kidney cells (Vero; ATCC CRL 1587), human embryonic kidney cells (293-HEK; ATCC CRL 1573), baby hamster kidney cells (BHK-21; ATCC CRL 8544), canine kidney cells (MDCK; ATCC CCL 34), Chinese hamster ovary cells (CHO-K1; ATCC CCL61), rat pituitary cells (GH1; ATCC CCL82), HeLa S3 cells (ATCC CCL2.2), rat hepatoma cells (H-4-II-E; ATCC CRL 1548) SV40-transformed monkey kidney cells (COS-1; ATCC CRL 1650) and murine embryonic cells (NIH-3T3; ATCC CRL 1658).
  • Vero African green monkey kidney cells
  • human embryonic kidney cells (293-HEK; ATCC CRL 1573
  • baby hamster kidney cells BHK-21;
  • the transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a particular gene which has a high level of expression.
  • viral sources such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a particular gene which has a high level of expression.
  • Suitable transcriptional and translational regulatory sequences also can be obtained from mammalian genes, such as actin, collagen, myosin, and metallothionein genes.
  • Transcriptional regulatory sequences include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Suitable eukaryotic promoters include the promoter of the mouse metallothionein I gene [Hamer et al., J. Molec. Appl. Genet. 1:273, 1982], the TK promoter of Herpes virus [McKnight, Cell 31:355, 1982], the SV40 early promoter [Benoist et al., Nature 290:304, 1981], the Rous sarcoma virus promoter [Gorman et al., Proc. Nat'l Acad. Sci.
  • a prokaryotic promoter such as the bacteriophage T3 RNA polymerase promoter, can be used to control TGF-beta binding-protein gene expression in mammalian cells if the prokaryotic promoter is regulated by a eukaryotic promoter (Zhou et al., Mol. Cell. Biol. 10:4529, 1990; Kaufman et al., Nucleic Acids Res. 19:4485, 1991).
  • TGF-beta binding-protein genes may also be expressed in bacterial, yeast, insect, or plant cells.
  • Suitable promoters that can be used to express TGF-beta binding-protein polypeptides in a prokaryotic host are well-known to those of skill in the art and include promoters capable of recognizing the T4, T3, Sp6 and T7 polymerases, the P R and P L promoters of bacteriophage lambda, the trp, recA, heat shock, lacUV5, tac, lpp-lacSpr, phoA, and lacZ promoters of E. coli, promoters of B.
  • subtilis the promoters of the bacteriophages of Bacillus, Streptomyces promoters, the int promoter of bacteriophage lambda, the bla promoter of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene.
  • Prokaryotic promoters have been reviewed by Glick, J. Ind. Microbiol. 1:277, 1987, Watson et al., Molecular Biology of the Gene, 4th Ed. (Benjamin Cummins 1987), and by Ausubel et al. (1995).
  • Preferred prokaryotic hosts include E. coli and Bacillus subtilus.
  • Suitable strains of E. coli include BL21(DE3), BL21(DE3)pLysS, BL21(DE3)pLysE, DH1, DH4I, DH5, DH5I, DH5IF′, DH5IMCR, DH10B, DH10B/p3, DH11S, C600, HB101, JM101, JM105, JM109, JM110, K38, RR1, Y1088, Y1089, CSH18, ER1451, and ER1647 (see, for example, Brown (Ed.), Molecular Biology Labfax (Academic Press 1991)).
  • Suitable strains of Bacillus subtilus include BR151, YB886, MI119, MI120, and B170 (see, for example, Hardy, “Bacillus Cloning Methods,” in DNA Cloning: A Practical Approach, Glover (Ed.) (IRL Press 1985)).
  • the baculovirus system provides an efficient means to introduce cloned TGF-beta binding-protein genes into insect cells.
  • Suitable expression vectors are based upon the Autographa californica multiple nuclear polyhedrosis virus (AcMNPV), and contain well-known promoters such as Drosophila heat shock protein (hsp) 70 promoter, Autographa californica nuclear polyhedrosis virus immediate-early gene promoter (ie-1) and the delayed early 39K promoter, baculovirus p10 promoter, and the Drosophila metallothionein promoter.
  • hsp Drosophila heat shock protein
  • ie-1 Autographa californica nuclear polyhedrosis virus immediate-early gene promoter
  • baculovirus p10 promoter the Drosophila metallothionein promoter.
  • Suitable insect host cells include cell lines derived from IPLB-Sf-21, a Spodoptera frugiperda pupal ovarian cell line, such as Sf9 (ATCC CRL 1711), Sf21AE, and Sf21 (Invitrogen Corporation; San Diego, Calif.), as well as Drosophila Schneider-2 cells.
  • Sf9 ATCC CRL 1711
  • Sf21AE Spodoptera frugiperda pupal ovarian cell line
  • Sf21 Invitrogen Corporation; San Diego, Calif.
  • Drosophila Schneider-2 cells Established techniques for producing recombinant proteins in baculovirus systems are provided by Bailey et al., “Manipulation of Baculovirus Vectors,” in Methods in Molecular Biology, Volume 7: Gene Transfer and Expression Protocols, Murray (ed.), pages 147-168 (The Humana Press, Inc.
  • Promoters for expression in yeast include promoters from GAL1 (galactose), PGK (phosphoglycerate kinase), ADH (alcohol dehydrogenase), AOX1 (alcohol oxidase), HIS4 (histidinol dehydrogenase), and the like.
  • Many yeast cloning vectors have been designed and are readily available. These vectors include YIp-based vectors, such as YIp5, YRp vectors, such as YRp17, YEp vectors such as YEp13 and YCp vectors, such as YCp19.
  • YIp5 YIp5
  • YRp vectors such as YRp17
  • YEp vectors such as YEp13
  • YCp vectors such as YCp19.
  • Expression vectors can also be introduced into plant protoplasts, intact plant tissues, or isolated plant cells. General methods of culturing plant tissues are provided, for example, by Miki et al., “Procedures for Introducing Foreign DNA into Plants,” in Methods in Plant Molecular Biology and Biotechnology, Glick et al. (eds.), pages 67-88 (CRC Press, 1993).
  • An expression vector can be introduced into host cells using a variety of standard techniques including calcium phosphate transfection, liposome-mediated transfection, microprojectile-mediated delivery, electroporation, and the like.
  • the transfected cells are selected and propagated to provide recombinant host cells that comprise the expression vector stably integrated in the host cell genome.
  • Techniques for introducing vectors into eukaryotic cells and techniques for selecting such stable transformants using a dominant selectable marker are described, for example, by Ausubel (1995) and- by Murray (ed.), Gene Transfer and Expression Protocols (Humana Press 1991). Methods for introducing expression vectors into bacterial, yeast, insect, and plant cells are also provided by Ausubel (1995).
  • TGF-beta binding-protein can be isolated by standard techniques, such as affinity chromatography, size exclusion chromatography, ion exchange chromatography, HPLC and the like. Additional variations in TGF-beta binding-protein isolation and purification can be devised by those of skill in the art. For example, anti-TGF-beta binding-protein antibodies, obtained as described below, can be used to isolate large quantities of protein by immunoaffinity purification.
  • the present invention provides antibodies that specifically bind to sclerostin as described herein in detail.
  • Antibodies to TGF-beta binding-protein can be obtained, for example, using the product of an expression vector as an antigen.
  • Antibodies that specifically bind to sclerostin may also be prepared by using peptides derived from any one of the sclerostin polypeptide sequences provided herein (SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, and 65).
  • Particularly useful anti-TGF-beta binding-protein antibodies “bind specifically” with TGF-beta binding-protein of Sequence ID Nos.
  • Antibodies of the present invention may be a polyclonal or, especially a monoclonal antibody.
  • the antibody may belong to any immunoglobulin class, and may be for example an IgG, (including isotypes of IgG, which for human antibodies are known in the art as IgG 1 , IgG 2 , IgG 3 , IgG 4 ); IgE; IgM; or IgA antibody.
  • An antibody may be obtained from fowl or mammals, preferably, for example, from a murine, rat, human or other primate antibody. When desired the antibody may be an internalising antibody.
  • Polyclonal antibodies to recombinant TGF-beta binding-protein can be prepared using methods well-known to those of skill in the art (see, for example, Green et al., “Production of Polyclonal Antisera,” in Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press 1992); Williams et al., “Expression of foreign proteins in E. coli using plasmid vectors and purification of specific polyclonal antibodies,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 15 (Oxford University Press 1995)).
  • an anti-TGF-beta binding-protein antibody of the present invention may also be derived from a subhuman primate antibody.
  • General techniques for raising diagnostically and therapeutically useful antibodies in baboons may be found, for example, in Goldenberg et al., international patent publication No. WO 91/11465 (1991), and in Losman et al., Int. J. Cancer 46:310, 1990.
  • variable region domain may be of any size or amino acid composition and will generally comprise at least one hypervariable amino acid sequence responsible for antigen binding embedded in a framework sequence.
  • variable (V) region domain may be any suitable arrangement of immunoglobulin heavy (V H ) and/or light (V L ) chain variable domains.
  • V H immunoglobulin heavy
  • V L light chain variable domains.
  • the V region domain may be monomeric and be a V H or V L domain where these are capable of independently binding antigen with acceptable affinity.
  • V region domain may be dimeric and contain V H -V H , V H -V L , or V L -V L , dimers in which the V H and V L chains are non-covalently associated (abbreviated hereinafter as F v ).
  • the chains may be covalently coupled either directly, for example via a disulphide bond between the two variable domains, or through a linker, for example a peptide linker, to form a single chain domain (abbreviated hereinafter as scF v ).
  • variable region domain may be any naturally occuring variable domain or an engineered version thereof.
  • engineered version is meant a variable region domain that has been created using recombinant DNA engineering techniques.
  • engineered versions include those created for example from natural antibody variable regions by insertions, deletions or changes in or to the amino acid sequences of the natural antibodies.
  • Particular examples of this type include those engineered variable region domains containing at least one CDR and optionally one or more framework amino acids from one antibody and the remainder of the variable region domain from a second antibody.
  • variable region domain may be covalently attached at a C-terminal amino acid to at least one other antibody domain or a fragment thereof.
  • a V H domain is present in the variable region domain this may be linked to an immunoglobulin C H 1 domain or a fragment thereof.
  • a V L domain may be linked to a C K domain or a fragment thereof.
  • the antibody may be a Fab fragment wherein the antigen binding domain contains associated V H and V L domains covalently linked at their C-termini to a CH1 and C K domain respectively.
  • the CH1 domain may be extended with further amino acids, for example to provide a hinge region domain as found in a Fab′ fragment, or to provide further domains, such as antibody CH2 and CH3 domains.
  • CDR peptides (“minimal recognition units”) can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al., Methods: A Companion to Methods in Enzymology 2:106, 1991; Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies,” in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al.
  • Antibodies for use in the invention may in general be monoclonal (prepared by conventional immunisation and cell fusion procedures) or in the case of fragments, derived therefrom using any suitable standard chemical such as reduction or enzymatic cleavage and/or digestion techniques, for example by treatment with pepsin. More specifically, monoclonal anti-TGF-beta binding-protein antibodies can be generated utilizing a variety of techniques. Rodent monoclonal antibodies to specific antigens may be obtained by methods known to those skilled in the art (see, for example, Kohler et al., Nature 256:495, 1975; and Coligan et al.
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising a TGF-beta binding-protein gene product, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B-lymphocytes, fusing the B-lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • an anti-TGF-beta binding-protein antibody of the present invention may be derived from a human monoclonal antibody.
  • Human monoclonal antibodies are obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.
  • Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., Nature Genet. 7:13, 1994; Lonberg et al., Nature 368:856, 1994; and Taylor et al., Int. Immun. 6:579, 1994.
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al., “Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)).
  • antibody fragments can be obtained, for example, by proteolytic hydrolysis of the antibody.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′) 2 . This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab′ monovalent fragments.
  • the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages.
  • an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an Fc fragment directly.
  • the antibody may be a recombinant or engineered antibody obtained by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions.
  • DNA is known and/or is readily available from DNA libraries including for example phage-antibody libraries (see Chiswell, D J and McCafferty, J. Tibtech. 10 80-84 (1992)) or where desired can be synthesised. Standard molecular biology and/or chemistry procedures may be used to sequence and manipulate the DNA, for example, to introduce codons to create cysteine residues, to modify, add or delete other amino acids or domains as desired.
  • One or more replicable expression vectors containing the DNA encoding a variable and/or constant region may be prepared and used to transform an appropriate cell line, e.g. a non-producing myeloma cell line, such as a mouse NSO line or a bacterial, such as E.coli, in which production of the antibody will occur.
  • an appropriate cell line e.g. a non-producing myeloma cell line, such as a mouse NSO line or a bacterial, such as E.coli
  • the DNA sequence in each vector should include appropriate regulatory sequences, particularly a promoter and leader sequence operably linked to a variable domain sequence. Particular methods for producing antibodies in this way are generally well known and routinely used.
  • the-antibody according to the invention may have one or more effector or reporter molecules attached to it and the invention extends to such modified proteins.
  • a reporter molecule may be a detectable moiety or label such as an enzyme, cytotoxic agent or other reporter molecule, including a dye, radionuclide, luminescent group, fluorescent group, or biotin, or the like.
  • the TGF-beta binding protein-specific immunoglobulin or fragment thereof may be radiolabeled for diagnostic or therapeutic applications. Techniques for radiolabeling of antibodies are known in the art. See, e.g., Adams 1998 In Vivo 12:11-21; Hiltunen 1993 Acta Oncol. 32:831-9.
  • TGF-beta binding protein specific antibody or fragment thereof
  • the effector or reporter molecules may be attached to the antibody through any available amino acid side-chain, terminal amino acid or, where present carbohydrate functional group located in the antibody, provided that the attachment or the attachment process does not adversely affect the binding properties and the usefulness of the molecule.
  • Particular functional groups include, for example any free amino, imino, thiol, hydroxyl, carboxyl or aldehyde group. Attachment of the antibody and the effector and/or reporter molecule(s) may be achieved via such groups and an appropriate functional group in the effector or reporter molecules.
  • the linkage may be direct or indirect through spacing or bridging groups.
  • Effector molecules include, for example, antineoplastic agents, toxins (such as enzymatically active toxins of bacterial (such as P. aeruginosa exotoxin A) or plant origin and fragments thereof (e.g. ricin and fragments thereof; plant gelonin, bryodin from Bryonia dioica, or the like. See, e.g., Thrush et al., 1996 Annu. Rev. Immunol. 14:49-71; Frankel et al., 1996 Cancer Res. 56:926-32); biologically active proteins, for example enzymes; nucleic acids and fragments thereof such as.
  • toxins such as enzymatically active toxins of bacterial (such as P. aeruginosa exotoxin A) or plant origin and fragments thereof (e.g. ricin and fragments thereof; plant gelonin, bryodin from Bryonia dioica, or the like. See, e.g., Thrush et al., 1996
  • DNA, RNA and fragments thereof DNA, RNA and fragments thereof; naturally occurring and synthetic polymers (e.g., polysaccharides and polyalkylene polymers such as poly(ethylene glycol) and derivatives thereof); radionuclides, particularly radioiodide; and chelated metals.
  • Suitable reporter groups include chelated metals, fluorescent compounds, or compounds that may be detected by NMR or ESR spectroscopy. Particularly useful effector groups are calichaemicin and derivatives thereof (see, for example, South African Patent Specifications Nos. 85/8794, 88/8127 and 90/2839).
  • toxins including chemotherapeutic agents, anti-mitotic agents, antibiotics, inducers of apoptosis (or “apoptogens”, see, e.g., Green and Reed, 1998, Science 281:1309-1312), or the like, are known to those familiar with the art, and the examples provided herein are intended to be illustrative without limiting the scope and spirit of the invention.
  • Particular antineoplastic agents include cytotoxic and cytostatic agents, for example alkylating agents, such as nitrogen mustards (e.g., chlorambucil, melphalan, mechlorethamine, cyclophosphamide, or uracil mustard) and derivatives thereof, triethylenephosphoramide, triethylenethiophosphor-amide, busulphan, or cisplatin; antimetabolites, such as methotrexate, fluorouracil, floxuridine, cytarabine, mercaptopurine, thioguanine, fluoroacetic acid or fluorocitric acid, antibiotics, such as bleomycins (e.g., bleomycin sulphate), doxorubicin, daunorubicin, mitomycins (e.g., mitomycin C), actinomycins (e.g., dactinomycin) plicamycin, calichaemicin and derivatives thereof, or esperamicin and
  • Chelated metals include chelates of di-or tripositive metals having a coordination number from 2 to 8 inclusive.
  • Particular examples of such metals include technetium (Tc), rhenium (Re), cobalt (Co), copper (Cu), gold (Au), silver (Ag), lead (Pb), bismuth (Bi), indium (In), gallium (Ga), yttrium (Y), terbium (Tb), gadolinium (Gd), and scandium (Sc).
  • the metal is preferably a radionuclide.
  • radionuclides include 99m Tc, 186 Re, 188 Re, 58 Co, 60 Co, 67 Cu, 195 Au, 199 Au, 110 Ag, 203 Pb, 206 Bi, 207 Bi, 111 In, 67 Ga, 68 Ga, 88 Y, 90 Y, 160 Tb, 153 Gd, and 47 Sc.
  • the chelated metal may be for example one of the above types of metal chelated with any suitable polydentate chelating agent, for example acyclic or cyclic polyamines, polyethers, (e.g., crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives.
  • a suitable polydentate chelating agent for example acyclic or cyclic polyamines, polyethers, (e.g., crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives.
  • the type of chelating agent will depend on the metal in use.
  • One particularly useful group of chelating agents in conjugates according to the invention comprises acyclic and cyclic polyamines, especially.
  • polyaminocarboxylic acids for example diethylenetriaminepentaacetic acid and derivatives thereof, and macrocyclic amines, such as cyclic tri-aza and tetra-aza derivatives (for example, as described in International Patent Specification No. WO 92/22583), and polyamides, especially desferrioxamine and derivatives thereof.
  • a thiol group in the antibody when used as the point of attachment this may be achieved through reaction with a thiol reactive group present in the effector or reporter molecule.
  • a thiol reactive group present in the effector or reporter molecule.
  • examples of such groups include an á-halocarboxylic acid or ester, such as iodoacetamide, an imide, such as maleimide, a vinyl sulphone, or a disulphide.
  • the present invention provides methods for selecting and/or isolating compounds that are capable of increasing bone density. For example, within one aspect of the present invention methods are provided for determining whether a selected molecule (e.g., a candidate agent) is capable of increasing bone mineral content, comprising the steps of (a) mixing (or contacting) a selected molecule with TGF-beta binding protein and a selected member of the TGF-beta family of proteins, (b) determining whether the selected molecule stimulates signaling by the TGF-beta family of proteins, or inhibits the binding of the TGF-beta binding protein to at least one member of the TGF-beta family of proteins.
  • the molecule enhances the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation.
  • methods for determining whether a selected molecule (candidate agent) is capable of increasing bone mineral content, comprising the steps of (a) exposing (contacting, mixing, combining) a selected molecule to cells which express TGF-beta binding-protein and (b) determining whether the expression (or activity) of TGF-beta binding-protein in the exposed cells decreases, or whether an activity of the TGF-beta binding protein decreases, and therefrom determining whether the compound is capable of increasing bone mineral content.
  • the cells are selected from the group consisting of the spontaneously transformed or untransformed normal human bone from bone biopsies and rat parietal bone osteoblasts.
  • Immunoassays may be used for detecting and quantifying the expression of a TGF-beta binding protein and include, for example, Countercurrent Immuno-Electrophoresis (CIEP), radioimmunoassays, radioimmunoprecipitations, Enzyme-Linked Immuno-Sorbent Assays (ELISA), immunoblot assays such as dot blot assays and Western blots, inhibition or competition assays, and sandwich assays (see U.S. Pat. Nos.
  • CIEP Countercurrent Immuno-Electrophoresis
  • ELISA Enzyme-Linked Immuno-Sorbent Assays
  • immunoblot assays such as dot blot assays and Western blots, inhibition or competition assays
  • sandwich assays see U.S. Pat. Nos.
  • Such immunoassays may use an antibody that is specific for a TGF-beta binding protein such as the anti-sclerostin antibodies described herein, or may use an antibody that is specific for a reporter molecule that is attached to the TGF-beta binding protein.
  • the level of polypeptide expression may also be determined by quantifying the amount of TGF-beta binding protein that binds to a TGF-beta binding protein ligand.
  • binding of sclerostin in a sample to a BMP may be detected by surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • the level of expression of mRNA encoding the specific TGF-beta binding protein may be quantified.
  • a family member of the TGF-beta super-family or a TGF-beta binding protein is first bound to a solid phase, followed by addition of a candidate molecule.
  • a labeled family member of the TGF-beta super-family or a TGF-beta binding protein is then added to the assay (i.e., the labeled polypeptide is the ligand for whichever polypeptide was bound to the solid phase), the solid phase washed, and the quantity of bound or labeled TGF-beta super-family member or TGF-beta binding protein on the solid support determined.
  • Molecules which are suitable for use in increasing bone mineral content as described herein are those molecules which decrease the binding of TGF-beta binding protein to a member or members of the TGF-beta super-family in a statistically significant manner.
  • assays suitable for use within the present invention should not be limited to the embodiments described within Examples 2 and 3. In particular, numerous parameters may be altered, such as by binding TGF-beta to a solid phase, or by elimination of a solid phase entirely.
  • methods for determining whether a selected molecule is capable of increasing bone mineral content, comprising the steps of (a) exposing (contacting, mixing, combining) a selected molecule (candidate agent) to cells which express TGF-beta and (b) determining whether the activity of TGF-beta from said exposed cells is altered, and therefrom determining whether the compound is capable of increasing bone mineral content.
  • the candidate agent is an antibody that binds to the TGF-beta binding protein sclerostin disclosed herein.
  • a method for identifying an antibody that modulates a TGF-beta signaling pathway comprising contacting an antibody that specifically binds to a SOST polypeptide with a SOST peptide, including but not limited to the peptides disclosed herein, under conditions and for a time sufficient to permit formation of an antibody plus (+) SOST (antibody/SOST) complex and then detecting the level (e.g., quantifying the amount) of the SOST/antibody complex to determine the presence of an antibody that modulates a TGF-beta signaling pathway.
  • a TGF-beta signaling pathway includes a signaling pathway by which a BMP binds to a type I and a type II receptor on a cell to stimulate or induce the pathway that modulates bone mineral content.
  • an antibody that specifically binds to SOST stimulates or enhances the pathway for increasing bone mineral content. Such an antibody may be identified using the methods disclosed herein to detect binding of an antibody to SOST specific peptides.
  • the subject invention methods may also be used for identifying antibodies that impair, inhibit (including competitively inhibit), or prevent binding of a BMP to a SOST polypeptide by detecting whether an antibody binds to SOST peptides that are located in regions or portions of regions on SOST to which a BMP binds, such as peptides at the amino terminal end of SOST and peptides that include amino terminal amino acid residues and a portion of the core region (docking core) of SOST (e.g., SEQ ID NOS: 47-64, 66-73, and 92-95).
  • the methods of the present invention may also be used to identify an antibody that impairs, prevents, or inhibits, formation of SOST homodimers.
  • Such an antibody that binds specifically to SOST may be identified by detecting binding of the antibody to peptides that are derived from the core or the carboxy terminal region of SOST (e.g., SEQ ID NOS: 74-91 and 96-99).
  • methods for determining whether a selected molecule is capable of increasing bone mineral content, comprising the steps of (a) mixing or contacting a selected molecule (candidate agent) with a TGF-beta-binding-protein and a selected member of the TGF-beta family of proteins, (b) determining whether the selected molecule up-regulates the signaling of the TGF-beta family of proteins, or inhibits the binding of the TGF-beta binding-protein to the TGF-beta family of proteins.
  • the molecule enhances the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation.
  • methods for determining whether a selected molecule (candidate agent) is capable of increasing bone mineral content, comprising the step of determining whether a selected molecule inhibits the binding of TGF-beta binding-protein to bone, or an analogue thereof.
  • bone or analogues thereof refers to hydroxyapatite, or a surface composed of a powdered form of bone, crushed bone or intact bone.
  • a wide variety of methods may be utilized to assess the inhibition of TGF-beta binding-protein localization to bone matrix.
  • One such representative method is provided below in Example 7 (see also Nicolas et al., Calcif. Tissue Int. 47:206-12 (1995)).
  • an antibody or antigen-binding fragment thereof that specifically binds to a sclerostin polypeptide is capable of competitively inhibiting binding of a TGF-beta family member to the sclerostin polypeptide.
  • the capability of the antibody or antibody fragment to impair or blocking binding of a TGF-beta family member, such as a BMP, to sclerostin may be determined according to any of the methods described herein.
  • the antibody or fragment thereof that specifically binds to sclerostin may impair, block, or prevent binding of a TGF-beta family member to sclerostin by impairing sclerostin homodimer formation.
  • An antibody that specifically binds to sclerostin may also be used to identify an activity of sclerostin by inhibiting or impairing sclerostin from binding to a BMP.
  • the antibody or fragment thereof may be incorporated in a cell-based assay or in an animal model in which sclerostin has a defined activity to determine whether the antibody alters (increases or decreases in a statistically significant manner) that activity.
  • An antibody or fragment thereof that specifically binds to sclerostin may be used to examine the effect of such an antibody in a signal transduction pathway and thereby modulate (stimulate or inhibit) the signaling pathway.
  • binding of an antibody to SOST results in a stimulation or induction of a signaling pathway.
  • the methods recited herein may refer to the analysis of an individual test molecule, that the present invention should not be so limited.
  • the selected molecule may be contained within a mixture of compounds.
  • the recited methods may further comprise the step of isolating a molecule that inhibits the binding of TGF-beta binding-protein to a TGF-beta family member.
  • a wide variety of molecules may be assayed for their ability to inhibit the binding of TGF-beta binding-protein to a TGF-beta family member.
  • Representative examples discussed in more detail below include organic molecules (e.g., organic small molecules), proteins or peptides, and nucleic acid molecules.
  • organic small molecules may be assayed for their ability to inhibit the binding of TGF-beta binding-protein to a TGF-beta family member.
  • suitable organic molecules may be selected from either a chemical library, wherein chemicals are assayed individually, or from combinatorial chemical libraries where multiple compounds are assayed at once, then deconvoluted to determine and isolate the most active compounds.
  • combinatorial chemical libraries include those described by Agrafiotis et al., “System and method of automatically generating chemical compounds with desired properties,” U.S. Pat. No. 5,463,564; Armstrong, R. W., “Synthesis of combinatorial arrays of organic compounds through the use of multiple component combinatorial array syntheses,” WO 95/02566; Baldwin, J. J. et al., “Sulfonamide derivatives and their use,” WO 95/24186; Baldwin, J. J.
  • a wide range of proteins and peptides may likewise be utilized as candidate molecules for inhibitors of the binding of TGF-beta binding-protein to a TGF-beta family member.
  • Peptide molecules which are putative inhibitors of the binding of TGF-beta binding-protein to a TGF-beta family member may be obtained through the screening of combinatorial peptide libraries.
  • Such libraries may either be prepared by one of skill in the art (see e.g., U.S. Pat. Nos. 4,528,266 and 4,359,535, and Patent Cooperation Treaty Publication Nos. WO 92/15679, WO 92/15677, WO 90/07862, WO 90/02809, or purchased from commercially available sources (e.g., New England Biolabs Ph.D.TM Phage Display Peptide Library Kit).
  • the present invention provides antibodies that specifically bind to a sclerostin polypeptide methods for using such antibodies.
  • the present invention also provides sclerostin polypeptide immunogens that may be used for generation and analysis of these antibodies.
  • the antibodies may be useful to block or impair binding of a sclerostin polypeptide, which is a TGF-beta binding protein, to a ligand, particularly a bone morphogenic protein, and may also block or impair binding of the sclerostin polypeptide to one or more other ligands.
  • a molecule such as an antibody that inhibits the binding of the TGF-beta binding protein to one or more members of the TGF-beta family of proteins, including one or more bone morphogenic proteins (BMPs), should be understood to refer to, for example, a molecule that allows the activation of a TGF-beta family member or BMP, or allows binding of TGF-beta family members including one or more BMPs to their respective receptors by removing or preventing the TGF-beta member from binding to the TGF-binding-protein.
  • BMPs bone morphogenic proteins
  • the present invention also provides peptide and polypeptide immunogens that may be used to generate and/or identify antibodies or fragments thereof that are capable of inhibiting, preventing, or impairing binding of the TGF-beta binding protein sclerostin to one or more BMPs.
  • the present invention also provides peptide and polypeptide immunogens that may be used to generate and/or identify antibodies or fragments thereof that are capable of inhibiting, preventing, or impairing (e.g., decreasing in a statistically significant manner) the formation of sclerostin homodimers.
  • the antibodies of the present invention are useful for increasing the mineral content and mineral density of bone, thereby ameliorating numerous conditions that result in the loss of bone mineral content, including for example, disease, genetic predisposition, accidents that result in the lack of use of bone (e.g., due to fracture), therapeutics that effect bone resorption or that kill bone forming cells, and normal aging.
  • Polypeptides or peptides useful for immunization and/or analysis of sclerostin-specific antibodies may also be selected by analyzing the primary, secondary, and tertiary structure of a TGF-beta binding protein according to methods known to those skilled in the art and described herein, in order to determine amino acid sequences more likely to generate an antigenic response in a host animal. See, e.g., Novotny, Mol. Immunol. 28:201-207 (1991); Berzofsky, Science 229:932-40 (1985)).
  • Modeling and x-ray crystallography data may also be used to predict and/or identify which portions or regions of a TGF-beta binding protein interact with which portions of a TGF-beta binding protein ligand, such as a BMP.
  • TGF-beta binding protein peptide immunogens may be designed and prepared that include amino acid sequences within or surrounding the portions or regions of interaction. These antibodies may be useful to block or impair binding of the TGF-beta binding protein to the same ligand and may also block or impair binding of the TGF-beta binding protein to one or more other ligands.
  • Antibodies or antigen binding fragments thereof contemplated by the present invention include antibodies that are capable of specifically binding to sclerostin and competitively inhibiting binding of a TGF-beta polypeptide, such as a BMP, to sclerostin.
  • a TGF-beta polypeptide such as a BMP
  • the antibodies contemplated by the present invention competitively inhibit binding of the sclerostin polypeptide to the BMP Type I receptor site on a BMP, or to the BMP Type II receptor binding site, or may competitively inhibit binding of sclerostin to both the Type I and Type II receptor binding sites on a BMP.
  • an anti-sclerostin antibody competitively inhibits binding of the Type I and/or Type II binding sites of the BMP polypeptide to sclerostin, thus blocking the antagonistic activity of sclerostin, the receptor binding sites on BMP are available to bind to the Type I and Type II receptors, thereby increasing bone mineralization.
  • the binding interaction between a TGF-beta binding protein such as sclerostin and a TGF-beta polypeptide such as a BMP generally occurs when each of the ligand pairs forms a homodimer.
  • a sclerostin specific antibody may be used to block or impair sclerostin homodimer formation.
  • Noggin dimer which is a BMP antagonist that has the ability to bind a BMP with high affinity (Zimmerman et al., supra) was isolated in complex with one dimer of human BMP-7 and analyzed by multiwavelength anomalous diffraction (MAD) (Groppe et al., Nature 420:636-42 (2002)). As discussed herein, this study revealed that Noggin dimer may efficiently block all the receptor binding sites (two type I and two type II receptor binding sites) on a BMP dimer.
  • MAD multiwavelength anomalous diffraction
  • the location of the amino acids of Noggin that contact BMP-7 may be useful in modeling the interaction between other TGF-beta binding proteins, such as sclerostin (SOST), and BMPs, and thus aiding the design of peptides that may be used as immunogens to generate antibodies that block or impair such an interaction.
  • SOST sclerostin
  • an antibody, or an antigen-binding fragment thereof, that binds specifically to a SOST polypeptide competitively inhibits binding of the SOST polypeptide to at least one or both of a bone morphogenic protein (BMP) Type I Receptor binding site and a BMP Type II Receptor binding site that are located on a BMP.
  • BMP bone morphogenic protein
  • the epitopes on SOST to which these antibodies bind may include or be included within contiguous amino acid sequences that are located at the N-terminus of the SOST polypeptide (amino acids at about positions 1-56 of SEQ ID NO: 46).
  • the polypeptides may also include a short linker peptide sequence that connects the N-terminal region to the core region, for example, polypeptides as provided in SEQ ID NO: 92 (human) and SEQ ID NO: 93 (rat).
  • Shorter representative N-terminus peptide sequences of human SOST e.g., SEQ ID NO: 46
  • SEQ ID NOS: 47-51 e.g., SEQ ID NO: 65
  • peptide sequences include SEQ ID NOS: 57-60.
  • Antibodies that specifically bind to a SOST polypeptide and block or competitively inhibit binding of the SOST polypeptide to a BMP may also specifically bind to peptides that comprise an amino acid sequence corresponding to the core region of SOST (amino acids at about positions 57-146 of SEQ ID NO: 46).
  • Polypeptides that include the core region may also include additional amino acids extending at either or both the N-terminus and C-terminus, for example, to include cysteine residues that may be useful for conjugating the polypeptide to a carrier molecule.
  • Representative core polypeptides of human and rat SOST comprise the amino acid sequences set forth in SEQ ID NO: 94 and SEQ ID NO: 95, respectively. Such antibodies may also bind shorter polypeptide sequences.
  • Representative human SOST core peptide sequences are provided in SEQ ID NOS: 66-69 and representative rat SOST core sequences are provided in SEQ ID NOS: 70-73.
  • antibodies that specifically bind to a SOST polypeptide impair (inhibit, prevent, or block, e.g., decrease in a statistically significant manner) formation of a SOST homodimer. Because the interaction between SOST and a BMP may involve a homodimer of SOST and a homodimer of the BMP, an antibody that prevents or impairs homodimer formation of SOST may thereby alter bone mineral density, preferably increasing bone mineral density. In one embodiment, antibodies that bind to the core region of SOST prevent homodimer formation.
  • Such antibodies may also bind to peptides that comprise contiguous amino acid sequences corresponding the core region, for example, SEQ ID NOS: 74, 75, and 98 (human SOST) and SEQ ID NOS: 76 and 99 (rat SOST).
  • Antibodies that bind to an epitope located on the C-terminal region of a SOST polypeptide may also impair homodimer formation.
  • Representative C-terminal polypeptides of human and rat SOST for example, comprise the amino acid sequences set forth in SEQ ID NO: 96 and SEQ ID NO: 97, respectively.
  • Such antibodies may also bind shorter polypeptide sequences.
  • Representative human SOST C-terminal peptide sequences are provided in SEQ ID NOS: 78-81 and representative rat SOST C-terminal sequences are provided in SEQ ID NOS: 86-88.
  • the SOST polypeptides and peptides disclosed herein to which antibodies may specifically bind are useful as immunogens. These immunogens of the present invention may be used for immunizing an animal to generate a humoral immune response that results in production of antibodies that specifically bind to a Type I or Type II receptor binding site or both located on a BMP include peptides derived from the N-terminal region of SOST or that may prevent SOST homodimer formation.
  • Such SOST polypeptides and peptides that are useful as immunogens may also be used in methods for screening samples containing antibodies, for example, samples of purified antibodies, antisera, or cell culture supernatants or any other biological sample that may contain one or more antibodies specific for SOST.
  • These peptides may also be used in methods for identifying and selecting from a biological sample one or more B cells that are producing an antibody that specifically binds to SOST (e.g., plaque forming assays and the like). The B cells may then be used as source of a SOST specific antibody-encoding polynucleotide that can be cloned and/or modified by recombinant molecular biology techniques known in the art and described herein.
  • a “biological sample” as used herein refers in certain embodiments to a sample containing at least one antibody specific for a SOST polypeptide, and a biological sample may be provided by obtaining a blood sample, biopsy specimen, tissue explant, organ culture, or any other tissue or cell preparation from a subject or a biological source.
  • a sample may further refer to a tissue or cell preparation in which the morphological integrity or physical state has been disrupted, for example, by dissection, dissociation, solubilization, fractionation, homogenization, biochemical or chemical extraction, pulverization, lyophilization, sonication, or any other means for processing a sample derived from a subject or biological source.
  • the subject or biological source may be a human or non-human animal, a primary cell culture (e.g., B cells immunized in vitro), or culture adapted cell line including but not limited to genetically engineered cell lines that may contain chromosomally integrated or episomal recombinant nucleic acid sequences, immortalized or immortalizable cell lines, somatic cell hybrid cell lines, differentiated or differentiatable cell lines, transformed cell lines, and the like.
  • a primary cell culture e.g., B cells immunized in vitro
  • culture adapted cell line including but not limited to genetically engineered cell lines that may contain chromosomally integrated or episomal recombinant nucleic acid sequences, immortalized or immortalizable cell lines, somatic cell hybrid cell lines, differentiated or differentiatable cell lines, transformed cell lines, and the like.
  • SOST peptide immunogens may also be prepared by synthesizing a series of peptides that, in total, represent the entire polypeptide sequence of a SOST polypeptide and that each have a portion of the SOST amino acid sequence in common with another peptide in the series. This overlapping portion would preferably be at least four amino acids, and more preferably 5, 6, 7, 8, 9, or 10 amino acids.
  • Each peptide may be used to immunize an animal, the sera collected from the animal, and tested in an assay to identify which animal is producing antibodies that impair or block binding of SOST to a TGF-beta protein. Antibodies are then prepared from such identified immunized animals according to methods known in the art and described herein.
  • Antibodies which inhibit the binding of TGF-beta binding-protein to a TGF-beta family member may readily be prepared given the disclosure provided herein. Particularly useful are anti-TGF-beta binding-protein antibodies that “specifically bind” TGF-beta binding-protein of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65, but not to other TGF-beta binding-proteins such as Dan, Cerberus, SCGF, or Gremlin.
  • antibodies are understood to include monoclonal antibodies, polyclonal antibodies, single chain, chimeric, CDR-grafted immunoglobulings, anti-idiotypic antibodies, and antibody fragments thereof (e.g., Fab, Fd, Fab′, and F(ab′) 2 , F V variable regions, or complementarity determining regions).
  • antibodies are understood to be specific against TGF-beta binding-protein, or against a specific TGF-beta family member, if they bind with a K a of greater than or equal to 10 7 M ⁇ 1 , preferably greater than or equal to 10 8 M ⁇ 1 , and do not bind to other TGF-beta binding-proteins, or bind with a K a of less than or equal to 10 6 M ⁇ 1 .
  • Affinity of an antibody for its cognate antigen is also commonly expressed as a dissociation constant K D
  • an anti-SOST antibody specifically binds to a TGF-beta family member if it binds with a K D of less than or equal to about 10 ⁇ 5 M, more preferably less than or equal to about 10 ⁇ 6 M, still more preferably less than or equal to 10 ⁇ 7 M, and still more preferably less than or equal to 10 ⁇ 8 M.
  • antibodies of the present invention preferably block, impair, or inhibit (e.g., decrease with statistical significance) the binding of TGF-beta binding-protein to a TGF-beta family member.
  • Affinity may also be determined by surface plasmon resonance (SPR; BIAcore, Biosensor, Piscataway, N.J.).
  • SPR surface plasmon resonance
  • target molecules are immobilized on a solid phase and exposed to ligands in a mobile phase running along a flow cell. If ligand binding to the immobilized target occurs, the local refractive index changes, leading to a change in SPR angle, which can be monitored in real time by detecting changes in the intensity of the reflected light.
  • the rates of change of the SPR signal can be analyzed to yield apparent rate constants for the association and dissociation phases of the binding reaction.
  • the ratio of these values gives the apparent equilibrium constant (affinity) (see, e.g., Wolff et al., Cancer Res. 53:2560-65 (1993)).
  • An antibody according to the present invention may belong to any immunoglobulin class, for example IgG, IgE, IgM, IgD, or IgA, and may be any one of the different isotypes that may comprise a class (such as IgG1, IgG2, IgG3, and IgG4 of the human IgG class). It may be obtained from or derived from an animal, for example, fowl (e.g., chicken) and mammals, which includes but is not limited to a mouse, rat, hamster, rabbit, or other rodent, a cow, horse, sheep, goat, camel, human, or other primate. The antibody may be an internalising antibody.
  • fowl e.g., chicken
  • mammals which includes but is not limited to a mouse, rat, hamster, rabbit, or other rodent, a cow, horse, sheep, goat, camel, human, or other primate.
  • the antibody may be an internalising antibody.
  • Antibodies may be produced as genetically engineered immunoglobulins (Ig) or Ig fragments designed to have desirable properties.
  • Ig immunoglobulins
  • antibodies may include a recombinant IgG that is a chimeric fusion protein having at least one variable (V) region domain from a first mammalian species and at least one constant region domain from a second, distinct mammalian species. Most commonly, a chimeric antibody has murine variable region sequences and human constant region sequences.
  • Such a murine/human chimeric immunoglobulin may be “humanized” by grafting the complementarity determining regions (CDRs) derived from a murine antibody, which confer binding specificity for an antigen, into human-derived V region framework regions and human-derived constant regions. Fragments of these molecules may be generated by proteolytic digestion, or optionally, by proteolytic digestion followed by mild reduction of disulfide bonds and alkylation. Alternatively, such fragments may also be generated by recombinant genetic engineering techniques.
  • CDRs complementarity determining regions
  • Certain preferred antibodies are those antibodies that inhibit or block a TGF-beta binding protein activity within an in vitro assay, as described herein. Binding properties of an antibody to a TGF-beta binding protein may generally be assessed using immunodetection methods including, for example, an enzyme-linked immunosorbent assay (ELISA), immunoprecipitation, immunoblotting, countercurrent immunoelectrophoresis, radioimmunoassays, dot blot assays, inhibition or competition assays, and the like, which may be readily performed by those having ordinary skill in the art (see, e.g., U.S. Pat. Nos. 4,376,110 and 4,486,530; Harlow et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory (1988)).
  • ELISA enzyme-linked immunosorbent assay
  • An immunogen may be comprised of cells expressing a TGF-beta binding protein, purified or partially purified TGF-beta binding polypeptides, or variants or fragments (i.e., peptides) thereof, or peptides derived from a TGF-beta binding protein.
  • Such peptides may be generated by proteolytic cleavage of a larger polypeptide, by recombinant molecular methodologies, or may be chemically synthesized.
  • nucleic acid sequences encoding TGF-beta binding proteins are provided herein, such that those skilled in the art may routinely prepare TGF-beta binding proteins for use as immunogens.
  • Peptides may be chemically synthesized by methods as described herein and known in the art. Alternatively, peptides may be generated by proteolytic cleavage of a TGF-beta binding protein, and individual peptides isolated by methods known in the art such as polyacrylamide gel electrophoresis or any number of liquid chromatography or other separation methods. Peptides useful as immunogens typically may have an amino acid sequence of at least 4 or 5 consecutive amino acids from a TGF-beta binding protein amino acid sequence such as those described herein, and preferably have at least 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 18, 19 or 20 consecutive amino acids of a TGF-beta binding protein.
  • Certain other preferred peptide immunogens comprise at least 6 but no more than 12 or more consecutive amino acids of a TGF-beta binding protein sequence, and other preferred peptide immunogens comprise at least 21 but no more than 50 consecutive amino acids of a SOST polypeptide.
  • Other preferred peptide immunogens comprise 21-25, 26-30, 31-35, 36-40, 41-50, or any whole integer number of amino acids between and including 21 and 100 consecutive amino acids, and between 100 and 190 consecutive amino acids of a TGF-beta binding protein sequence.
  • polyclonal antibodies may be readily generated by one of ordinary skill in the art from a variety of warm-blooded animals such as horses, cows, various fowl, rabbits, mice, sheep, goats, baboons, or rats.
  • the TGF-beta binding-protein or unique peptide thereof of 13-20 amino acids or as described herein is used to immunize the animal through intraperitoneal, intramuscular, intraocular, intradermal, or subcutaneous injections, along with an adjuvant such as Freund's complete or incomplete adjuvant, or the Ribi Adjuvant System (Corixa Corporation, Seattle, Was.). See also, e.g., Harlow et al., supra.
  • animals receive one or more booster immunizations according to a preferred schedule that may vary according to, inter alia, the antigen, the adjuvant (if any), and/or the particular animal species.
  • the immune response may be monitored by periodically bleeding the animal and preparing and analyzing sera in an immunoassay, such as an ELISA or Ouchterlony diffusion assay, or the like, to determine the specific antibody titer.
  • an immunoassay such as an ELISA or Ouchterlony diffusion assay, or the like
  • Particularly preferred polyclonal antisera will give a detectable signal on one of these assays, such as an ELISA, that is preferably at least three times greater than background.
  • Polyclonal antibodies that bind specifically to the TGF-beta binding protein or peptide may then be purified from such antisera, for example, by affinity chromatography using protein A.
  • affinity chromatography may be performed wherein the TGF-beta binding protein or peptide or an antibody specific for an Ig constant region of the particular immunized animal species is immobilized on a suitable solid support.
  • Antibodies for use in the invention include monoclonal antibodies that are prepared by conventional immunization and cell fusion procedures as described herein an known in the art. Monoclonal antibodies may be readily generated using conventional techniques (see, e.g., Kohler et al., Nature 256:495, 1975; Coligan et al. (eds.), Current Protocols in Immunology, 1:2.5.1-2.6.7 (John Wiley & Sons 1991) [“Coligan”]; U.S. Pat. Nos.
  • Antibody fragments may be derived therefrom using any suitable standard technique such as proteolytic digestion, or optionally, by proteolytic digestion (for example, using papain or pepsin) followed by mild reduction of disulfide bonds and alkylation. Alternatively, such fragments may also be generated by recombinant genetic engineering techniques.
  • a subject animal such as a rat or mouse or hamster is immunized with TGF-beta binding-protein or a portion of a region thereof, including peptides within a region, as described herein.
  • the protein may be admixed with an adjuvant such as Freund's complete or incomplete adjuvant or Ribi adjuvant in order to increase the resultant immune response.
  • an adjuvant such as Freund's complete or incomplete adjuvant or Ribi adjuvant
  • spleen and lymph nodes are harvested.
  • the harvested spleen and/or lymph node cell suspensions are fused with a suitable myeloma cell that is drug-sensitized in order to create a “hybridoma” which secretes monoclonal antibody.
  • suitable myeloma lines include, for example, NS-0, SP20, NS-1 (ATCC No. TIB 18), and P3X63-Ag 8.653 (ATCC No. CRL 1580).
  • the lymphoid (e.g., spleen) cells and the myeloma cells may be combined for a few minutes with a membrane fusion-promoting agent, such as polyethylene glycol or a nonionic detergent, and then plated at low density on a selective medium that supports the growth of hybridoma cells but not unfused myeloma cells.
  • a membrane fusion-promoting agent such as polyethylene glycol or a nonionic detergent
  • the cells may be placed into culture plates containing a suitable medium, such as RPMI 1640, or DMEM (Dulbecco's Modified Eagles Medium) (JRH Biosciences, Lenexa, Kans.), as well as additional ingredients, such as fetal bovine serum (FBS, i.e., from Hyclone, Logan, Utah, or JRH Biosciences). Additionally, the medium should contain a reagent which selectively allows for the growth of fused spleen and myeloma cells such as HAT (hypoxanthine, aminopterin, and thymidine) (Sigma Chemical Co., St. Louis, Mo.).
  • HAT hypoxanthine, aminopterin, and thymidine
  • the resulting fused cells or hybridomas may be screened in order to determine the presence of antibodies which are reactive with TGF-beta binding-protein (depending on the antigen used), and which block, impair, or inhibit the binding of TGF-beta binding-protein to a TGF-beta family member.
  • Hybridomas that produce monoclonal antibodies that specifically bind to sclerostin or a variant thereof are preferred.
  • a wide variety of assays may be utilized to determine the presence of antibodies which are reactive against the proteins of the present invention, including for example countercurrent immuno-electrophoresis, radioimmunoassays, radioimmunoprecipitations, enzyme-linked immuno-sorbent assays (ELISA), dot blot assays, western blots, immunoprecipitation, inhibition or competition assays, and sandwich assays (see U.S. Pat. Nos. 4,376,110 and 4,486,530; see also Antibodies: A Laboratory Manual, Harlow and Lane (eds.), Cold Spring Harbor Laboratory Press, 1988).
  • the hybridomas are cloned, for example, by limited dilution cloning or by soft agar plaque isolation, and reassayed. Thus, a hybridoma producing antibodies reactive against the desired protein may be isolated.
  • the monoclonal antibodies from the hybridoma cultures may be isolated from the supernatants of hybridoma cultures.
  • An alternative method for production of a murine monoclonal antibody is to inject the hybridoma cells into the peritoneal cavity of a syngeneic mouse, for example, a mouse that has been treated (e.g., pristane-primed) to promote formation of ascites fluid containing the monoclonal antibody.
  • Monoclonal antibodies can be isolated and purified by a variety of well-established techniques.
  • Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al., “Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)).
  • Monoclonal antibodies may be purified by affinity chromatography using an appropriate ligand selected based on particular properties of the antibody (e.g., heavy or light chain isotype, binding specificity, etc.).
  • Examples of a suitable ligand, immobilized on a solid support include Protein A, Protein G, an anti-constant region (light chain or heavy chain) antibody, an anti-idiotype antibody, and a TGF-beta binding protein, or fragment or variant thereof.
  • an anti-TGF-beta binding-protein antibody of the present invention may be a human monoclonal antibody.
  • Human monoclonal antibodies may be generated by any number of techniques with which those having ordinary skill in the art will be familiar. Such methods include, but are not limited to, Epstein Barr Virus (EBV) transformation of human peripheral blood cells (e.g., containing B lymphocytes), in vitro immunization of human B cells, fusion of spleen cells from immunized transgenic mice carrying inserted human immunoglobulin genes, isolation from human immunoglobulin V region phage libraries, or other procedures as known in the art and based on the disclosure herein.
  • EBV Epstein Barr Virus
  • human monoclonal antibodies may be obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge.
  • Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., Nature Genet. 7:13, 1994; Lonberg et al., Nature 368:856, 1994; Taylor et al., Int. Immun. 6:579, 1994; U.S. Pat. No. 5,877,397; Bruggemann et al., 1997 Curr. Opin. Biotechnol. 8:455-58; Jakobovits et al., 1995 Ann. N. Y. Acad. Sci. 764:525-35.
  • human immunoglobulin transgenes may be mini-gene constructs, or transloci on yeast artificial chromosomes, which undergo B cell-specific DNA rearrangement and hypermutation in the mouse lymphoid tissue.
  • Human monoclonal antibodies may be obtained by immunizing the transgenic mice, which may then produce human antibodies specific for the antigen. Lymphoid cells of the immunized transgenic mice can be used to produce human antibody-secreting hybridomas according to the methods described herein. Polyclonal sera containing human antibodies may also be obtained from the blood of the immunized animals.
  • Another method for generating human TGF-beta binding protein specific monoclonal antibodies includes immortalizing human peripheral blood cells by EBV transformation. See, e.g., U.S. Pat. No. 4,464,456.
  • Such an immortalized B cell line (or lymphoblastoid cell line) producing a monoclonal antibody that specifically binds to a TGF-beta binding protein (or a variant or fragment thereof) can be identified by immunodetection methods as provided herein, for example, an ELISA, and then isolated by standard cloning techniques.
  • the stability of the lymphoblastoid cell line producing an anti-TGF-beta binding protein antibody may be improved by fusing the transformed cell line with a murine myeloma to produce a mouse-human hybrid cell line according to methods known in the art (see, e.g., Glasky et al., Hybridoma 8:377-89 (1989)).
  • Still another method to generate human monoclonal antibodies is in vitro immunization, which includes priming human splenic B cells with antigen, followed by fusion of primed B cells with a heterohybrid fusion partner. See, e.g., Boerner et al., 1991 J Immunol. 147:86-95.
  • a B cell that is producing an anti-SOST antibody is selected and the light chain and heavy chain variable regions are cloned from the B cell according to molecular biology techniques known in the art (WO 92/02551; U.S. Pat. No. 5,627,052; Babcook et al., Proc. Natl. Acad. Sci. USA 93:7843-48 (1996)) and described herein.
  • B cells from an immunized animal are isolated from the spleen, lymph node, or peripheral blood sample by selecting a cell that is producing an antibody that specifically binds to SOST.
  • B cells may also be isolated from humans, for example, from a peripheral blood sample.
  • Methods for detecting single B cells that are producing an antibody with the desired specificity are well known in the art, for example, by plaque formation, fluorescence-activated cell sorting, in vitro stimulation followed by detection of specific antibody, and the like.
  • Methods for selection of specific antibody producing B cells include, for example, preparing a single cell suspension of B cells in soft agar that contains SOST or a peptide fragment thereof. Binding of the specific antibody produced by the B cell to the antigen results in the formation of a complex, which may be visible as an immunoprecipitate. After the B cells producing the specific antibody are selected, the specific antibody genes may be cloned by isolating and amplifying DNA or mRNA according to methods known in the art and described herein.
  • fragments of anti-TGF-beta binding protein antibodies may be desired.
  • Antibody fragments, F(ab′) 2 , Fab, Fab′, Fv, Fe, Fd, retain the antigen binding site of the whole antibody and therefore bind to the same epitope.
  • These antigen-binding fragments derived from an antibody can be obtained, for example, by proteolytic hydrolysis of the antibody, for example, pepsin or papain digestion of whole antibodies according to conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′) 2 .
  • This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab′ monovalent fragments.
  • the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages.
  • an enzymatic cleavage using papain produces two monovalent Fab fragments and an Fc fragment directly.
  • an antibody fragment may also be any synthetic or genetically engineered protein that acts like an antibody in that it binds to a specific antigen to form a complex.
  • antibody fragments include isolated fragments consisting of the light chain variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (scFv proteins), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • the antibody of the present invention preferably comprises at least one variable region domain.
  • variable region domain may be of any size or amino acid composition and will generally comprise at least one hypervariable amino acid sequence responsible for antigen binding and which is adjacent to or in frame with one or more framework sequences.
  • the variable (V) region domain may be any suitable arrangement of immunoglobulin heavy (V H ) and/or light (V L ) chain variable domains.
  • V H immunoglobulin heavy
  • V L light chain variable domains.
  • the V region domain may be monomeric and be a V H or V L domain, which is capable of independently binding antigen with acceptable affinity.
  • the V region domain may be dimeric and contain V H -V H , V H -V L , or V L -V L , dimers.
  • the V region dimer comprises at least one V H and at least one V L chain that are non-covalently associated (hereinafter referred to as F v ).
  • the chains may be covalently coupled either directly, for example via a disulphide bond between the two variable domains, or through a linker, for example a peptide linker, to form a single chain Fv (scF v ).
  • variable region domain may be any naturally occurring variable domain or an engineered version thereof.
  • engineered version is meant a variable region domain that has been created using recombinant DNA engineering techniques.
  • engineered versions include those created, for example, from a specific antibody variable region by insertions, deletions, or changes in or to the amino acid sequences of the specific antibody.
  • Particular examples include engineered variable region domains containing at least one CDR and optionally one or more framework amino acids from a first antibody and the remainder of the variable region domain from a second antibody.
  • variable region domain may be covalently attached at a C-terminal amino acid to at least one other antibody domain or a fragment thereof.
  • a V H , domain that is present in the variable region domain may be linked to an immunoglobulin C H 1 domain, or a fragment thereof.
  • a V L domain may be linked to a C K domain or a fragment thereof.
  • the antibody may be a Fab fragment wherein the antigen binding domain contains associated V H and V L domains covalently linked at their C-termini to a CH1 and C K domain, respectively.
  • the CH1 domain may be extended with further amino acids, for example to provide a hinge region or a portion of a hinge region domain as found in a Fab′ fragment, or to provide further domains, such as antibody CH2 and CH3 domains.
  • CDR peptides (“minimal recognition units”) can be obtained by constructing polynucleotides that encode the CDR of an antibody of interest. Such polynucleotides are prepared, for example, by using the polymerase chain reaction to synthesize the variable region using mRNA of antibody-producing cells as a template (see, for example, Larrick et al., Methods: A Companion to Methods in Enzymology 2:106, 1991; Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies,” in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al.
  • the antibody may be a recombinant or engineered antibody obtained by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions.
  • DNA is known and/or is readily available from DNA libraries including for example phage-antibody libraries (see Chiswell and McCafferty, Tibtech. 10:80-84 (1992)) or if desired can be synthesized. Standard molecular biology and/or chemistry procedures may be used to sequence and manipulate the DNA, for example, to introduce codons to create cysteine residues, or to modify, add or delete other amino acids or domains as desired.
  • Chimeric antibodies specific for a TGF-beta binding protein, and which include humanized antibodies, may also be generated according to the present invention.
  • a chimeric antibody has at least one constant region domain derived from a first mammalian species and at least one variable region domain derived from a second, distinct mammalian species (see, e.g., Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-55 (1984)).
  • a chimeric antibody may be constructed by cloning the polynucleotide sequence that encodes at least one variable region domain derived from a non-human monoclonal antibody, such as the variable region derived from a murine, rat, or hamster monoclonal antibody, into a vector containing a nucleotide sequence that encodes at least one human constant region (see, e.g., Shin et al., Methods Enzymol. 178:459-76 (1989); Walls et al., Nucleic Acids Res. 21:2921-29 (1993)).
  • the polynucleotide sequence encoding the light chain variable region of a murine monoclonal -antibody may be inserted into a vector containing a nucleotide sequence encoding the human kappa light chain constant region sequence.
  • the polynucleotide sequence encoding the heavy chain variable region of the monoclonal antibody may be cloned in frame with sequences encoding a human IgG constant region, for example, the human IgG1 constant region.
  • the particular human constant region selected may depend upon the effector functions desired for the particular antibody (e.g. complement fixing, binding to a particular Fc receptor, etc.).
  • the constructed vectors will be transfected into eukaryotic cells for stable expression of the chimeric antibody.
  • Another method known in the art for generating chimeric antibodies is homologous recombination (e.g., U.S. Pat. No. 5,482,856).
  • a non-human/human chimeric antibody may be further genetically engineered to create a “humanized” antibody.
  • a humanized antibody may comprise a plurality of CDRs derived from an immunoglobulin of a non-human mammalian species, at least one human variable framework region, and at least one human immunoglobulin constant region.
  • Useful strategies for designing humanized antibodies may include, for example by way of illustration and not limitation, identification of human variable framework regions that are most homologous to the non-human framework regions of the chimeric antibody.
  • such a strategy may increase the likelihood that the humanized antibody will retain specific binding affinity for a TGF-beta binding protein, which in some preferred embodiments may be substantially the same affinity for a TGF-beta binding protein or variant or fragment thereof, and in certain other preferred embodiments may be a greater affinity for TGF-beta binding protein. See, e.g., Jones et al., 1986 Nature 321:522-25; Riechmann et al., 1988 Nature 332:323-27.
  • Designing such a humanized antibody may therefore include determining CDR loop conformations and structural determinants of the non-human variable regions, for example, by computer modeling, and then comparing the CDR loops and determinants to known human CDR loop structures and determinants. See, e.g., Padlan et al., 1995 FASEB 9:133-39; Chothia et al., 1989 Nature, 342:377-383. Computer modeling may also be used to compare human structural templates selected by sequence homology with the non-human variable regions. See, e.g., Bajorath et al., 1995 Ther. Immunol. 2:95-103; EP-0578515-A3.
  • veneering One such method for preparing a humanized antibody is called veneering.
  • the terms “veneered FRs” and “recombinantly veneered FRs” refer to the selective replacement of FR residues from, e.g., a rodent heavy or light chain V region, with human FR residues in order to provide a xenogeneic molecule comprising an antigen-binding site that retains substantially all of the native FR polypeptide folding structure.
  • Veneering techniques are based on the understanding that the ligand binding characteristics of an antigen-binding site are determined primarily by the structure and relative disposition of the heavy and light chain CDR sets within the antigen-binding surface. Davies et al., Ann. Rev. Biochem.
  • antigen binding specificity can be preserved in a humanized antibody only wherein the CDR structures, their interaction with each other, and their interaction with the rest of the V region domains are carefully maintained.
  • exterior (e.g., solvent-accessible) FR residues that are readily encountered by the immune system are selectively replaced with human residues to provide a hybrid molecule that comprises either a weakly immunogenic, or substantially non-immunogenic veneered surface.
  • the process of veneering makes use of the available sequence data for human antibody variable domains compiled by Kabat et al., in Sequences of Proteins of Immunological Interest, 4th ed., (U.S. Dept. of Health and Human Services, U.S. Government Printing Office, 1987), updates to the Kabat database, and other accessible U.S. and foreign databases (both nucleic acid and protein). Solvent accessibilities of V region amino acids can be deduced from the known three-dimensional structure for human and murine antibody fragments. Initially, the FRs of the variable domains of an antibody molecule of interest are compared with corresponding FR sequences of human variable domains obtained from the above-identified sources.
  • the most homologous human V regions are then compared residue by residue to corresponding murine amino acids.
  • the residues in the murine FR that differ from the human counterpart are replaced by the residues present in the human moiety using recombinant techniques well known in the art. Residue switching is only carried out with moieties which are at least partially exposed (solvent accessible), and care is exercised in the replacement of amino acid residues that may have a significant effect on the tertiary structure of V region domains, such as proline, glycine, and charged amino acids.
  • the resultant “veneered” antigen-binding sites are thus designed to retain the rodent CDR residues, the residues substantially adjacent to the CDRs, the residues identified as buried or mostly buried (solvent inaccessible), the residues believed to participate in non-covalent (e.g., electrostatic and hydrophobic) contacts between heavy and light chain domains, and the residues from conserved structural regions of the FRs which are believed to influence the “canonical” tertiary structures of the CDR loops.
  • An additional method for selecting antibodies that specifically bind to a TGF-beta binding protein or variant or fragment thereof is by phage display. See, e.g., Winter et al., 1994 Annu. Rev. Immunol. 12:433-55; Burton et al., 1994 Adv. Immunol. 57:191-280.
  • Human or murine immunoglobulin variable region gene combinatorial libraries may be created in phage vectors that can be screened to select Ig fragments (Fab, Fv, sFv, or multimers thereof) that bind specifically to TGF-beta binding protein or variant or fragment thereof. See, e.g., U.S. Pat. No. 5,223,409; William D.
  • mRNA is isolated from a B cell population, and utilized to create heavy and light chain immunoglobulin cDNA expression libraries in the ⁇ ImmunoZap(H) and ⁇ ImmunoZap(L) vectors. Positive plaques may subsequently be converted to a non-lytic plasmid which allows high level expression of monoclonal antibody fragments from E. coli.
  • a library containing a plurality of polynucleotide sequences encoding Ig variable region fragments may be inserted into the genome of a filamentous bacteriophage, such as M13 or a variant thereof, in frame with the sequence encoding a phage coat protein.
  • a fusion protein may be a fusion of the coat protein with the light chain variable region domain and/or with the heavy chain variable region domain.
  • immunoglobulin Fab fragments may also be displayed on a phage particle (see, e.g., U.S. Pat. No. 5,698,426). These vectors may be screened individually or co-expressed to form Fab fragments or antibodies (see Huse et al., supra; see also Sastry et al., supra).
  • portions or fragments, such as Fab and Fv fragments, of antibodies may also be constructed utilizing conventional enzymatic digestion or recombinant DNA techniques to incorporate the variable regions of a gene which encodes a specifically binding antibody.
  • the genes which encode the variable region from a hybridoma producing a monoclonal antibody of interest are amplified using nucleotide primers for the variable region. These primers may be synthesized by one of ordinary skill in the art, or may be purchased from commercially available sources.
  • Stratagene sells primers for mouse and human variable regions including, among others, primers for V Ha , V Hb , V Hc , V Hd , C H1 , V L and C L regions. These primers may be utilized to amplify heavy or light chain variable regions, which may then be inserted into vectors such as ImmunoZAPTM H or ImmunoZAPTM L (Stratagene), respectively. These vectors may then be introduced into E. coli, yeast, or mammalian-based systems for expression. Utilizing these techniques, large amounts of a single-chain protein containing a fusion of the V H and V L domains may be produced (see Bird et al., Science 242:423-426, 1988). In addition, such techniques may be utilized to change a “murine” antibody to a “human” antibody, without altering the binding specificity of the antibody.
  • combinatorial phage libraries may also be used for humanization of non-human variable regions. See, e.g., Rosok et al., 1996 J. Biol. Chem. 271:22611-18; Rader et al., 1998 Proc. Natl. Acad. Sci. USA 95:8910-15.
  • a phage library may be screened to select an Ig variable region fragment of interest by immunodetection methods known in the art and described herein, and the DNA sequence of the inserted immunoglobulin gene in the phage so selected may be determined by standard techniques. See, Sambrook et al., 2001 Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press.
  • the selected Ig-encoding sequence may then be cloned into another suitable vector for expression of the Ig fragment or, optionally, may be cloned into a vector containing Ig constant regions, for expression of whole immunoglobulin chains.
  • the invention contemplates SOST-specific antibodies that are multimeric antibody fragments.
  • Useful methodologies are described generally, for example in Hayden et al. 1997, Curr Opin. Immunol. 9:201-12; Coloma et al., 1997 Nat. Biotechnol. 15:159-63).
  • multimeric antibody fragments may be created by phage techniques to form miniantibodies (U.S. Pat. No. 5,910,573) or diabodies (Holliger et al., 1997, Cancer Immunol. Immunother. 45:128-130).
  • an antibody specific for SOST may be an antibody that is expressed as an intracellular protein.
  • intracellular antibodies are also referred to as intrabodies and may comprise an Fab fragment, or preferably comprise a scFv fragment (see, e.g., Lecerf et al., Proc. Natl. Acad. Sci. USA 98:4764-49 (2001).
  • the framework regions flanking the CDR regions can be modified to improve expression levels and solubility of an intrabody in an intracellular reducing environment (see, e.g., Worn et al., J. Biol. Chem. 275:2795-803 (2000).
  • An intrabody may be directed to a particular cellular location or organelle, for example by constructing a vector that comprises a polynucleotide sequence encoding the variable regions of an intrabody that may be operatively fused to a polynucleotide sequence that encodes a particular target antigen within the cell (see, e.g., Graus-Porta et al., Mol. Cell Biol. 15:1182-91 (1995); Lener et al., Eur. J. Biochem. 267:1196-205 (2000)).
  • An intrabody may be introduced into a cell by a variety of techniques available to the skilled artisan including via a gene therapy vector, or a lipid mixture (e.g., ProvectinTM manufactured by Imgenex Corporation, San Diego, Calif.), or according to photochemical internalization methods.
  • a gene therapy vector or a lipid mixture (e.g., ProvectinTM manufactured by Imgenex Corporation, San Diego, Calif.), or according to photochemical internalization methods.
  • Introducing amino acid mutations into an immunoglobulin molecule specific for a TGF-beta binding protein may be useful to increase the specificity or affinity for TGF-beta binding protein or to alter an effector function.
  • Immunoglobulins with higher affinity for TGF-beta binding protein may be generated by site-directed mutagenesis of particular residues.
  • Computer assisted three-dimensional molecular modeling may be employed to identify the amino acid residues to be changed, in order to improve affinity for the TGF-beta binding protein. See, e.g., Mountain et al., 1992, Biotechnol. Genet Eng. Rev. 10: 1-142.
  • combinatorial libraries of CDRs may be generated in M13 phage and screened for immunoglobulin fragments with improved affinity. See, e.g., Glaser et al., 1992, J. Immunol. 149:3903-3913; Barbas et al., 1994 Proc. Natl. Acad. Sci. USA 91:3809-13; U.S. Pat. No. 5,792,456.
  • Effector functions may also be altered by site-directed mutagenesis. See, e.g., Duncan et al., 1988 Nature 332:563-64; Morgan et al., 1995 Immunology 86:319-24; Eghtedarzedeh-Kondri et al., 1997 Biotechniques 23:830-34.
  • mutation of the glycosylation site on the Fc portion of the immunoglobulin may alter the ability of the immunoglobulin to fix complement.
  • Other mutations in the constant region domains may alter the ability of the immunoglobulin to fix complement, or to effect antibody-dependent cellular cytotoxicity.
  • non-human, human, or humanized heavy chain and light chain variable regions of any of the Ig molecules described herein may be constructed as single chain Fv (scFv) polypeptide fragments (single chain antibodies).
  • scFv single chain Fv
  • Multi-functional scFv fusion proteins may be generated by linking a polynucleotide sequence encoding an scFv polypeptide in-frame with at least one polynucleotide sequence encoding any of a variety of known effector proteins.
  • effector proteins may include immunoglobulin constant region sequences. See, e.g., Hollenbaugh et al., 1995 J. Immunol. Methods 188:1-7.
  • effector proteins are enzymes. As a non-limiting example, such an enzyme may provide a biological activity for therapeutic purposes (see, e.g., Siemers et al., 1997 Bioconjug. Chem.
  • scFv fusion proteins include Ig-toxin fusions, or immunotoxins, wherein the scFv polypeptide is linked to a toxin.
  • the scFv or any antibody fragment described herein may, in certain embodiments, be fused to peptide or polypeptide domains that permits detection of specific binding between the fusion protein and antigen (e.g., a TGF-beta binding protein).
  • the fusion polypeptide domain may be an affinity tag polypeptide for detecting binding of the scFv fusion protein to a TGF-beta binding protein by any of a variety of techniques with which those skilled in the art will be familiar.
  • a peptide tag include avidin, streptavidin or His (e.g., polyhistidine).
  • Detection techniques may also include, for example, binding of an avidin or streptavidin fusion protein to biotin or to a biotin mimetic sequence (see, e.g., Luo et al., 1998 J. Biotechnol. 65:225 and references cited therein), direct covalent modification of a fusion protein with a detectable moiety (e.g., a labeling moiety), non-covalent binding of the fusion protein to a specific labeled reporter molecule, enzymatic modification of a detectable substrate by a fusion protein that includes a portion having enzyme activity, or immobilization (covalent or non-covalent) of the fusion protein on a solid-phase support.
  • a detectable moiety e.g., a labeling moiety
  • enzymatic modification of a detectable substrate by a fusion protein that includes a portion having enzyme activity enzymatic modification of a detectable substrate by a fusion protein that includes a portion having enzyme activity, or im
  • affinity polypeptides for construction of scFv fusion proteins may include streptavidin fusion proteins, as disclosed, for example, in WO 89/03422, U.S. Pat. No. 5,489,528, U.S. Pat. No. 5,672,691, WO 93/24631, U.S. Pat. No. 5,168,049, U.S. Pat. No. 5,272,254; avidin fusion proteins (see, e.g., EP 511,747); an enzyme such as glutathione-S-transferase; and Staphylococcus aureus protein A polypeptide.
  • the polynucleotides encoding an antibody or fragment thereof that specifically bind a TGF-beta binding protein, as described herein, may be propagated and expressed according to any of a variety of well-known procedures for nucleic acid excision, ligation, transformation, and transfection using any number of known expression vectors.
  • expression of an antibody fragment may be preferred in a prokaryotic host, such as Escherichia coli (see, e.g., Pluckthun et al., 1989 Methods Enzymol. 178:497-515).
  • expression of the antibody or a fragment thereof may be preferred in a eukaryotic host cell, including yeast (e.g., Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Pichia pastoris ), animal cells (including mammalian cells) or plant cells.
  • yeast e.g., Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Pichia pastoris
  • animal cells including mammalian cells
  • suitable animal cells include, but are not limited to, myeloma (such as a mouse NSO line), COS, CHO, or hybridoma cells.
  • plant cells include tobacco, corn, soybean, and rice cells.
  • suitable antibodies may be isolated or purified by many techniques well known to those of ordinary skill in the art (see Antibodies: A Laboratory Manual, Harlow and Lane (eds.), Cold Spring Harbor Laboratory Press, 1988). Suitable techniques include peptide or protein affinity columns (including use of anti-constant region antibodies attached to the column matrix), HPLC or RP-HPLC, purification on protein A or protein G columns, or any combination of these techniques.
  • TGF-beta binding-protein As described herein and below in the Examples (e.g., Examples 8 and 9), altered versions of TGF-beta binding-protein which compete with native TGF-beta binding-protein's ability to block the activity of a particular TGF-beta family member should lead to increased bone density.
  • mutants of TGF-beta binding-protein which bind to the TGF-beta family member but do not inhibit the function of the TGF-beta family member would meet the criteria.
  • the mutant versions must effectively compete with the endogenous inhibitory functions of TGF-beta binding-protein.
  • Polypeptides described herein include the TGF binding protein sclerostin and variants thereof and antibodies or fragments thereof that specifically bind to sclerostin.
  • the polynucleotides that encode these polypeptides include derivatives of the genes that are substantially similar to the genes and isolated nucleic acid molecules, and, when appropriate, the proteins (including peptides and polypeptides) that are encoded by the genes and their derivatives.
  • nucleotide sequence is deemed to be “substantially similar” if (a) the nucleotide sequence is derived from the coding region of the above-described genes and nucleic acid molecules and includes, for example, portions of the sequence or allelic variations of the sequences discussed above, or alternatively, encodes a molecule which inhibits the binding of TGF-beta binding-protein to a member of the TGF-beta family; (b) the nucleotide sequence is capable of hybridization to nucleotide sequences of the present invention under moderate, high or very high stringency (see Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, NY, 1989); and/or (c) the DNA sequences are degenerate as a result of the genetic code to the DNA sequences defined in (a) or (b).
  • nucleic acid molecule disclosed herein includes both complementary and non-complementary sequences, provided the sequences otherwise meet the criteria set forth herein.
  • high stringency means standard hybridization conditions (e.g., 5 ⁇ SSPE, 0.5% SDS at 65° C., or the equivalent).
  • the structure of the proteins encoded by the nucleic acid molecules described herein may be predicted from the primary translation products using the hydrophobicity plot function of, for example, P/C Gene or Intelligenetics Suite (Intelligenetics, Mountain View, Calif.), or according to the methods described by Kyte and Doolittle ( J. Mol. Biol. 157:105-132, 1982).
  • Proteins of the present invention may be prepared in the form of acidic or basic salts, or in neutral form.
  • individual amino acid residues may be modified by oxidation or reduction.
  • various substitutions, deletions, or additions may be made to the amino acid or nucleic acid sequences, the net effect of which is to retain or further enhance or decrease the biological activity of the mutant or wild-type protein.
  • due to degeneracy in the genetic code for example, there may be considerable variation in nucleotide sequences encoding the same amino acid sequence.
  • proteins disclosed herein include conjugates of the proteins along with other proteins or polypeptides. This may be accomplished, for example, by the synthesis of N-terminal or C-terminal fusion proteins which may be added to facilitate purification or identification of proteins (see U.S. Pat. No. 4,851,341, see also, Hopp et al., Bio/Technology 6:1204, 1988.) Alternatively, fusion proteins such as Flag®/TGF-beta binding-protein be constructed in order to assist in the identification, expression, and analysis of the protein.
  • Proteins of the present invention may be constructed using a wide variety of techniques described herein. Further, mutations may be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a derivative having the desired amino acid insertion, substitution, or deletion.
  • oligonucleotide-directed site-specific (or segment specific) mutagenesis procedures may be employed to provide an altered gene or nucleic acid molecule having particular codons altered according to the substitution, deletion, or insertion required.
  • Exemplary methods of making the alterations set forth above are disclosed by Walder et al. ( Gene 42:133, 1986); Bauer et al. ( Gene 37:73, 1985); Craik ( BioTechniques, January 1985, 12-19); Smith et al. ( Genetic Engineering: Principles and Methods, Plenum Press, 1981); and Sambrook et al. (supra).
  • Deletion or truncation derivatives of proteins may also be constructed by utilizing convenient restriction endonuclease sites adjacent to the desired deletion. Subsequent to restriction, overhangs may be filled in and the DNA religated. Exemplary methods of making the alterations set forth above are disclosed by Sambrook et al. ( Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, 1989).
  • Mutations which are made in the nucleic acid molecules of the present invention preferably preserve the reading frame of the coding sequences. Furthermore, the mutations will preferably not create complementary regions that when transcribed could hybridize to produce secondary mRNA structures, such as loops or hairpins, that would adversely affect translation of the mRNA. Although a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutants screened for gain or loss or retention of biological activity.
  • mutations may be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a derivative having the desired amino acid insertion, substitution, or deletion.
  • Nucleic acid molecules which encode proteins of the present invention may also be constructed utilizing techniques such as PCR mutagenesis, chemical mutagenesis (Drinkwater and Klinedinst, PNAS 83:3402-3406, 1986), by forced nucleotide misincorporation (e.g., Liao and Wise Gene 88:107-111, 1990), or by use of randomly mutagenized oligonucleotides (Horwitz et al., Genome 3:112-117, 1989).
  • the present invention also provides for the manipulation and expression of the above described genes and nucleic acid molecules by culturing host cells containing a vector capable of expressing the above-described genes.
  • vectors or vector constructs include either synthetic or cDNA-derived nucleic acid molecules encoding the desired protein, which are operably linked to suitable transcriptional or translational regulatory elements.
  • Suitable regulatory elements may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, insect, or plant genes. Selection of appropriate regulatory elements is dependent on the host cell chosen, and may be readily accomplished by one of ordinary skill in the art. Examples of regulatory elements include a transcriptional promoter and enhancer or RNA polymerase binding sequence, a transcriptional terminator, and a ribosomal binding sequence, including a translation initiation signal.
  • Nucleic acid molecules that encode any of the proteins described above may be readily expressed by a wide variety of prokaryotic and eukaryotic host cells, including bacterial, mammalian, yeast or other fungi, viral, insect, or plant cells. Methods for transforming or transfecting such cells to express foreign DNA are well known in the art (see, e.g., Itakura et al., U.S. Pat. No. 4,704,362; Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929-1933, 1978; Murray et al., U.S. Pat. No. 4,801,542; Upshall et al., U.S. Pat. No.
  • Bacterial host cells suitable for carrying out the present invention include E. coli, B. subtilis, Salmonella typhimurium, and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, as well as many other bacterial species well known to one of ordinary skill in the art and described herein.
  • a representative example of a bacterial host cell includes E. coli DH5 ⁇ (Stratagene, La Jolla, Calif.).
  • Bacterial expression vectors preferably comprise a promoter which functions in the host cell, one or more selectable phenotypic markers, and a bacterial origin of replication.
  • Representative promoters include the ⁇ -lactamase (penicillinase) and lactose promoter system (see Chang et al., Nature 275:615, 1978), the T7 RNA polymerase promoter (Studier et al., Meth. Enzymol. 185:60-89, 1990), the lambda promoter (Elvin et al., Gene 87:123-126, 1990), the trp promoter (Nichols and Yanofsky, Meth.
  • telomeres are well known in the art, including among others, pBR322 (see Bolivar et al., Gene 2:95, 1977), the pUC plasmids pUC18, pUC19, pUC118, pUC119 (see Messing, Meth.
  • Yeast and fungi host cells suitable for carrying out the present invention include, among others, Saccharomyces pombe, Saccharomyces cerevisiae, the genera Pichia or Kluyveromyces and various species of the genus Aspergillus (McKnight et al., U.S. Pat. No. 4,935,349).
  • Suitable expression vectors for yeast and fungi include, among others, YCp50 (ATCC No. 37419) for yeast, and the amdS cloning vector pV3 (Turnbull, Bio/Technology 7:169, 1989), YRp7 (Struhl et al., Proc. Natl. Acad. Sci.
  • promoters for use in yeast include promoters from yeast glycolytic genes (Hitzeman et al., J. Biol. Chem. 255:12073-12080, 1980; Alber and Kawasaki, J. Mol. Appl. Genet. 1:419-434, 1982) or alcohol dehydrogenase genes (Young et al., in Genetic Engineering of Microorganisms for Chemicals, Hollaender et al. (eds.), p. 355, Plenum, New York, 1982; Ammerer, Meth. Enzymol. 101:192-201, 1983).
  • useful promoters for fungi vectors include those derived from Aspergillus nidulans glycolytic genes, such as the adh3 promoter (McKnight et al., EMBO J 4:2093-2099, 1985).
  • the expression units may also include a transcriptional terminator.
  • An example of a suitable terminator is the adh3 terminator (McKnight et al., supra, 1985).
  • the yeast vectors will generally include a selectable marker, which may be one of any number of genes that exhibit a dominant phenotype for which a phenotypic assay exists to enable transformants to be selected.
  • selectable markers are those that complement host cell auxotrophy, provide antibiotic resistance, or enable a cell to utilize specific carbon sources, and include leu2 (Broach et al., ibid.), ura3 (Botstein et al., Gene 8:17, 1979), or his3 (Struhl et al., ibid.).
  • Another suitable selectable marker is the cat gene, which confers chloramphenicol resistance on yeast cells.
  • Protocols for the transformation of yeast are also well known to those of ordinary skill in the art.
  • transformation may be readily accomplished either by preparation of spheroplasts of yeast with DNA (see Hinnen et al., PNAS USA 75:1929, 1978) or by treatment with alkaline salts such as LiCl (see Itoh et al., J. Bacteriology 153:163, 1983). Transformation of fungi may also be carried out using polyethylene glycol as described by Cullen et al. ( Bio/Technology 5:369, 1987).
  • Viral vectors include those that comprise a promoter that directs the expression of an isolated nucleic acid molecule that encodes a desired protein as described above.
  • a wide variety of promoters may be utilized within the context of the present invention, including for example, promoters such as MoMLV LTR, RSV LTR, Friend MuLV LTR, adenoviral promoter (Ohno et al., Science 265:781-784, 1994), neomycin phosphotransferase promoter/enhancer, late parvovirus promoter (Koering et al., Hum. Gene Therap.
  • the promoter is a tissue-specific promoter (see e.g., WO 91/02805; EP 0,415,731; and WO 90/07936).
  • tissue specific promoters include neural specific enolase promoter, platelet derived growth factor beta promoter, bone morphogenic protein promoter, human alpha1-chimaerin promoter, synapsin I promoter and synapsin II promoter.
  • viral-specific promoters e.g., retroviral promoters (including those noted above, as well as others such as HIV promoters), hepatitis, herpes (e.g., EBV), and bacterial, fungal or parasitic (e.g., malarial)-specific promoters may be utilized in order to target a specific cell or tissue which is infected with a virus, bacteria, fungus, or parasite.
  • retroviral promoters including those noted above, as well as others such as HIV promoters
  • hepatitis e.g., herpes (e.g., EBV)
  • bacterial, fungal or parasitic e.g., malarial
  • Mammalian cells suitable for carrying out the present invention include, among others COS, CHO, SaOS, osteosarcomas, KS483, MG-63, primary osteoblasts, and human or mammalian bone marrow stroma.
  • Mammalian expression vectors for use in carrying out the present invention will include a promoter capable of directing the transcription of a cloned gene, nucleic acid molecule, or cDNA.
  • Preferred promoters include viral promoters and cellular promoters.
  • Bone specific promoters include the promoter for bone sialo-protein and the promoter for osteocalcin.
  • Viral promoters include the cytomegalovirus immediate early promoter (Boshart et al., Cell 41:521-530, 1985), cytomegalovirus immediate late promoter, SV40 promoter (Subramani et al., Mol. Cell. Biol. 1:854-864, 1981), MMTV LTR, RSV LTR, metallothionein-1, adenovirus E1a.
  • Cellular promoters include the mouse metallothionein-1 promoter (Palmiter et al., U.S. Pat. No. 4,579,821), a mouse V ⁇ promoter (Bergman et al., Proc. Natl Acad. Sci.
  • Such expression vectors may also contain a set of RNA splice sites located downstream from the promoter and upstream from the DNA sequence encoding the peptide or protein of interest. Preferred RNA splice sites may be obtained from adenovirus and/or immunoglobulin genes. Also contained in the expression vectors is a polyadenylation signal located downstream of the coding sequence of interest. Suitable polyadenylation signals include the early or late polyadenylation signals from SV40 (Kaufman and Sharp, ibid.), the polyadenylation signal from the Adenovirus 5 E1B region and the human growth hormone gene terminator (DeNoto et al., Nucleic Acids Res. 9:3719-3730, 1981).
  • the expression vectors may include a noncoding viral leader sequence, such as the Adenovirus 2 tripartite leader, located between the promoter and the RNA splice sites.
  • Preferred vectors may also include enhancer sequences, such as the SV40 enhancer.
  • Expression vectors may also include sequences encoding the adenovirus VA RNAs. Suitable expression vectors can be obtained from commercial sources (e.g., Stratagene, La Jolla, Calif.).
  • Vector constructs comprising cloned DNA sequences can be introduced into cultured mammalian cells by, for example, calcium phosphate-mediated transfection (Wigler et al., Cell 14:725, 1978; Corsaro and Pearson, Somatic Cell Genetics 7:603, 1981; Graham and Van der Eb, Virology 52:456, 1973), electroporation (Neumann et al., EMBO J. 1:841-845, 1982), or DEAE-dextran mediated transfection (Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley and Sons, Inc., NY, 1987).
  • a selectable marker is generally introduced into the cells along with the gene or cDNA of interest.
  • Preferred selectable markers for use in cultured mammalian cells include genes that confer resistance to drugs, such as neomycin, hygromycin, and methotrexate.
  • the selectable marker may be an amplifiable selectable marker.
  • Preferred amplifiable selectable markers are the DHFR gene and the neomycin resistance gene. Selectable markers are reviewed by Thilly ( Mammalian Cell Technology, Butterworth Publishers, Stoneham, Mass., which is incorporated herein by reference).
  • Mammalian cells containing a suitable vector are allowed to grow for a period of time, typically 1-2 days, to begin expressing the DNA sequence(s) of interest. Drug selection is then applied to select for growth of cells that are expressing the selectable marker in a stable fashion. For cells that have been transfected with an amplifiable, selectable marker, the drug concentration may be increased in a stepwise manner to select for increased copy number of the cloned sequences, thereby increasing expression levels. Cells expressing the introduced sequences are selected and screened for production of the protein of interest in the desired form or at the desired level. Cells that satisfy these criteria can then be cloned and scaled up for production.
  • Protocols for the transfection of mammalian cells are well known to those of ordinary skill in the art. Representative methods include calcium phosphate mediated transfection, electroporation, lipofection, retroviral, adenoviral and protoplast fusion-mediated transfection (see Sambrook et al., supra). Naked vector constructs can also be taken up by muscular cells or other suitable cells subsequent to injection into the muscle of a mammal (or other animals).
  • proteins of the present invention may be expressed in a transgenic animal whose germ cells and somatic cells contain a gene which encodes the desired protein and which is operably linked to a promoter effective for the expression of the gene.
  • transgenic animals may be prepared that lack the desired gene (e.g., “knock-out” mice).
  • Such transgenics may be prepared in a variety of non-human animals, including mice, rats, rabbits, sheep, dogs, goats, and pigs (see Hammer et al., Nature 315:680-683, 1985, Palmiter et al., Science 222:809-814, 1983, Brinster et al., Proc. Natl Acad. Sci.
  • an expression vector including a nucleic acid molecule to be expressed together with appropriately positioned expression control sequences, is introduced into pronuclei of fertilized eggs, for example, by microinjection. Integration of the injected DNA is detected by blot analysis of DNA from tissue samples. It is preferred that the introduced DNA be incorporated into the germ line of the animal so that it is passed on to the animal's progeny.
  • Tissue-specific expression may be achieved through the use of a tissue-specific promoter, or through the use of an inducible promoter, such as the metallothionein gene promoter (Palmiter et al., 1983, supra), which allows regulated expression of the transgene.
  • tissue-specific promoter or through the use of an inducible promoter, such as the metallothionein gene promoter (Palmiter et al., 1983, supra), which allows regulated expression of the transgene.
  • Proteins can be isolated by, among other methods, culturing suitable host and vector systems to produce the recombinant translation products as described herein.
  • Supernatants from such cell lines, or protein inclusions, or whole cells from which the protein is not excreted into the supernatant can then be treated by a variety of purification procedures in order to isolate the desired proteins.
  • the supernatant may be first concentrated using commercially available protein concentration filters, such as an Amicon or Millipore Pellicon ultrafiltration unit. Following concentration, the concentrate may be applied to a suitable purification matrix such as, for example, an anti-protein antibody (e.g., an antibody that specifically binds to the polypeptide to be isolated) bound to a suitable support.
  • an anti-protein antibody e.g., an antibody that specifically binds to the polypeptide to be isolated
  • anion or cation exchange resins may be employed in order to purify the protein.
  • one or more reverse-phase high performance liquid chromatography (RP-HPLC) steps may be employed to further purify the protein.
  • RP-HPLC reverse-phase high performance liquid chromatography
  • an isolated polypeptide may be determined by techniques known in the art and described herein, such as gel electrophoresis and chromatography methods. Preferably, such isolated polypeptides are at least about 90% pure, more preferably at least about 95% pure, and most preferably at least about 99% pure.
  • a protein is deemed to be “isolated” within the context of the present invention if no other undesired protein is detected pursuant to SDS-PAGE analysis followed by Coomassie blue staining.
  • the desired protein can be isolated such that no other undesired protein is detected pursuant to SDS-PAGE analysis followed by silver staining.
  • nucleic acid molecules are provided which are capable of inhibiting TGF-beta binding-protein binding to a member of the TGF-beta family.
  • antisense oligonucleotide molecules are provided which specifically inhibit expression of TGF-beta binding-protein nucleic acid sequences (see generally, Hirashima et al. in Molecular Biology of RNA: New Perspectives (M. Inouye and B. S. Dudock, eds., 1987 Academic Press, San Diego, p. 401); Oligonucleotides: Antisense Inhibitors of Gene Expression (J. S.
  • ribozymes are provided which are capable of inhibiting the TGF-beta binding-protein binding to a member of the TGF-beta family.
  • ribozymes are intended to include RNA molecules that contain anti-sense sequences -for specific recognition, and an RNA-cleaving enzymatic activity. The catalytic strand cleaves a specific site in a target RNA at greater than stoichiometric concentration.
  • ribozymes may be utilized within the context of the present invention, including for example, the hammerhead ribozyme (for example, as described by Forster and Symons, Cell 48:211-220, 1987; Haseloff and Gerlach, Nature 328:596-600, 1988; Walbot and Bruening, Nature 334:196, 1988; Haseloff and Gerlach, Nature 334:585, 1988); the hairpin ribozyme (for example, as described by Haseloff et al., U.S. Pat. No. 5,254,678, issued Oct. 19, 1993 and Hempel et al., European Patent Publication No. 0 360 257, published Mar.
  • the hairpin ribozyme for example, as described by Haseloff et al., U.S. Pat. No. 5,254,678, issued Oct. 19, 1993 and Hempel et al., European Patent Publication No. 0 360 257, published Mar.
  • Ribozymes of the present invention typically consist of RNA, but may also be composed of DNA, nucleic acid analogs (e.g., phosphorothioates), or chimerics thereof (e.g., DNA/RNA/RNA).
  • the gene product or any of the candidate molecules described above and below, may be labeled with a variety of compounds, including for example, fluorescent molecules, toxins, and radionuclides.
  • fluorescent molecules include fluorescein, Phycobili proteins, such as phycoerythrin, rhodamine, Texas red and luciferase.
  • toxins include ricin, abrin diphtheria toxin, cholera toxin, gelonin, pokeweed antiviral protein, tritin, Shigella toxin, and Pseudomonas exotoxin A.
  • radionuclides include Cu-64, Ga-67, Ga-68, Zr-89, Ru-97, Tc-99m, Rh-105, Pd-109, In-111, I-123, I-125, I-131, Re-186, Re-188, Au-198, Au-199, Pb-203, At-211, Pb-212 and Bi-212.
  • the antibodies described above may also be labeled or conjugated to one partner of a ligand binding pair.
  • Representative examples include avidin-biotin, streptavidin-biotin, and riboflavin-riboflavin binding protein.
  • the present invention also provides a variety of pharmaceutical compositions, comprising one of the above-described molecules which inhibits the TGF-beta binding-protein binding to a member of the TGF-beta family along with a pharmaceutically or physiologically acceptable carrier, excipients or diluents.
  • a pharmaceutically or physiologically acceptable carrier excipients or diluents.
  • such carriers should be nontoxic to recipients at the dosages and concentrations employed.
  • compositions entails combining the therapeutic agent with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, maltose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, maltose, sucrose or dextrins
  • chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • Neutral buffered saline or saline mixed with nonspecific serum albumin are exemplary appropriate diluents.
  • compositions of the present invention may be prepared for administration by a variety of different routes.
  • the type of carrier is selected based on the mode of administration.
  • Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, intrathecal, rectal, vaginal, sublingual or parenteral administration, including subcutaneous, intravenous, intramuscular, intrasternal, intracavernous, intrameatal, or intraurethral injection or infusion.
  • a pharmaceutical composition (e.g., for oral administration or delivery by injection) may be in the form of a liquid (e.g., an elixir, syrup, solution, emulsion or suspension).
  • a liquid pharmaceutical composition may include, for example, one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils that may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents; antioxidants; chelating agents; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. The use of physiological saline is preferred, and an injectable pharmaceutical composition is preferably sterile.
  • compositions described herein may be formulated for sustained release (i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration).
  • sustained release i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration.
  • Such compositions may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site.
  • Sustained-release formulations may contain an agent dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane.
  • Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release.
  • a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • Illustrative carriers useful in this regard include microparticles of poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose, dextran and the like.
  • Other illustrative delayed-release carriers include supramolecular biovectors, which comprise a non-liquid hydrophilic core (e.g., a cross-linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphilic compound, such as a phospholipid (see e.g., U.S. Pat. No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638).
  • biodegradable microspheres e.g., polylactate polyglycolate
  • Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344, 5,407,609 and 5,942,252.
  • Modified hepatitis B core protein carrier systems such as described in WO/99 40934, and references cited therein, will also be useful for many applications.
  • Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which are capable of inducing a class I-restricted cytotoxic T lymphocyte responses in a host.
  • calcium phosphate core particles are employed as carriers or as controlled release matrices for the compositions of this invention.
  • Exemplary calcium phosphate particles are disclosed, for example, in published patent application No. WO/0046147.
  • the polynucleotide may be present within any of a variety of delivery systems known to those of ordinary skill in the art, including nucleic acid, and bacterial, viral and mammalian expression systems. Techniques for incorporating DNA into such expression systems are well known to those of ordinary skill in the art.
  • the DNA may also be “naked,” as described, for example, in Ulmer et al., Science 259:1745-1749, 1993 and reviewed by Cohen, Science 259:1691-1692, 1993.
  • the uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.
  • compositions disclosed herein may be delivered via oral administration to an animal.
  • these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally will contain a preservative to prevent the growth of microorganisms.
  • Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see U.S. Pat. No. 5,466,468).
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., vegetable oils
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the solution for parenteral administration in an aqueous solution, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, Remington's Pharmaceutical Sciences, 15th ed., pp.
  • compositions disclosed herein may be formulated in a neutral or salt form.
  • Illustrative pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the carriers can further comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • pharmaceutically-acceptable refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • compositions of the present invention are used for the introduction of the compositions of the present invention into suitable host cells/organisms.
  • the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like.
  • compositions of the present invention can be bound, either covalently or non-covalently, to the surface of such carrier vehicles.
  • Liposomes have been used successfully with a number of cell types that are normally difficult to transfect by other procedures, including T cell suspensions, primary hepatocyte cultures and PC 12 cells (Renneisen et al., J. Biol. Chem. 265(27):16337-42, 1990; Muller et al., DNA Cell Biol. 9(3):221-29, 1990).
  • liposomes are free of the DNA length constraints that are typical of viral-based delivery systems. Liposomes have been used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals. Furthermore, he use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery.
  • liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles (MLVs).
  • MLVs multilamellar vesicles
  • the invention provides for pharmaceutically-acceptable nanocapsule formulations of the compositions of the present invention.
  • Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guerrero et al., Drug Dev. Ind. Pharm. 24(12):1113-28, 1998).
  • ultrafine particles sized around 0.1 ⁇ m
  • Such particles can be made as described, for example, by Couvreur et al., Crit. Rev. Ther. Drug Carrier Syst.
  • compositions of the present invention may be placed within containers, along with packaging material that provides instructions regarding the use of such pharmaceutical compositions.
  • instructions will include a tangible expression describing the reagent concentration, as well as within certain embodiments, relative amounts of excipient ingredients or diluents (e.g., water, saline or PBS) which may be necessary to reconstitute the pharmaceutical composition.
  • the present invention also provides methods for increasing the mineral content and mineral density of bone. Briefly, numerous conditions result in the loss of bone mineral content, including for example, disease, genetic predisposition, accidents which result in the lack of use of bone (e.g., due to fracture), therapeutics which effect bone resorption, or which kill bone forming cells and normal aging. Through use of the molecules described herein which inhibit the TGF-beta binding-protein binding to a TGF-beta family member such conditions may be treated or prevented. As utilized herein, it should be understood that bone mineral content has been increased if bone mineral content has been increased in a statistically significant manner (e.g., greater than one-half standard deviation), at a selected site.
  • a statistically significant manner e.g., greater than one-half standard deviation
  • a wide variety of conditions that result in loss of bone mineral content may be treated with the molecules described herein. Patients with such conditions may be identified through clinical diagnosis utilizing well known techniques (see, e.g., Harrison's Principles of Internal Medicine, McGraw-Hill, Inc.). Representative examples of diseases that may be treated included dysplasias, wherein there is abnormal growth or development of bone.
  • Representative examples of such conditions include achondroplasia, cleidocranial dysostosis, enchondromatosis, fibrous dysplasia, Gaucher's Disease, hypophosphatemic rickets, Marfan's Syndrome, multiple hereditary exotoses, neurofibromatosis, osteogenesis imperfecta, osteopetrosis, osteopoikilosis, sclerotic lesions, fractures, periodontal disease, pseudoarthrosis, and pyogenic osteomyelitis.
  • Other conditions which may be treated or prevented include a wide variety of causes of osteopenia (i.e., a condition that causes greater than one standard deviation of bone mineral content or density below peak skeletal mineral content at youth).
  • causes of osteopenia include anemic states, conditions caused by steroids, conditions caused by heparin, bone marrow disorders, scurvy, malnutrition, calcium deficiency, idiopathic osteoporosis, congenital osteopenia or osteoporosis, alcoholism, chronic liver disease, senility, postmenopausal state, oligomenorrhea, amenorrhea, pregnancy, diabetes mellitus, hyperthyroidism, Cushing's disease, acromegaly, hypogonadism, immobilization or disuse, reflex sympathetic dystrophy syndrome, transient regional osteoporosis, and osteomalacia.
  • bone mineral content or density may be increased by administering to a warm-blooded animal a therapeutically effective amount of a molecule that inhibits binding of the TGF-beta binding-protein to a TGF-beta family member.
  • warm-blooded animals that may be treated include both vertebrates and mammals, including for example humans, horses, cows, pigs, sheep, dogs, cats, rats and mice.
  • therapeutic molecules include ribozymes, ribozyme genes, antisense oligonucleotides, and antibodies (e.g., humanized antibodies or any other antibody described herein).
  • methods for increasing bone density, comprising the steps of introducing into cells which home to bone, a vector that directs the expression of a molecule which inhibits binding of the TGF-beta binding-protein to a member of the TGF-beta family, and administering the vector-containing cells to a warm-blooded animal.
  • cells that home to bone may be obtained directly from the bone of patients (e.g., cells obtained from the bone marrow such as CD34+, osteoblasts, osteocytes, and the like), from peripheral blood, or from cultures.
  • a vector that directs the expression of a molecule that inhibits the binding of TGF-beta binding-protein to a member of the TGF-beta family may be introduced into cells.
  • suitable vectors include viral vectors such as herpes viral vectors (e.g., U.S. Pat. No.
  • adenoviral vectors e.g., WO 94/26914, WO 93/9191; Kolls et al., PNAS 91(1):215-219, 1994; Kass-Eisler et al., PNAS 90(24):11498-502, 1993; Guzman et al., Circulation 88(6):2838-48, 1993; Guzman et al., Cir. Res. 73(6):1202-1207, 1993; Zabner et al., Cell 75(2):207-216, 1993; Li et al., Hum Gene Ther. 4(4):403-409, 1993; Caillaud et al., Eur. J. Neurosci.
  • Viral vectors may likewise be constructed which contain a mixture of different elements (e.g., promoters, envelope sequences, and the like) from different viruses, or non-viral sources.
  • the viral vector itself, or a viral particle which contains the viral vector may be utilized in the methods and compositions described below.
  • nucleic acid molecules which encode a molecule which inhibits binding of the TGF-beta binding-protein to a member of the TGF-beta family may be administered by a variety of techniques, including, for example, administration of asialoosomucoid (ASOR) conjugated with poly-L-lysine DNA complexes (Cristano et al., PNAS 92122-92126, 1993), DNA linked to killed adenovirus (Curiel et al., Hum. Gene Ther.
  • ASOR asialoosomucoid
  • cytofectin-mediated introduction DMRIE-DOPE, Vical, Calif.
  • direct DNA injection Acsadi et al., Nature 352:815-818, 1991
  • DNA ligand Wang et al., J. of Biol. Chem. 264:16985-16987, 1989
  • lipofection Felgner et al., Proc. Natl. Acad. Sci. USA 84:7413-7417, 1989
  • liposomes Pickering et al., Circ.
  • nucleic acids which encode the protein itself either alone (Vile and Hart, Cancer Res. 53: 3860-3864, 1993), or utilizing PEG-nucleic acid complexes.
  • Representative examples of molecules that may be expressed by the vectors of present invention include ribozymes and antisense molecules, each of which are discussed in more detail above.
  • Determination of increased bone mineral content may be determined directly through the use of X-rays (e.g., Dual Energy X-ray Absorptometry or “DEXA”), or by inference through bone turnover markers (such as osteoblast specific alkaline phosphatase, osteocalcin, type 1 procollagen C′ propeptide (PICP), and total alkaline phosphatase; see Comier, C., Curr. Opin. in Rheu. 7:243, 1995), or by markers of bone resorption (pyridinoline, deoxypryridinoline, N-telopeptide, urinary hydroxyproline, plasma tartrate-resistant acid phosphatases and galactosyl hydroxylysine; see Comier, supra).
  • the amount of bone mass may also be calculated from body weights or by other methods known in the art (see Guinness-Hey, Metab. Bone Dis. and Relat. Res. 5:177-181, 1984).
  • compositions may be administered by a variety of techniques, as noted above.
  • the DNA samples for this analysis came from 29 sclerosteosis patients in 24 families, 59 unaffected family members and a set of unrelated control individuals from the same population.
  • the pools consisted of 4-6 individuals, either affected individuals, affected individuals from consanguineous families, parents and unaffected siblings, or unrelated controls.
  • analysis of the markers showed several allele sizes for each marker.
  • First-strand cDNA was prepared from the following total RNA samples using a commercially available kit (“Superscript Preamplification System for First-Strand cDNA Synthesis”, Life Technologies, Rockville, Md.): human brain, human liver, human spleen, human thymus, human placenta, human skeletal muscle, human thyroid, human pituitary, human osteoblast (NHOst from Clonetics Corp., San Diego, Calif.), human osteosarcoma cell line (Saos-2, ATCC# HTB-85), human bone, human bone marrow, human cartilage, vervet monkey bone, saccharomyces cerevisiae, and human peripheral blood monocytes.
  • kit (“Superscript Preamplification System for First-Strand cDNA Synthesis”, Life Technologies, Rockville, Md.): human brain, human liver, human spleen, human thymus, human placenta, human skeletal muscle, human thyroid, human pituitary, human osteoblast (NHOst from Clonetics Corp
  • RNA samples were purchased from a commercial source (Clontech, Palo Alto, Calif.), except the following which were prepared in-house: human osteoblast, human osteosarcoma cell line, human bone, human cartilage and vervet monkey bone. These in-house RNA samples were prepared using a commercially available kit (“TRI Reagent”, Molecular Research Center, Inc., Cincinnati, Ohio).
  • PCR was performed on these samples, and additionally on a human genomic sample as a control.
  • the sense Beer oligonucleotide primer had the sequence 5′-CCGGAGCTGGAGAACAACAAG-3′ (SEQ ID NO: 19).
  • the antisense Beer oligonucleotide primer had the sequence 5′-GCACTGGCCGGAGCACACC-3′ (SEQ ID NO: 20).
  • PCR was performed using primers for the human beta-actin gene, as a control.
  • the sense beta-actin oligonucleotide primer had the sequence 5′-AGGCCAACCGCGAGAAGATGA CC-3′ (SEQ ID NO: 21).
  • the antisense beta-actin oligonucleotide primer had the sequence 5′-GAAGT CCAGGGCGACGTAGCA-3′ (SEQ ID NO: 22).
  • PCR was performed using standard conditions in 25 ul reactions, with an annealing temperature of 61 degrees Celsius. Thirty-two cycles of PCR were performed with the Beer primers and twenty-four cycles were performed with the beta-actin primers.
  • the mouse Beer cRNA probe detected a specific message expressed in the neural tube, limb buds, blood vessels and ossifying cartilages of developing mouse embryos.
  • Panel A in FIG. 3 shows expression in the apical ectodermal ridge (aer) of the limb (l) bud, blood vessels (bv) and the neural tube (nt).
  • Panel B shows expression in the 4 th ventricle of the brain (4).
  • Panel C shows expression in the mandible (ma) cervical vertebrae (cv), occipital bone (oc), palate (pa) and a blood vessel (bv).
  • Panel D shows expression in the ribs (r) and a heart valve (va).
  • Panel A is a transverse section of 10.5 dpc embryo.
  • Panel B is a sagittal section of 12.5 dpc embryo and panels C and D are sagittal sections of 15.5 dpc embryos.
  • ba branchial arch
  • h heart
  • te telencephalon (forebrain)
  • b brain
  • f frontonasal mass
  • g gut
  • h heart
  • j jaw
  • li liver
  • lu lung
  • ot otic vesicle
  • skm skeletal muscle
  • ns nasal sinus
  • th thymus
  • to tongue
  • fl forelimb
  • di diaphragm
  • the DNA sequence encoding the full length human Beer protein was amplified using the following PCR oligonucleotide primers:
  • the 5′ oligonucleotide primer had the sequence 5′-AAGCTTGGTACCATGCAGCTCCCAC-3′ (SEQ ID NO: 23) and contained a HindIII restriction enzyme site (in bold) followed by 19 nucleotides of the Beer gene starting 6 base pairs prior to the presumed amino terminal start codon (ATG).
  • the 3′ oligonucleotide primer had the sequence 5′-AAGCTTCTA CTTGTCATCGTCGTCCT TGTAGTC GTAGGCGTTCTCCAGCT-3′ (SEQ ID NO: 24) and contained a HindIII restriction enzyme site (in bold) followed by a reverse complement stop codon (CTA) followed by the reverse complement of the FLAG epitope (underlined, Sigma-Aldrich Co., St. Louis, Mo.) flanked by the reverse complement of nucleotides coding for the carboxy terminal 5 amino acids of the Beer.
  • the PCR product was TA cloned (“Original TA Cloning Kit”, Invitrogen, Carlsbad, Calif.) and individual clones were screened by DNA sequencing.
  • a sequence-verified clone was then digested by HindIII and purified on a 1.5% agarose gel using a commercially available reagents (“QIAquick Gel Extraction Kit”, Qiagen Inc., Valencia, Calif.). This fragment was then ligated to HindIII digested, phosphatase-treated pcDNA3.1 (Invitrogen, Carlsbad, Calif.) plasmid with T4 DNA ligase. DH10B E. coli were transformed and plated on LB, 100 ⁇ g/ml ampicillin plates.
  • Colonies bearing the desired recombinant in the proper orientation were identified by a PCR-based screen, using a 5′ primer corresponding to the T7 promoter/priming site in pcDNA3.1 and a 3′ primer with the sequence 5′-GCACTGGCCGGAGCACACC-3′ (SEQ ID NO: 25) that corresponds to the reverse complement of internal BEER sequence. The sequence of the cloned fragment was confirmed by DNA sequencing.
  • COS-1 cells (ATCC# CRL-1650) were used for transfection. 50 ⁇ g of the expression plasmid pcDNA-Beer-Flag was transfected using a commercially available kit following protocols supplied by the manufacturer (“DEAE-Dextran Transfection Kit”, Sigma Chemical Co., St. Louis, Mo.). The final media following transfection was DMEM (Life Technologies, Rockville, Md.) containing 0.1% Fetal Bovine Serum. After 4 days in culture, the media was removed. Expression of recombinant BEER was analyzed by SDS-PAGE and Western Blot using anti-FLAG® M2 monoclonal antibody (Sigma-Aldrich Co., St. Louis, Mo.).
  • the human Beer gene sequence was amplified using PCR with standard conditions and the following primers: Sense primer: 5′-GTCGTCGGATCCATGGGGTGGCAGGCGTTCAAGAATGAT-3′ (SEQ ID NO:26) Antisense primer: 5′-GTCGTCAAGCTTCTACTTGTCATCGTCCTTGTAGTCGTAGGCGTTCTCCAGCTCGGC-3′ (SEQ ID NO:27)
  • the resulting cDNA contained the coding region of Beer with two modifications.
  • the N-terminal secretion signal was removed and a FLAG epitope tag (Sigma) was fused in frame to the C-terminal end of the insert.
  • BamH1 and HindIII cloning sites were added and the gene was subcloned into pMelBac vector (Invitrogen) for transfer into a baculoviral expression vector using standard methods.
  • SF9 cells (Invitrogen) were maintained in TNM_FH media (Invitrogen) containing 10% fetal calf serum.
  • TNM_FH media Invitrogen
  • SF9 cultures in spinner flasks were infected at an MOI of greater than 10. Samples of the media and cells were taken daily for five days, and Beer expression monitored by western blot using an anti-FLAG M2 monoclonal antibody (Sigma) or an anti-Beer rabbit polyclonal antiserum.
  • the baculovirus-infected SF9 cells were harvested by centrifugation and cell associated protein was extracted from the cell pellet using a high salt extraction buffer (1.5 M NaCl, 50 mM Tris pH 7.5).
  • the extract (20 ml per 300 ml culture) was clarified by centrifugation, dialyzed three times against four liters of Tris buffered saline (150 mM NaCl, 50 mM Tris pH 7.5), and clarified by centrifugation again.
  • This high salt fraction was applied to Hitrap Heparin (Pharmacia; 5 ml bed volume), washed extensively with HEPES buffered saline (25 mM HEPES 7.5, 150 mM Nacl) and bound proteins were eluted with a gradient from 150 mM NaCl to 1200 mM NaCl. Beer elution was observed at aproximately 800 mM NaCl. Beer containing fractions were supplemented to 10% glycerol and 1 mM DTT and frozen at ⁇ 80 degrees C.
  • the listed primers amplified the entire coding region minus the secretion signal sequence.
  • These include restriction sites for subcloning into the bacterial expression vector pQE-30 (Qiagen Inc., Valencia, Calif.) at sites BamHI/HindIII for Beer and Gremlin, and sites SacI/HindIII for Dan.
  • pQE30 contains a coding sequence for a 6 ⁇ His tag at the 5′ end of the cloning region.
  • the completed constructs were transformed into E. coli strain M-15/pRep (Qiagen Inc) and individual clones verified by sequencing. Protein expression in M-15/pRep and purification (6 ⁇ His affinity tag binding to Ni-NTA coupled to Sepharose) were performed as described by the manufacturer (Qiagen, The QIAexpressionist).
  • the E. coli -derived Beer protein was recovered in significant quantity using solubilization in 6M guanidine and dialyzed to 2-4M to prevent precipitation during storage.
  • Gremlin and Dan protein were recovered in higher quantity with solubilization in 6M guanidine and a post purification guanidine concentration of 0.5M.
  • Rabbit antisera and chicken egg Igy fraction were screened for activity via Western blot. Each of the three antigens was separated by PAGE and transferred to 0.45 um nitrocellulose (Novex, San Diego, Calif.). The membrane was cut into strips with each strip containing approximately 75 ng of antigen. The strips were blocked in 3% Blotting Grade Block (Bio-Rad Laboratories, Hercules, Calif.) and washed 3 times in 1 ⁇ Tris buffer saline (TBS)/0.02% TWEEN buffer. The primary antibody (preimmunization bleeds, rabbit antisera or chicken egg IgY in dilutions ranging from 1:100 to 1:10,000 in blocking buffer) was incubated with the strips for one hour with gentle rocking.
  • TBS Tris buffer saline
  • nitrocellulose strips of Beer, Gremlin or Dan were incubated with dilutions (1:5000 and 1:10,000) of their respective rabbit antisera or chicken egg IgY as well as to antisera or chicken egg Igy (dilutions 1:1000 and 1:5000) made to the remaining two antigens.
  • the increased levels of nonmatching antibodies was performed to detect low affinity binding by those antibodies that may be seen only at increased concentration.
  • the protocol and duration of development is the same for all three binding events using the protocol described above. There was no antigen cross-reactivity observed for any of the antigens tested.
  • TGF ⁇ -1, TGF ⁇ -2, TGF ⁇ -3, BMP-4, BMP-5, BMP-6 and GDNF were obtained from commerical sources (R&D systems; Minneapolis, Minn.).
  • a representative protocol is as follows. Partially purified Beer was dialyzed into HEPES buffered saline (25 mM HEPES 7.5, 150 mM NaCl).
  • IP buffer 150 mM NaCl, 25 mM Tris pH 7.5, 1 mM EDTA, 1.4 mM ⁇ -mercaptoethanol, 0.5 % triton ⁇ 100, and 10% glycerol.
  • 30 ng recombinant human BMP-5 protein was applied to 15 ul of FLAG affinity matrix (Sigma; St Louis Mo.)) in the presence and absence of 500 ng FLAG epitope-tagged Beer. The proteins were incubated for 4 hours@ 4° C. and then the affinity matrix-associated proteins were washed 5 times in IP buffer (1 ml per wash).
  • the bound proteins were eluted from the affinity matrix in 60 microliters of 1 ⁇ SDS PAGE sample buffer.
  • the proteins were resolved by SDS PAGE and Beer associated BMP-5 was detected by western blot using anti-BMP-5 antiserum (Research Diagnostics, Inc) (see FIG. 5).
  • FLAG-Beer protein (20 ng) is added to 100 ul PBS/0.2% BSA and adsorbed into each well of 96 well microtiter plate previously coated with anti-FLAG monoclonal antibody (Sigma; St Louis Mo.) and blocked with 10% BSA in PBS. This is conducted at room temperature for 60 minutes. This protein solution is removed and the wells are washed to remove unbound protein. BMP-5 is added to each well in concentrations ranging from 10 pM to 500 nM in PBS/0.2% BSA and incubated for 2 hours at room temperature. The binding solution is removed and the plate washed with three times with 200 ul volumes of PBS/0.2% BSA.
  • BMP-5 levels are then detected using BMP-5 anti-serum via ELISA (F. M. Ausubel et al (1998) Current Protocols in Mol Biol. Vol 2 11.2.1-11.2.22). Specific binding is calculated by subtracting non-specific binding from total binding and analyzed by the LIGAND program (Munson and Podbard, Anal. Biochem., 107, p220-239, (1980).
  • Beer is engineered and expressed as a human Fc fusion protein.
  • ligand BMP is engineered and expressed as mouse Fc fusion.
  • TGF-beta binding-protein binding includes organic compounds derived from commercial or internal collections representing diverse chemical structures. These compounds are prepared as stock solutions in DMSO and are added to assay wells at ⁇ 1% of final volume under the standard assay conditions. These are incubated for 2 hours at room temperature with the BMP and Beer, the solution removed and the bound BMP is quantitated as described.
  • Agents that inhibit 40% of the BMP binding observed in the absence of compound or antibody are considered antagonists of this interaction. These are further evaluated as potential inhibitors based on titration studies to determine their inhibition constants and their influence on TGF-beta binding-protein binding affinity. Comparable specificity control assays may also be conducted to establish the selectivity profile for the identified antagonist through studies using assays dependent on the BMP ligand action (e.g. BMP/BMP receptor competition study).
  • TGF-beta binding-protein is prepared as described by Nicolas (supra). Hydroxyapatite is added to each well of a 96 well microtiter plate equipped with a polypropylene filtration membrane (Polyfiltroninc, Weymouth Mass.). TGF-beta binding-protein is added to 0.2% albumin in PBS buffer. The wells containing matrix are washed 3 times with this buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3M NaOH and quantitated.
  • Inhibitor identification is conducted via incubation of TGF-beta binding-protein with test molecules and applying the mixture to the matrix as described above. The matrix is washed 3 times with 0.2% albumin in PBS buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3 M NaOH and quantitated. Agents that inhibit 40% of the TGF-beta binding-protein binding observed in the absence of compound or antibody are considered bone localization inhibitors. These inhibitors are further characterized through dose response studies to determine their inhibition constants and their influence on TGF-beta binding-protein binding affinity.
  • a full-length TGF-beta binding-protein cDNA in pBluescript SK serves as a template for mutagenesis. Briefly, appropriate primers (see the discussion provided above) are utilized to generate the DNA fragment by polymerase chain reaction using Vent DNA polymerase (New England Biolabs, Beverly, Mass.). The polymerase chain reaction is run for 23 cycles in buffers provided by the manufacturer using a 57° C. annealing temperature. The product is then exposed to two restriction enzymes and after isolation using agarose gel electrophoresis, ligated back into pRBP4-503 from which the matching sequence has been removed by enzymatic digestion. Integrity of the mutant is verified by DNA sequencing.
  • mutant TGF-beta binding-protein cDNAs are transferred into the pcDNA3.1 mammalian expression vector described in EXAMPLE 3. After verifying the sequence, the resultant constructs are transfected into COS-1 cells, and secreted protein is purified as described in EXAMPLE 3.
  • a 41 kb SalI fragment, containing the entire gene body, plus ⁇ 17 kb of 5′ flanking and ⁇ 20 kb of 3′ flanking sequence was sub-cloned into the BamHI site of the SuperCosI cosmid vector (Stratagene, La Jolla, Calif.) and propagated in the E. coli strain DH10B.
  • a 35 kb MluI—AviII restriction fragment (Sequence No. 6), including the entire mouse Beer gene, as well as 17 kb and 14 kb of 5′ and 3′ flanking sequence, respectively, was then gel purified, using conventional means, and used for microinjection of mouse zygotes (DNX Transgenics; U.S. Pat. No. 4,873,191). Founder animals in which the cloned DNA fragment was integrated randomly into the genome were obtained at a frequency of 5-30% of live-born pups. The presence of the transgene was ascertained by performing Southern blot analysis of genomic DNA extracted from a small amount of mouse tissue, such as the tip of a tail.
  • DNA was extracted using the following protocol: tissue was digested overnight at 55° C. in a lysis buffer containing 200 mM NaCl, 100 mM Tris pH8.5, 5 mM EDTA, 0.2% SDS and 0.5 mg/ml Proteinase K. The following day, the DNA was extracted once with phenol/chloroform (50:50), once with chloroform/isoamylalcohol (24:1) and precipitated with ethanol.
  • TE 10 mM Tris pH7.5, 1 mM EDTA
  • 8-10 ug of each DNA sample were digested with a restriction endonuclease, such as EcoRI, subjected to gel electrophoresis and transferred to a charged nylon membrane, such as HyBondN+ (Amersham, Arlington Heights, Ill.).
  • the resulting filter was then hybridized with a radioactively labelled fragment of DNA deriving from the mouse Beer gene locus, and able to recognize both a fragment from the endogenous gene locus and a fragment of a different size deriving from the transgene.
  • mice were bred to normal non-transgenic mice to generate sufficient numbers of transgenic and non-transgenic progeny in which to determine the effects of Beer gene overexpression.
  • animals at various ages for example, 1 day, 3 weeks, 6 weeks, 4 months are subjected to a number of different assays designed to ascertain gross skeletal formation, bone mineral density, bone mineral content, osteoclast and osteoblast activity, extent of endochondral ossification, cartilage formation, etc.
  • the transcriptional activity from the transgene may be determined by extracting RNA from various tissues, and using an RT-PCR assay which takes advantage of single nucleotide polymorphisms between the mouse strain from which the transgene is derived (129 Sv/J) and the strain of mice used for DNA microinjection [(C57BL5/J ⁇ SJL/J)F2].
  • ES cells Homologous recombination in embryonic stem (ES) cells can be used to inactivate the endogenous mouse Beer gene and subsequently generate animals carrying the loss-of-function mutation.
  • a reporter gene such as the E. coli ⁇ -galactosidase gene, was engineered into the targeting vector so that its expression is controlled by the endogenous Beer gene's promoter and translational initiation signal. In this way, the spatial and temporal patterns of Beer gene expression can be determined in animals carrying a targeted allele.
  • the targeting vector was constructed by first cloning the drug-selectable phosphoglycerate kinase (PGK) promoter driven neomycin-resistance gene (neo) cassette from pGT-N29 (New England Biolabs, Beverly, Mass.) into the cloning vector pSP72 (Promega, Madson, Wis.). PCR was used to flank the PGKneo cassette with bacteriophage P1 loxP sites, which are recognition sites for the P1 Cre recombinase (Hoess et al., PNAS USA, 79:3398, 1982).
  • PGK drug-selectable phosphoglycerate kinase
  • neo neomycin-resistance gene
  • PCR primers were comprised of the 34 nucleotide (ntd) loxP sequence, 15-25 ntd complementary to the 5′ and 3′ ends of the PGKneo cassette, as well as restriction enzyme recognition sites (BamHI in the sense primer and EcoRI in the anti-sense primer) for cloning into pSP72.
  • sequence of the sense primer was 5′-AATCTGGATCCATAACTTCGTATAGCATACATTATACGAAGTTATCTGCAG GATTCGAGGGCCCCT-3′ (SEQ ID NO: 34); sequence of the anti-sense primer was 5′-AATCTGAATTCCACCGGTGTTAATTAAATAACTTCGT ATAATGTATGCTATACGAAGTTATAGATCTAGAG TCAGCTTCTGA-3′ (SEQ ID NO: 35).
  • the next step was to clone a 3.6 kb XhoI-HindIII fragment, containing the E. coli ⁇ -galactosidase gene and SV40 polyadenylation signal from pSV ⁇ (Clontech, Palo Alto, Calif.) into the pSP72-PGKneo plasmid.
  • the “short arm” of homology from the mouse Beer gene locus was generated by amplifying a 2.4 kb fragment from the BAC clone 15G5.
  • the approach taken for introducing the “short arm” into the pSP72- ⁇ gal-PGKneo plasmid was to linearize the plasmid at a site upstream of the ⁇ -gal gene and then to co-transform this fragment with the “short arm” PCR product and to select for plasmids in which the PCR product was integrated by homologous recombination.
  • the sense primer for the “short arm” amplification included 30 ntd complementary to the pSP72 vector to allow for this recombination event.
  • the sequence of the sense primer was 5′-ATTTAGGTGACACT ATAGAACTCGAGCAGCTGAAGCTTAACCACATGGTGGCTCACAACCAT-3′ (SEQ ID NO: 36) and the sequence of the anti-sense primer was 5′-AACGACGGCCAGTGAATCCGTA ATCATGGTCATGCTGCCAGGTGGAGGAGGGCA-3′ (SEQ ID NO: 37).
  • the “long arm” from the Beer gene locus was generated by amplifying a 6.1 kb fragment from BAC clone 15G5 with primers which also introduce the rare-cutting restriction enzyme sites SgrAI, FseI, AscI and PacI.
  • the sequence of the sense primer was 5′-ATTACCACCGGTGACACCCGCTTCCTGACAG-3′ (SEQ ID NO: 38);
  • the sequence of the anti-sense primer was 5′-ATTACTTAATTAAACATGGCGCGCCAT ATGGCCGGCCCCTAATTGCGGCGCATCGTTAATT-3′ (SEQ ID NO: 39).
  • the resulting PCR product was cloned into the TA vector (Invitrogen, Carlsbad, Calif.) as an intermediate step.
  • the mouse Beer gene targeting construct also included a second selectable marker, the herpes simplex virus I thymidine kinase gene (HSVTK) under the control of rous sarcoma virus long terminal repeat element (RSV LTR). Expression of this gene renders mammalian cells sensitive (and inviable) to gancyclovir; it is therefore a convenient way to select against neomycin-resistant cells in which the construct has integrated by a non-homologous event (U.S. Pat. No. 5,464,764).
  • HVTK herpes simplex virus I thymidine kinase gene
  • RSV LTR rous sarcoma virus long terminal repeat element
  • the RSVLTR-HSVTK cassette was amplified from pPS1337 using primers that allow subsequent cloning into the FseI and AscI sites of the “long arm”-TA vector plasmid.
  • the sequence of the sense primer was 5′-ATTACGGCCGGCCGCAAAGGAATTCAAGA TCTGA-3′ (SEQ ID NO: 40); the sequence of the anti-sense primer was 5′-ATTACGGCGCGCCCCTC ACAGGCCGCACCCAGCT-3′ (SEQ ID NO: 41).
  • the final step in the construction of the targeting vector involved cloning the 8.8 kb SgrAI-AscI fragment containing the “long arm” and RSVLTR-HSVTK gene into the SgrAI and AscI sites of the pSP72-“short arm”- ⁇ gal-PGKneo plasmid.
  • This targeting vector was linearized by digestion with either AscI or PacI before electroporation into ES cells.
  • 17-nucleotide antisense oligonucleotides are prepared in an overlapping format, in such a way that the 5′ end of the first oligonucleotide overlaps the translation initiating AUG of the Beer transcript, and the 5′ ends of successive oligonucleotides occur in 5 nucleotide increments moving in the 5′ direction (up to 50 nucleotides away), relative to the Beer AUG.
  • Corresponding control oligonucleotides are designed and prepared using equivalent base composition but redistributed in sequence to inhibit any significant hybridization to the coding mRNA. Reagent delivery to the test cellular system is conducted through cationic lipid delivery (P. L. Felgner, Proc. Natl.
  • the cells are harvested, resuspended in lysis buffer (50 mM Tris pH 7.5, 20 mM NaCl, 1 mM EDTA, 1% SDS) and the soluble protein collected. This material is applied to 10-20 % gradient denaturing SDS PAGE. The separated proteins are transferred to nitrocellulose and the western blot conducted as above using the antibody reagents described. In parallel, the control oligonucleotides are added to identical cultures and experimental operations are repeated. Decrease in Beer mRNA or protein levels are considered significant if the treatment with the antisense oligonucleotide results in a 50% change in either instance compared to the control scrambled oligonucleotide. This methodology enables selective gene inactivation and subsequent phenotype characterization of the mineralized nodules in the tissue culture model.
  • Homology recognition techniques e.g., PSI-BLAST (Altschul et al., Nucleic Acids Res. 25:3389-402 (1997)), FUGUE (Shi et al., J. Mol. Biol. 310:243-57 (2001)) suggested that the core region of SOST (SOST_Core) adopts a cystine-knot fold.
  • FUGUE is a sensitive method for detecting homology between sequences and structures.
  • Human Chorionic Gonadotropin ⁇ (hCG- ⁇ ) for which an experimentally determined 3D structure is known, was identified by FUGUE (Shi et al., supra) as the closest homologue of SOST_Core. Therefore, hCG- ⁇ was used as the structural template to build 3D models for SOST_Core.
  • FIG. 7 An alignment of SOST_Core and its close homologues is shown in FIG. 7. Among the homologues shown in the alignment, only hCG- ⁇ (CGHB) had known 3D structure. The sequence identity between SOST_Core and hCG- ⁇ was approximately 25%. Eight CYS residues were conserved throughout the family, emphasizing the overall structural similarity between SOST_Core and hCG- ⁇ . Three pairs of cystines (1-5, 3-7, 4-8) formed disulfide bonds (shown with solid lines in FIG. 7) in a “knot” configuration, which was characteristic to the cystine-knot fold. An extra disulfide bond (2-6), shown as a dotted line in FIG. 7, was unique to this family and distinguished the family of proteins from other cystine-knot families (e.g., TGF- ⁇ , BMP).
  • CGHB hCG- ⁇
  • CYS residues were conserved throughout the family, emphasizing the overall structural similarity between SOST_Core and hCG- ⁇ .
  • SOST_Core was modeled using PDB (Berman et al., Acta Crystallogr. D. Biol. Crystallogr. 58(Pt 6 Pt1):899-907 (2002)) entry 1HCN, the 3D structure of hCG- ⁇ (Wu et al., Structure 2:545-58 (1994)), as the structural template. Models were calculated with MODELER (Sali & Blundell, J. Mol. Biol. 234:779-815 (1993)). A snapshot of the best model is shown in FIG. 8.
  • cystine-knot proteins form dimers because of the lack of hydrophobic core in a monomer (Scheufler et al., supra; Schlunegger and Grutter, J. Mol. Biol. 231:445-58 (1993)); Wu et al., supra). SOST likely follows the same rule and forms a homodimer to increase its stability.
  • hCG- ⁇ was replaced with hCG- ⁇ from the heterodimer structure (1HCN) using structure superimposition techniques combined with manual adjustment, and then a SOST_Core homodimer model was built according to the pseudo hCG- ⁇ homodimer structure.
  • the final model is shown in FIG. 9.
  • This example describes protein modeling of type I and type II receptor binding sites on BMP that are involved with interaction between BMP and SOST.
  • Noggin another BMP antagonist that binds to BMP with high affinity, contacts BMP at both type I and type II receptor binding sites via the N-terminal portion of Noggin (Groppe et al., supra). The two ⁇ -strands in the core region near the C-terminal also contact BMP at the type II receptor binding site.
  • An amino acid sequence alignment is presented in FIG. 10.
  • the cysteine residues that form the characteristic cys-knot were conserved between Noggin and SOST.
  • the overall sequence identity was 24%, and the sequence identity within the N-terminal binding region (alignment positions 1-45) was 33%.
  • the sequence similarity within the core region was about 20%, but the cys-knot scaffold was maintained and a sufficient number of key residues was conserved, supporting homology between Noggin and SOST.
  • the Noggin structure was compared to SOST also to understand how two SOST monomers dimerize. As shown in FIG. 11, the Noggin structure suggested that the linker between the N-terminal region and the core region not only played a role in connecting the two regions, but also formed part of the dimerization interface between two Noggin monomers. One major difference between Noggin and SOST was that the linker between the N-terminal region and the core region was much shorter in SOST.
  • the C-terminal region of SOST may play a role in SOST dimerization.
  • the sequence of Noggin ended with the core region, while SOST had an extra C-terminal region.
  • SOST had an extra C-terminal region.
  • a disulfide bond connected the C-termini of two Noggin monomers.
  • the C-terminal region of SOST started close to the interface of two monomers and could contribute to dimerization.
  • secondary structure prediction showed that some portions of the C-terminal region of SOST had a tendency to form helices. This region in SOST may be responsible for the dimerization activity, possibly through helix-helix packing, which mimicked the function of the longer linker in Noggin.
  • Noggin Another difference between the structure of Noggin and SOST was the amino acid insertion in the SOST core region at alignment positions 169-185 (see FIG. 10). This insertion extended a ⁇ -hairpin, which pointed towards the dimerization interface in the Noggin structure (shown in FIG. 11 as a loop region in the middle of the monomers and above the C-terminal Cys residue). This elongated ⁇ -hairpin could also contribute to SOST dimerization.
  • This Example describes the design of SOST peptide immunogens that are used for immunizing animals and generating antibodies that block interactions between BMP and SOST and prevent dimer formation of SOST monomers.
  • SOST may interact with BMP in a similar manner to Noggin. That is, the N-terminal region of SOST may interact with both the type I and type II receptor binding sites on BMP, and a portion of the core region (amino acid alignment positions 190-220 in FIG. 10) may interact with the type II receptor binding site such that antibodies specific for these SOST regions may block or impair binding of BMP to SOST.
  • amino acid sequences of these SOST polypeptide fragments for rat and human SOST are provided as follows.
  • SOST_N_Linker The N-terminal region (includes the short linker that connects to the core region) Human: QGWQAFKNDATEIIPELGEYPEPPPELENNKTMNRAENGGRPPHHPFETKDVSEYS (SEQ ID NO:92) Rat: QGWQAFKNDATEIIPGLREYPEPPQELENNQTMNRAENGGRPPHHPYDTKDVSEYS (SEQ ID NO:93)
  • SOST_Core_Bind Portion of the core region that is likely to contact BMP at its type II receptor binding site (extended slightly at both termini to include the CYS residue anchors): Human: CIPDRYRAQRVQLLCPGGEAPRARKVRLVASC (SEQ ID NO:94) Rat: CIPDRYRAQRVQLLCPGGAAPRSRKVRLVASC (SEQ ID NO:95)
  • the C-terminal region of SOST is likely to be involved in the formation of SOST homodimers (see Example 12).
  • the elongated ⁇ -hairpin may also play a role in homodimer formation.
  • Antibodies that specifically bind to such regions may prevent or impair dimerization of SOST monomers, which may in turn interfere with interaction between SOST and BMP.
  • Polypeptide fragments in rat and human SOST corresponding to these regions are as follows.
  • SOST_C the C-terminal region (SEQ ID NO:96) Human: LTRFHNQSELKDFGTEAARPQKGRKPRPRARSAKANQAELENAY (SEQ ID NO:97) Rat: LTRFHNQSELKDFGPETARPQKGRKPRPRARGAKANQAELENAY
  • SOST_Core_Dimer Portion of the core region that is likely involved in SOST dimerization (extended slightly at both termini to include the Cys residue anchors): Human: CGPARLLPNAIGRGKWWRPSGPDFRC (SEQ ID NO:98) Rat: CGPARLLPNAIGRVKWWRPNGPDFRC (SEQ ID NO:99)
  • the key N-terminal binding region of SOST was modeled on the basis of the Noggin/BMP-7 complex structure (Protein Data Bank Entry No: 1M4U) and the amino acid sequence alignment (see FIG. 10) to identify amino acid residues of the SOST N-terminus that likely interact with BMP.
  • the model of SOST is presented in FIG. 12.
  • phenylalanine (Phe, F) at alignment position 8 (see arrow and accompanying text) in the SOST sequence projects into a hydrophobic pocket on the surface of the BMP dimer.
  • Peptides were designed to encompass the SOST N-terminal region predicted to make contact with BMP proteins.
  • the peptide sequences are presented below.
  • the peptide sequences were designed to overlap, and an additional cysteine was added to the C-terminal end to facilitate crosslinking to KLH.
  • the peptides were then used for immunization.
  • the peptide sequences of the immunogens are as follows.
  • the following peptides were designed to contain the amino acid portion of core region that was predicted to make contact with BMP proteins. Cysteine was added at the C-terminal end of each peptide for conjugation to KLH, and the conjugated peptides were used for immunization. In the Docking Core N-terminal Peptide an internal cysteine was changed to a serine to avoid double conjugation to KLH.
  • Two regions within SOST that potentially interact to form SOST homodimers include the amino acids with the SOST core region that are not present in Noggin.
  • Human SOST peptides designed to contain this sequence had a C-terminal or N-terminal Cys that was conjugated to KLH.
  • a cysteine was added to the carboxy terminus of the sequence (SEQ ID NO: 76). The KLH conjugated peptides were used for immunization.
  • the second region within SOST that potentially interacts to form SOST homodimers includes the C-terminal region.
  • Peptide immunogens were designed to include amino acid sequences within this region (see below). For conjugation to KLH, a cysteine residue was added to the C-terminal end, and the conjugated peptides were used for immunization.
  • This example describes an assay for detecting binding of a ligand, for example, an antibody or antibody fragment thereof, to sclerostin.
  • a FLAG®-sclerostin fusion protein was prepared according to protocols provided by the manufacturer (Sigma Aldrich, St. Louis, Mo.) and as described in U.S. Pat. No. 6,395,511. Each well of a 96 well microtiter plate is coated with anti-FLAG® monoclonal antibody (Sigma Aldrich) and then blocked with 10% BSA in PBS. The fusion protein (20 ng) is added to 100 ⁇ l PBS/0.2% BSA and adsorbed onto the 96-well plate for 60 minutes at room temperature. This protein solution is removed and the wells are washed to remove unbound fusion protein.
  • a BMP for example, BMP-4, BMP-5, BMP-6, or BMP-7
  • BMP-7 is diluted in PBS/0.2% BSA and added to each well at concentrations ranging from 10 pM to 500 nM. After an incubation for 2 hours at room temperature, the binding solution is removed and the plate is washed three times with 200 ⁇ l volumes of PBS/0.2% BSA. Binding of the BMP to sclerostin is detected using polyclonal antiserum or monoclonal antibody specific for the BMP and an appropriate enzyme-conjugated second step reagent according to standard ELISA techniques (see, e.g., Ausubel et al., Current Protocols in Mol Biol. Vol 2 11.2.1-11.2.22 (1998)). Specific binding is calculated by subtracting non-specific binding from total binding and analyzed using the LIGAND program (Munson and Podbard, Anal. Biochem. 107:220-39 (1980)).
  • Binding of sclerostin to a BMP is also detected by homogeneous time resolved fluorescence detection (Mellor et al., J Biomol. Screening, 3:91-99 (1998)).
  • a polynucleotide sequence encoding sclerostin is operatively linked to a human immunoglobulin constant region in a recombinant nucleic acid construct and expressed as a human Fc-sclerostin fusion protein according to methods known in the art and described herein.
  • a BMP ligand is engineered and expressed as a BMP-mouse Fe fusion protein. These two fusion proteins are incubated together and the assay conducted as described by Mellor et al.
  • This example describes a method for detecting an antibody that inhibits binding of a TGF-beta family member to sclerostin.
  • An ELISA is performed essentially as described in Example 14 except that the BMP concentration is held fixed at its Kd (determined, for example, by BIAcore analysis).
  • an antibody or a library or collection of antibodies is added to the wells to a concentration of 1 ⁇ M.
  • Antibodies are incubated for 2 hours at room temperature with the BMP and sclerostin, the solution removed, and the bound BMP is quantified as described (see Example 14). Antibodies that inhibit 40% of the BMP binding observed in the absence of antibody are considered antagonists of this interaction.
  • TGF-beta binding-protein is prepared as described by Nicolas (supra). Hydroxyapatite is added to each well of a 96-well microtiter plate equipped with a polypropylene filtration membrane (Polyfiltroninc, Weymouth Mass.). TGF-beta binding-protein diluted in 0.2% albumin in PBS buffer is then added to the wells. The wells containing matrix are washed 3 times with 0.2% albumin in PBS buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3 M NaOH and then quantified.
  • An antibody that inhibits or impairs binding of the sclerostin TGF-beta binding protein to the hydroxyapatite is identified by incubating the TGF-beta binding protein with the antibody and applying the mixture to the matrix as described above. The matrix is washed 3 times with 0.2% albumin in PBS buffer. Adsorbed sclerostin is eluted with 0.3 M NaOH and then quantified. An antibody that inhibits the level of binding of sclerostin to the hydroxyapatite by at least 40% compared to the level of binding observed in the absence of antibody is considered a bone localization inhibitor. Such an antibody is further characterized in dose response studies to determine its inhibition constant and its effect on TGF-beta binding-protein binding affinity.

Abstract

A novel class or family of TGF-β binding proteins is disclosed. Also disclosed are assays for selecting molecules for increasing bone mineralization and methods for utilizing such molecules. In particular, compositions and methods relating to antibodies that specifically bind to TGF-beta binding proteins are provided. These methods and compositions relate to altering bone mineral density by interfering with the interaction between a TGF-beta binding protein sclerostin and a TGF-beta superfamily member, particularly a bone morphogenic protein. Increasing bone mineral density has uses in diseases and conditions in which low bone mineral density typifies the condition, such as osteopenia, osteoporosis, and bone fractures.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. application Ser. No. 10/095,248 (filed Mar. 7, 2002), which is a continuation of U.S. application Ser. No. 09/449,218 (filed Nov. 24, 1999), now issued as U.S. Pat. No. 6,395,511, which claims priority from U.S. Provisional Application No. 60/110,283 filed Nov. 27, 1998. The contents of all the above applications are incorporated herein by reference in their entirety.[0001]
  • TECHNICAL FIELD
  • The present invention relates generally to pharmaceutical products and methods and, more specifically, to methods and compositions suitable for increasing the mineral content of bone. Such compositions and methods may be utilized to treat a wide variety of conditions, including for example, osteopenia, osteoporosis, fractures and other disorders in which low bone mineral density are a hallmark of the disease. [0002]
  • BACKGROUND OF THE INVENTION
  • Two or three distinct phases of changes to bone mass occur over the life of an individual (see Riggs, [0003] West J Med. 154:63-77, 1991). The first phase occurs in both men and women, and proceeds to attainment of a peak bone mass. This first phase is achieved through linear growth of the endochondral growth plates, and radial growth due to a rate of periosteal apposition. The second phase begins around age 30 for trabecular bone (flat bones such as the vertebrae and pelvis) and about age 40 for cortical bone (e.g., long bones found in the limbs) and continues to old age. This phase is characterized by slow bone loss, and occurs in both men and women. In women, a third phase of bone loss also occurs, most likely due to postmenopausal estrogen deficiencies. During this phase alone, women may lose an additional 10% of bone mass from the cortical bone and 25% from the trabecular compartment (see Riggs, supra).
  • Loss of bone mineral content can be caused by a wide variety of conditions, and may result in significant medical problems. For example, osteoporosis is a debilitating disease in humans characterized by marked decreases in skeletal bone mass and mineral density, structural deterioration of bone including degradation of bone microarchitecture and corresponding increases in bone fragility and susceptibility to fracture in afflicted individuals. Osteoporosis in humans is preceded by clinical osteopenia (bone mineral density that is greater than one standard deviation but less than 2.5 standard deviations below the mean value for young adult bone), a condition found in approximately 25 million people in the United States. Another 7-8 million patients in the United States have been diagnosed with clinical osteoporosis (defined as bone mineral content greater than 2.5 standard deviations below that of mature young adult bone). Osteoporosis is one of the most expensive diseases for the health care system, costing tens of billions of dollars annually in the United States. In addition to health care-related costs, long-term residential care and lost working days add to the financial and social costs of this disease. Worldwide approximately 75 million people are at risk for osteoporosis. [0004]
  • The frequency of osteoporosis in the human population increases with age, and among Caucasians is predominant in women (who comprise 80% of the osteoporosis patient pool in the United States). The increased fragility and susceptibility to fracture of skeletal bone in the aged is aggravated by the greater risk of accidental falls in this population. More than 1.5 million osteoporosis-related bone fractures are reported in the United States each year. Fractured hips, wrists, and vertebrae are among the most common injuries associated with osteoporosis. Hip fractures in particular are extremely uncomfortable and expensive for the patient, and for women correlate with high rates of mortality and morbidity. [0005]
  • Although osteoporosis has been defined as an increase in the risk of fracture due to decreased bone mass, none of the presently available treatments for skeletal disorders can substantially increase the bone density of adults. There is a strong perception among all physicians that drugs are needed which could increase bone density in adults, particularly in the bones of the wrist, spinal column and hip that are at risk in osteopenia and osteoporosis. [0006]
  • Current strategies for the prevention of osteoporosis may offer some benefit to individuals but cannot ensure resolution of the disease. These strategies include moderating physical activity (particularly in weight-bearing activities) with the onset of advanced age, including adequate calcium in the diet, and avoiding consumption of products containing alcohol or tobacco. For patients presenting with clinical osteopenia or osteoporosis, all current therapeutic drugs and strategies are directed to reducing further loss of bone mass by inhibiting the process of bone absorption, a natural component of the bone remodeling process that occurs constitutively. [0007]
  • For example, estrogen is now being prescribed to retard bone loss. There is, however, some controversy over whether there is any long term benefit to patients and whether there is any effect at all on patients over 75 years old. Moreover, use of estrogen is believed to increase the risk of breast and endometrial cancer. [0008]
  • High doses of dietary calcium, with or without vitamin D has also been suggested for postmenopausal women. However, high doses of calcium can often have unpleasant gastrointestinal side effects, and serum and urinary calcium levels must be continuously monitored (see Khosla and Rigss, [0009] Mayo Clin. Proc. 70:978-982, 1995).
  • Other therapeutics which have been suggested include calcitonin, bisphosphonates, anabolic steroids and sodium fluoride. Such therapeutics however, have undesirable side effects (e.g., calcitonin and steroids may cause nausea and provoke an immune reaction, bisphosphonates and sodium fluoride may inhibit repair of fractures, even though bone density increases modestly) that may prevent their usage (see Khosla and Rigss, supra). [0010]
  • No currently practiced therapeutic strategy involves a drug that stimulates or enhances the growth of new bone mass. The present invention provides compositions and methods which can be utilized to increase bone mineralization, and thus may be utilized to treat a wide variety of conditions where it is desired to increase bone mass. Further, the present invention provides other, related advantages. [0011]
  • SUMMARY OF THE INVENTION
  • As noted above, the present invention provides a novel class or family of TGF-beta binding-proteins, as well as assays for selecting compounds which increase bone mineral content and bone mineral density, compounds which increase bone mineral content and bone mineral density and methods for utilizing such compounds in the treatment or prevention of a wide variety of conditions. [0012]
  • Within one aspect of the present invention, isolated nucleic acid molecules are provided, wherein said nucleic acid molecules are selected from the group consisting of: (a) an isolated nucleic acid molecule comprising sequence ID Nos. 1, 5, 7, 9, 11, 13, or, 15, or complementary sequence thereof; (b) an isolated nucleic acid molecule that specifically hybridizes to the nucleic acid molecule of (a) under conditions of high stringency; and (c) an isolated nucleic acid that encodes a TGF-beta binding-protein according to (a) or (b). Within related aspects of the present invention, isolated nucleic acid molecules are provided based upon hybridization to only a portion of one of the above-identified sequences (e.g., for (a) hybridization may be to a probe of at least 20, 25, 50, or 100 nucleotides selected from nucleotides 156 to 539 or 555 to 687 of Sequence ID No. 1). As should be readily evident, the necessary stringency to be utilized for hybridization may vary based upon the size of the probe. For example, for a 25-mer probe high stringency conditions could include: 60 mM Tris pH 8.0, 2 mM EDTA, 5× Denhardt's, 6×SSC, 0.1% (w/v) N-laurylsarcosine, 0.5% (w/v) NP-40 (nonidet P-40) overnight at 45 degrees C., followed by two washes with 0.2×SSC/0.1% SDS at 45-50 degrees. For a 100-mer probe under low stringency conditions, suitable conditions might include the following: 5× SSPE, 5× Denhardt's, and 0.5% SDS overnight at 42-50 degrees, followed by two washes with 2× SSPE (or 2×SSC)/0.1% SDS at 42-50 degrees. [0013]
  • Within related aspects of the present invention, isolated nucleic acid molecules are provided which have homology to Sequence ID Nos. 1, 5, 7, 9, 11, 13, or 15, at a 50%, 60%, 75%, 80%, 90%, 95%, or 98% level of homology utilizing a Wilbur-Lipman algorithm. Representative examples of such isolated molecules include, for example, nucleic acid molecules which encode a protein comprising Sequence ID NOs. 2, 6, 10, 12, 14, or 16, or have homology to these sequences at a level of 50%, 60%, 75%, 80%, 90%, 95%, or 98% level of homology utilizing a Lipman-Pearson algorithm. [0014]
  • Isolated nucleic acid molecules are typically less than 100 kb in size, and, within certain embodiments, less than 50 kb, 25 kb, 10 kb, or even 5 kb in size. Further, isolated nucleic acid molecules, within other embodiments, do not exist in a “library” of other unrelated nucleic acid molecules (e.g., a subclone BAC such as described in GenBank Accession No. AC003098 and EMB No. AQ171546). However, isolated nucleic acid molecules can be found in libraries of related molecules (e.g., for shuffling, such as is described in U.S. Pat. Nos. 5,837,458; 5,830,721; and 5,811,238). Finally, isolated nucleic acid molecules as described herein do not include nucleic acid molecules which encode Dan, Cerberus, Gremlin, or SCGF (U.S. Pat. No. 5,780,263). [0015]
  • Also provided by the present invention are cloning vectors which contain the above-noted nucleic acid molecules, and expression vectors which comprise a promoter (e.g., a regulatory sequence) operably linked to one of the above-noted nucleic acid molecules. Representative examples of suitable promoters include tissue-specific promoters, and viral-based promoters (e.g., CMV-based promoters such as CMV I-E, SV40 early promoter, and MuLV LTR). Expression vectors may also be based upon, or derived from viruses (e.g., a “viral vector”). Representative examples of viral vectors include herpes simplex viral vectors, adenoviral vectors, adenovirus-associated viral vectors and retroviral vectors. Also provided are host cells containing or comprising any of above-noted vectors (including for example, host cells of human, monkey, dog, rat, or mouse origin). [0016]
  • Within other aspects of the present invention, methods of producing TGF-beta binding-proteins are provided, comprising the step of culturing the aforementioned host cell containing vector under conditions and for a time sufficient to produce the TGF-beta binding protein. Within further embodiments, the protein produced by this method may be further purified (e.g., by column chromatography, affinity purification, and the like). Hence, isolated proteins which are encoded by the above-noted nucleic acid molecules (e.g., Sequence ID NOs. 2, 4, 6, 8, 10, 12, 14, or 16) may be readily produced given the disclosure of the subject application. [0017]
  • It should also be noted that the aforementioned proteins, or fragments thereof, may be produced as fusion proteins. For example, within one aspect fusion proteins are provided comprising a first polypeptide segment comprising a TGF-beta binding-protein encoded by a nucleic acid molecule as described above, or a portion thereof of at least 10, 20, 30, 50, or 100 amino acids in length, and a second polypeptide segment comprising a non-TGF-beta binding-protein. Within certain embodiments, the second polypeptide may be a tag suitable for purification or recognition (e.g., a polypeptide comprising multiple anionic amino acid residues—see U.S. Pat. No. 4,851,341), a marker (e.g., green fluorescent protein, or alkaline phosphatase), or a toxic molecule (e.g., ricin). [0018]
  • Within another aspect of the present invention, antibodies are provided which are capable of specifically binding the above-described class of TGF-beta binding proteins (e.g., human BEER). Within various embodiments, the antibody may be a polyclonal antibody, or a monoclonal antibody (e.g., of human or murine origin). Within further embodiments, the antibody is a fragment of an antibody which retains the binding characteristics of a whole antibody (e.g., an F(ab′)[0019] 2, F(ab)2, Fab′, Fab, or Fv fragment, or even a CDR). Also provided are hybridomas and other cells which are capable of producing or expressing the aforementioned antibodies.
  • Within related aspects of the invention, methods are provided detecting a TGF-beta binding protein, comprising the steps of incubating an antibody as described above under conditions and for a time sufficient to permit said antibody to bind to a TGF-beta binding protein, and detecting the binding. Within various embodiments the antibody may be bound to a solid support to facilitate washing or separation, and/or labeled. (e.g., with a marker selected from the group consisting of enzymes, fluorescent proteins, and radioisotopes). [0020]
  • Within other aspects of the present invention, isolated oligonucleotides are provided which hybridize to a nucleic acid molecule according to Sequence ID NOs. 1, 3, 5, 7, 9, 11, 13, 15, 17, or 18 or the complement thereto, under conditions of high stringency. Within further embodiments, the oligonucleotide may be found in the sequence which encodes Sequence ID Nos. 2, 4, 6, 8, 10, 12, 14, or 16. Within certain embodiments, the oligonucleotide is at least 15, 20, 30, 50, or 100 nucleotides in length. Within further embodiments, the oligonucleotide is labeled with another molecule (e.g., an enzyme, fluorescent molecule, or radioisotope). Also provided are primers which are capable of specifically amplifying all or a portion of the above-mentioned nucleic acid molecules which encode TGF-beta binding-proteins. As utilized herein, the term “specifically amplifying” should be understood to refer to primers which amplify the aforementioned TGF-beta binding-proteins, and not other TGF-beta binding proteins such as Dan, Cerberus, Gremlin, or SCGF (U.S. Pat. No. 5,780,263). [0021]
  • Within related aspects of the present invention, methods are provided for detecting a nucleic acid molecule which encodes a TGF-beta binding protein, comprising the steps of incubating an oligonucleotide as described above under conditions of high stringency, and detecting hybridization of said oligonucleotide. Within certain embodiments, the oligonucleotide may be labeled and/or bound to a solid support. [0022]
  • Within other aspects of the present invention, ribozymes are provided which are capable of cleaving RNA which encodes one of the above-mentioned TGF-beta binding-proteins (e.g., Sequence ID NOs. 2, 6, 8, 10, 12, 14, or 16). Such ribozymes may be composed of DNA, RNA (including 2′-O-methyl ribonucleic acids), nucleic acid analogs (e.g., nucleic acids having phosphorothioate linkages) or mixtures thereof. Also provided are nucleic acid molecules (e.g., DNA or cDNA) which encode these ribozymes, and vectors which are capable of expressing or producing the ribozymes. Representative examples of vectors include plasmids, retrotransposons, cosmids, and viral-based vectors (e.g., viral vectors generated at least in part from a retrovirus, adenovirus, or, adeno-associated virus). Also provided are host cells (e.g., human, dog, rat, or mouse cells) which contain these vectors. In certain embodiments, the host cell may be stably transformed with the vector. [0023]
  • Within further aspects of the invention, methods are provided for producing ribozymes either synthetically, or by in vitro or in vivo transcription. Within further embodiments, the ribozymes so produced may be further purified and/or formulated into pharmaceutical compositions (e.g., the ribozyme or nucleic acid molecule encoding the ribozyme along with a pharmaceutically acceptable carrier or diluent). Similarly, the antisense oligonucleotides and antibodies or other selected molecules described herein may be formulated into pharmaceutical compositions. [0024]
  • Within other aspects of the present invention, antisense oligonucleotides are provided comprising a nucleic acid molecule which hybridizes to a nucleic acid molecule according to Sequence ID NOs. 1, 3, 5, 7, 9, 11, 13, or 15, or the complement thereto, and wherein said oligonucleotide inhibits the expression of TGF-beta binding protein as described herein (e.g., human BEER). Within various embodiments, the oligonucleotide is 15, 20, 25, 30, 35, 40, or 50 nucleotides in length. Preferably, the oligonucleotide is less than 100, 75, or 60 nucleotides in length. As should be readily evident, the oligonucleotide may be comprised of one or more nucleic acid analogs, ribonucleic acids, or deoxyribonucleic acids. Further, the oligonucleotide may be modified by one or more linkages, including for example, covalent linkage such as a phosphorothioate linkage, a phosphotriester linkage, a methyl phosphonate linkage, a methylene(methylimino) linkage, a morpholino linkage, an amide linkage, a polyamide linkage, a short chain alkyl intersugar linkage, a cycloalkyl intersugar linkage, a short chain heteroatomic intersugar linkage and a heterocyclic intersugar linkage. One representative example of a chimeric oligonucleotide is provied in U.S. Pat. No. 5,989,912. [0025]
  • Within yet another aspect of the present invention, methods are provided for increasing bone mineralization, comprising introducing into a warm-blooded animal an effective amount of the ribozyme as described above. Within related aspects, such methods comprise the step of introducing into a patient an effective amount of the nucleic acid molecule or vector as described herein which is capable of producing the desired ribozyme, under conditions favoring transcription of the nucleic acid molecule to produce the ribozyme. [0026]
  • Within other aspects of the invention transgenic, non-human animals are provided. Within one embodiment a transgenic animal is provided whose germ cells and somatic cells contain a nucleic acid molecule encoding a TGF-beta binding-protein as described above which is operably linked to a promoter effective for the expression of the gene, the gene being introduced into the animal, or an ancestor of the animal, at an embryonic stage, with the proviso that said animal is not a human. Within other embodiments, transgenic knockout animals are provided, comprising an animal whose germ cells and somatic cells comprise a disruption of at least one allele of an endogenous nucleic acid molecule which hybridizes to a nucleic acid molecule which encodes a TGF-binding protein as described herein, wherein the disruption prevents transcription of messenger RNA from said allele as compared to an animal without the disruption, with the proviso that the animal is not a human. Within various embodiments, the disruption is a nucleic acid deletion, substitution, or, insertion. Within other embodiments the transgenic animal is a mouse, rat, sheep, pig, or dog. [0027]
  • Within further aspects of the invention, kits are provided for the detection of TGF-beta binding-protein gene expression, comprising a container that comprises a nucleic acid molecule, wherein the nucleic acid molecule is selected from the group consisting of (a) a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS: 1, 3, 5, 7, 9, 1 1, 13, 15, 100, or 101; (b) a nucleic acid molecule comprising the complement of the nucleotide sequence of (a); (c) a nucleic acid molecule that is a fragment of (a) or (b) of at least 15, 20 30, 50, 75, or, 100 nucleotides in length. Also provided are kits for the detection of a TGF-beta binding-protein which comprise a container that comprise one of the TGF-beta binding protein antibodies described herein. [0028]
  • For example, within one aspect of the present invention methods are provided for determining whether a selected molecule is capable of increasing bone mineral content, comprising the steps of (a) mixing one or more candidate molecules with TGF-beta-binding-protein encoded by the nucleic acid molecule according to [0029] claim 1 and a selected member of the TGF-beta family of proteins (e.g., BMP 5 or 6), (b) determining whether the candidate molecule alters the signaling of the TGF-beta family member, or alters the binding of the TGF-beta binding-protein to the TGF-beta family member. Within certain embodiments, the molecule alters the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation. Within this aspect of the present invention, the candidate molecule(s) may alter signaling or binding by, for example, either decreasing (e.g., inhibiting), or increasing (e.g., enhancing) signaling or binding.
  • Within yet another aspect, methods are provided for determining whether a selected molecule is capable of increasing bone mineral content, comprising the step of determining whether a selected molecule inhibits the binding of TGF-beta binding-protein to bone, or an analogue thereof. Representative examples of bone or analogues thereof include hydroxyapatite and primary human bone samples obtained via biopsy. [0030]
  • Within certain embodiments of the above-recited methods, the selected molecule is contained within a mixture of molecules and the methods may further comprise the step of isolating one or more molecules which are functional within the assay. Within yet other embodiments, TGF-beta family of proteins is bound to a solid support and the binding of TGF-beta binding-protein is measured or TGF-beta binding-protein are bound to a solid support and the binding of TGF-beta proteins are measured. [0031]
  • Utilizing methods such as those described above, a wide variety of molecules may be assayed for their ability to increase bone mineral content by inhibiting the binding of the TGF-beta binding-protein to the TGF-beta family of proteins. Representative examples of such molecules include proteins or peptides, organic molecules, and nucleic acid molecules. [0032]
  • Within other related aspects of the invention, methods are provided for increasing bone mineral content in a warm-blooded animal, comprising the step of administering to a warm-blooded animal a therapeutically effective amount of a molecule identified from the assays recited herein. Within another aspect, methods are provided for increasing bone mineral content in a warm-blooded animal, comprising the step of administering to a warm-blooded animal a therapeutically effective amount of a molecule which inhibits the binding of the TGF-beta binding-protein to the TGF-beta super-family of proteins, including bone morphogenic proteins (BMPs). Representative examples of suitable molecules include antisense molecules, ribozymes, ribozyme genes, and antibodies (e.g., a humanized antibody) which specifically recognize and alter the activity of the TGF-beta binding-protein. [0033]
  • Within another aspect of the present invention, methods are provided for increasing bone mineral content in a warm-blooded animal, comprising the steps of (a) introducing into cells which home to the bone a vector which directs the expression of a molecule which inhibits the binding of the TGF-beta binding-protein to the TGF-beta family of proteins and bone morphogenic proteins (BMPs), and (b) administering the vector-containing cells to a warm-blooded animal. As utilized herein, it should be understood that cells “home to bone” if they localize within the bone matrix after peripheral administration. Within one embodiment, such methods further comprise, prior to the step of introducing, isolating cells from the marrow of bone which home to the bone. Within a further embodiment, the cells which home to bone are selected from the group consisting of CD34+ cells and osteoblasts. [0034]
  • Within other aspects of the present invention, molecules are provided (preferably isolated) which inhibit the binding of the TGF-beta binding-protein to the TGF-beta super-family of proteins. [0035]
  • Within further embodiments, the molecules may be provided as a composition, and can further comprise an inhibitor of bone resorption. Representative examples of such inhibitors include calcitonin, estrogen, a bisphosphonate, a growth factor having anti-resorptive activity and tamoxifen. [0036]
  • Representative examples of molecules which may be utilized in the aforementioned therapeutic contexts include, e.g., ribozymes, ribozyme genes, antisense molecules, and/or antibodies (e.g., humanized antibodies). Such molecules may depending upon their selection, used to alter, antagonize, or agonize the signalling or binding of a TGF-beta binding-protein family member as described herein. [0037]
  • Within various embodiments of the invention, the above-described molecules and methods of treatment or prevention may be utilized on conditions such as osteoporosis, osteomalasia, periodontal disease, scurvy, Cushing's Disease, bone fracture and conditions due to limb immobilization and steroid usage. [0038]
  • The present invention also provides antibodies that specifically bind to a TGF-beta binding protein, sclerostin (SOST), and provides immunogens comprising sclerostin peptides derived from regions of sclerostin that interact with a member of the TGF-beta superfamily such as a bone morphogenic protein. In one embodiment, the invention provides an antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide, said sclerostin polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the antibody competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in GenBank Ace. Nos. NM[0039] 004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); or AAB33865 (SEQ ID NO: 110) and wherein the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. NOS. U25110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); or NM001204 (SEQ ID NO: 113). In another embodiment, the invention provides an antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide and that impairs formation of a sclerostin homodimer, wherein the sclerostin polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65.
  • In certain particular embodiments of the invention, the antibody is a polyclonal antibody. In other embodiments, the antibody is a monoclonal antibody, which is a mouse, human, rat, or hamster monoclonal antibody. The invention also provides a hybridoma cell or a host cell that is capable of producing the monoclonal antibody. In other embodiments of the invention, the antibody is a humanized antibody or a chimeric antibody. The invention further provides a host cell that produces the humanized or chimeric antibody. In certain embodiments the antigen-binding fragment of the antibody is a F(ab′)[0040] 2, Fab′, Fab, Fd, or Fv fragment. The invention also provides an antibody that is a single chain antibody and provides a host cell that is capable of expressing the single chain antibody. In another embodiment, the invention provides a composition comprising such antibodies and a physiologically acceptable carrier.
  • In another embodiment, the invention provides an immunogen comprising a peptide comprising at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in GenBank Ace. Nos. NM[0041] 004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); or AAB33865 (SEQ ID NO: 110) and wherein the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. NOs. U25110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); or NM001204 (SEQ ID NO: 113). The invention also provides an immunogen comprising a peptide that comprises at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that impairs formation of a SOST homodimer.
  • In certain particular embodiments, the subject invention immunogens are associated with a carrier molecule. In certain embodiments, the carrier molecule is a carrier polypeptide, and in particular embodiments, the carrier polypeptide is keyhole limpet hemocyanin. [0042]
  • The invention also provides a method for producing an anti-body that specifically binds to a SOST polypeptide, comprising immunizing a non-human animal with an immunogen comprising a peptide comprising at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, wherein (a) the SOST polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65; (b) the antibody competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site; (c) the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. Nos. NM[0043] 004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); or AAB33865 (SEQ ID NO: 110); and (d) the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in GenBank Ace. NOs. U25110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); or NM001204 (SEQ ID NO: 113).
  • In another embodiment, the invention provides a method for producing an antibody that specifically binds to a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, comprising immunizing a non-human animal with an immunogen comprising a peptide that comprises at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the antibody impairs formation of a SOST homodimer. [0044]
  • These and other aspects of the present invention will become evident upon reference to the following detailed description and attached drawings. In addition, documents including various references set forth herein that describe in more detail certain procedures or compositions (e.g., plasmids, etc.), are incorporated by reference in their entirety.[0045]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic illustration comparing the amino acid sequence of Human Dan; Human Gremlin; Human Cerberus and Human Beer. Arrows indicate the Cysteine backbone. [0046]
  • FIG. 2 summarizes the results obtained from surveying a variety of human tissues for the expression of a TGF-beta binding-protein gene, specifically, the Human Beer gene. A semi-quantitative Reverse Transcription-Polymerase Chain Reaction (RT-PCR) procedure was used to amplify a portion of the gene from first-strand cDNA synthesized from total RNA (described in more detail in EXAMPLE 2A). [0047]
  • FIGS. [0048] 3A-3D summarize the results obtained from RNA in situ hybridization of mouse embryo sections, using a cRNA probe that is complementary to the mouse Beer transcript (described in more detail in EXAMPLE 2B). Panel 3A is a transverse section of 10.5 dpc embryo. Panel 3B is a sagittal section of 12.5 dpc embryo and panels 3C and 3D are sagittal sections of 15.5 dpc embryos.
  • FIGS. [0049] 4A-4C illustrate, by western blot analysis, the specificity of three different polyclonal antibodies for their respective antigens (described in more detail in EXAMPLE 4). FIG. 4A shows specific reactivity of an anti-H. Beer antibody for H. Beer antigen, but not H. Dan or H. Gremlin. FIG. 4B shows reactivity of an anti-H. Gremlin antibody for H. Gremlin antigen, but not H. Beer or H. Dan. FIG. 4C shows reactivity of an anti-H. Dan antibody for H. Dan, but not H. Beer or H. Gremlin.
  • FIG. 5 illustrates, by western blot analysis, the selectivity of the TGF-beta binding-protein, Beer, for BMP-5 and BMP-6, but not BMP-4 (described in more detail in EXAMPLE 5). [0050]
  • FIG. 6 demonstrates that the ionic interaction between the TGF-beta binding-protein, Beer, and BMP-5 has a dissociation constant in the 15-30 nM range. [0051]
  • FIG. 7 presents an alignment of the region containing the characteristic cystine-knot of a SOST (sclerostin) polypeptide and its closest homologues. Three disulphide bonds that form the cystine-knot are illustrated as solid lines. An extra disulphide bond, shown by a dotted line, is unique to this family, which connects two β-hairpin tips in the 3D structure. The polypeptides depicted are SOST: sclerostin (SEQ ID NO: 126); CGHB: Human Chorionic Gonadotropin β (SEQ ID NO: 127); FSHB: follicle-stimulating hormone beta subunit (SEQ ID NO: 128); TSHB: thyrotropin beta chain precursor (SEQ ID NO: 129); VWF: Von Willebrand factor (SEQ ID NO: 130); MUC2: human mucin 2 precursor (SEQ ID NO: 131); CER1: Cerberus 1 ([0052] Xenopus laevis homolog) (SEQ ID NO: 132); DRM: gremlin (SEQ ID NO: 133); DAN: (SEQ ID NO: 134); CTGF: connective tissue growth factor precursor (SEQ ID NO: 135); NOV: NovH (nephroblastoma overexpressed gene protein homolog) (SEQ ID NO: 136); CYR6: (SEQ ID NO: 137).
  • FIG. 8 illustrates a 3D model of the core region of SOST (SOST_Core). [0053]
  • FIG. 9 presents a 3D model of the core region of SOST homodimer. [0054]
  • FIGS. 10A and 10B provide an amino acid sequence alignment of Noggin from five different animals: human (NOGG_HUMAN (SEQ ID NO: 138); chicken (NOGG_CHICK, SEQ ID NO: 139); African clawed frog (NOGG_XENLA, SEQ ID NO: 140); NOGG_FUGRU, SEQ ID NO: 141); and zebrafish (NOGG_ZEBRA, SEQ ID NO: 142); and SOST from human (SOST_HUMAN, SEQ ID NO: 46), rat (SOST_RAT, SEQ ID NO: 65), and mouse (SOST Mouse, SEQ ID NO: 143). [0055]
  • FIG. 11 illustrates the Noggin/BMP-7 complex structure. The BMP homodimer is shown on the bottom portion of the figure in surface mode. The Noggin homodimer is shown on top of the BMP dimer in cartoon mode. The circles outline the N-terminal binding region, the core region, and the linker between the N-terminal and core regions. [0056]
  • FIG. 12 depicts a 3D model of the potential BMP-binding fragment located at the SOST N-terminal region. A BMP dimer is shown in surface mode, and the potential BMP-binding fragment is shown in stick mode. A phenylalanine residue fitting into a hydrophobic pocket on the BMP surface is noted.[0057]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Definitions [0058]
  • Prior to setting forth the invention in detail, it may be helpful to an understanding thereof to set forth definitions of certain terms and to list and to define the abbreviations that will be used hereinafter. [0059]
  • “Molecule” should be understood to include proteins or peptides (e.g., antibodies, recombinant binding partners, peptides with a desired binding affinity), nucleic acids (e.g., DNA, RNA, chimeric nucleic acid molecules, and nucleic acid analogues such as PNA); and organic or inorganic compounds. [0060]
  • “TGF-beta” should be understood to include any known or novel member of the TGF-beta super-family, which also includes bone morphogenic proteins (BMPs). [0061]
  • “TGF-beta receptor” should be understood to refer to the receptor specific for a particular member of the TGF-beta super-family (including bone morphogenic proteins (BMPs)). [0062]
  • “TGF-beta binding-protein” should be understood to refer to a protein with specific binding affinity for a particular member or subset of members of the TGF-beta super-family (including bone morphogenic proteins (BMPs)). Specific examples of TGF-beta binding-proteins include proteins encoded by Sequence ID Nos. 1, 5, 7, 9, 11, 13, 15, 100, and 101. [0063]
  • Inhibiting the “binding of the TGF-beta binding-protein to the TGF-beta family of proteins and bone morphogenic proteins (BMPs)” should be understood to refer to molecules which allow the activation of TGF-beta or bone morphogenic proteins (BMPs), or allow the binding of TGF-beta family members including bone morphogenic proteins (BMPs) to their respective receptors, by removing or preventing TGF-beta from binding to TGF-binding-protein. Such inhibition may be accomplished, for example, by molecules which inhibit the binding of the TGF-beta binding-protein to specific members of the TGF-beta super-family. [0064]
  • “Vector” refers to an assembly that is capable of directing the expression of desired protein. The vector must include transcriptional promoter elements that are operably linked to the gene(s) of interest. The vector may be composed of deoxyribonucleic acids (“DNA”), ribonucleic acids (“RNA”), or a combination of the two (e.g., a DNA-RNA chimeric). Optionally, the vector may include a polyadenylation sequence, one or more restriction sites, as well as one or more selectable markers such as neomycin phosphotransferase or hygromycin phosphotransferase. Additionally, depending on the host cell chosen and the vector employed, other genetic elements such as an origin of replication, additional nucleic acid restriction sites, enhancers, sequences conferring inducibility of transcription, and selectable markers, may also be incorporated into the vectors described herein. [0065]
  • An “isolated nucleic acid molecule” is a nucleic acid molecule that is not integrated in the genomic DNA of an organism. For example, a DNA molecule that encodes a TGF-binding protein that has been separated from the genomic DNA of a eukaryotic cell is an isolated DNA molecule. Another example of an isolated nucleic acid molecule is a chemically-synthesized nucleic acid molecule that is not integrated in the genome of an organism. The isolated nucleic acid molecule may be genomic DNA, cDNA, RNA, or composed at least in part of nucleic acid analogs. [0066]
  • An “isolated polypeptide” is a polypeptide that is essentially free from contaminating cellular components, such as carbohydrate, lipid, or other proteinaceous impurities associated with the polypeptide in nature. Preferably, such isolated polypeptides are at least about 90% pure, more preferably at least about 95% pure, and most preferably at least about 99% pure. Within certain embodiments, a particular protein preparation contains an isolated polypeptide if it appears nominally as a single band on SDS-PAGE gel with Coomassie Blue staining. The term “isolated” when referring to organic molecules (e.g., organic small molecules) means that the compounds are greater than 90% pure utilizing methods which are well known in the art (e.g., NMR, melting point). [0067]
  • “Sclerosteosis” is a term that was applied by Hansen (1967) (Hansen, H. G., Sklerosteose. in: Opitz, H.; Schmid, F., [0068] Handbuch der Kinderheilkunde. Berlin: Springer (pub.) 6 1967. Pp. 351-355) to a disorder similar to van Buchem hyperostosis corticalis generalisata but possibly differing in radiologic appearance of the bone changes and in the presence of asymmetric cutaneous syndactyly of the index and middle fingers in many cases. The jaw has an unusually square appearance in this condition.
  • “Humanized antibodies” are recombinant proteins in which murine or other non-human animal complementary determining regions of monoclonal antibodies have been transferred from heavy and light variable chains of the murine or other non-human animal immunoglobulin into a human variable domain. [0069]
  • As used herein, an “antibody fragment” is a portion of an antibody such as F(ab′)[0070] 2, F(ab)2, Fab′, Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-TGF-beta binding-protein monoclonal antibody fragment binds to an epitope of TGF-beta binding-protein.
  • The term antibody fragment or antigen-binding fragment also includes any synthetic or genetically engineered protein that acts like an antibody by binding to a specific antigen to form a complex. For example, antibody fragments include isolated fragments consisting of the light chain variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (“sFv proteins”), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region. [0071]
  • A “detectable label” is a molecule or atom that can be conjugated to a polypeptide moiety such as an antibody moiety or a nucleic acid moiety to produce a molecule useful for diagnosis. Examples of detectable labels include chelators, photoactive agents, radioisotopes, fluorescent agents, paramagnetic ions, enzymes, and other marker moieties. [0072]
  • As used herein, an “immunoconjugate” is a molecule comprising an anti-TGF-beta binding-protein antibody, or an antibody fragment, and a detectable label or an effector molecule. Preferably, an immunoconjugate has roughly the same, or only slightly reduced, ability to bind TGF-beta binding-protein after conjugation as before conjugation. [0073]
  • Abbreviations: TGF-beta—“Transforming Growth Factor-beta”; TGF-bBP—“Transforming Growth Factor-beta binding-protein” (one representative TGF-bBP is designated “H. Beer”); BMP—“bone morphogenic protein”; PCR—“polymerase chain reaction”; RT-PCR—PCR process in which RNA is first transcribed into DNA using reverse transcriptase (RT); cDNA—any DNA made by copying an RNA sequence into DNA form. [0074]
  • As noted above, the present invention provides a novel class of TGF-beta binding-proteins, as well as methods and compositions for increasing bone mineral content in warn-blooded animals. Briefly, the present inventions are based upon the unexpected discovery that a mutation in the gene which encodes a novel member of the TGF-beta binding-protein family results in a rare condition (sclerosteosis) characterized by bone mineral contents which are one- to four-fold higher than in normal individuals. Thus, as discussed in more detail below this discovery has led to the development of assays which may be utilized to select molecules which inhibit the binding of the TGF-beta binding-protein to the TGF-beta family of proteins and bone morphogenic proteins (BMPs), and methods of utilizing such molecules for increasing the bone mineral content of warm-blooded animals (including for example, humans). [0075]
  • Discussion of the Disease Known as Sclerosteosis [0076]
  • Sclerosteosis is a disease related to abnormal bone mineral density in humans. Sclerosteosis is a term that was applied by Hansen (1967) (Hansen, H. G., Sklerosteose. In: Opitz, H.; Schmid, F., Handbuch der Kinderheilkunde. Berlin: Springer (pub.) 6 1967. Pp. 351-355) to a disorder similar to van Buchem hyperostosis corticalis generalisata but possibly differing in radiologic appearance of the bone changes and differing in the presence of asymmetric cutaneous syndactyly of the index and middle fingers in many cases. [0077]
  • Sclerosteosis is now known to be an autosomal semi-dominant disorder that is characterized by widely disseminated sclerotic lesions of the bone in the adult. The condition is progressive. Sclerosteosis also has a developmental aspect that is associated with syndactyly (two or more fingers are fused together). The Sclerosteosis Syndrome is associated with large stature and many affected individuals attain a height of six feet or more. The bone mineral content of homozygotes can be 1 to 6 fold greater than observed in normal individuals, and bone mineral density can be 1 to 4 fold above normal values (e.g., from unaffected siblings). [0078]
  • The Sclerosteosis Syndrome occurs primarily in Afrikaaners of Dutch descent in South Africa. Approximately {fraction (1/140)} individuals in the Afrikaaner population are carriers of the mutated gene (heterozygotes). The mutation shows 100% penetrance. There are anecdotal reports of increased of bone mineral density in heterozygotes with no associated pathologies (syndactyly or skull overgrowth). [0079]
  • No abnormality of the pituitary-hypothalamus axis has been observed in patients with sclerosteosis. In particular, there appears to be no over-production of growth hormone and cortisone. In addition, sex hormone levels are normal in affected individuals. However, bone turnover markers (osteoblast specific alkaline phosphatase, osteocalcin, [0080] type 1 procollagen C′ propeptide (PICP), and total alkaline phosphatase; (see Comier, C., Curr. Opin. in Rheu. 7:243, 1995) indicate that there is hyperosteoblastic activity associated with the disease but that there is normal to slightly decreased osteoclast activity as measured by markers of bone resorption (pyridinoline, deoxypryridinoline, N-telopeptide, urinary hydroxyproline, plasma tartrate-resistant acid phosphatases and galactosyl hydroxylysine (see Comier, supra)).
  • Sclerosteosis is characterized by the continual deposition of bone throughout the skeleton during the lifetime of the affected individuals. In homozygotes the continual deposition of bone mineral leads to an overgrowth of bone in areas of the skeleton where there is an absence of mechanoreceptors (skull, jaw, cranium). In homozygotes with Sclerosteosis, the overgrowth of the bones of the skull leads to cranial compression and eventually to death due to excessive hydrostatic pressure on the brain stem. In all other parts of the skeleton there is a generalized and diffuse sclerosis. Cortical areas of the long bones are greatly thickened resulting in a substantial increase in bone strength. Trabecular connections are increased in thickness which in turn increases the strength of the trabecular bone. Sclerotic bones appear unusually opaque to x-rays. [0081]
  • As described in more detail in Example 1, the rare genetic mutation that is responsible for the Sclerosteosis syndrome has been localized to the region of human chromosome 17 that encodes a novel member of the TGF-beta binding-protein family (one representative example of which is designated “H. Beer”). As described in more detail below, based upon this discovery, the mechanism of bone mineralization is more fully understood, allowing the development of assays for molecules that increase bone mineralization, and use of such molecules to increase bone mineral content, and in the treatment or prevention of a wide number of diseases. [0082]
  • TGF-Beta Super-Family [0083]
  • The Transforming Growth Factor-beta (TGF-beta) super-family contains a variety of growth factors that share common sequence elements and structural motifs (at both the secondary and tertiary levels). This protein family is known to exert a wide spectrum of biological responses that affect a large variety of cell types. Many of the TGF-beta family members have important functions during the embryonal development in pattern formation and tissue specification; in adults the family members are involved, e.g., in wound healing and bone repair and bone remodeling, and in the modulation of the immune system. In addition to the TGF-beta's, the super-family includes the Bone Morphogenic Proteins (BMPs), Activins, Inhibins, Growth and Differentiation Factors (GDFs), and Glial-Derived Neurotrophic Factors (GDNFs). Primary classification is established through general sequence features that bin a specific protein into a general sub-family. Additional stratification within the sub-family is possible due to stricter sequence conservation between members of the smaller group. In certain instances, such as with BMP-5, BMP-6 and BMP-7, the amino acid identity can be as high as 75% among members of the smaller group. This level of identity enables a single representative sequence to illustrate the key biochemical elements of the sub-group that separates it from other members of the larger family. [0084]
  • The crystal structure of TGF-beta2 has been determined. The general fold of the TGF-beta2 monomer contains a stable, compact, cysteine knotlike structure formed by three disulphide bridges. Dimerization, stabilized by one disulfide bridge, is antiparallel. [0085]
  • TGF-beta signals by inducing the formation of hetero-oligomeric complexes of type I and type II receptors. Transduction of TGF-beta signals involves these two distinct type I and type II subfamilies of transmembrane serine/threonine kinase receptors. At least seven type I receptors and five type II receptors have been identified (see Kawabata et al., [0086] Cytokine Growth Factor Rev. 9:49-61 (1998); Miyazono et al., Adv. Immunol. 75:115-57 (2000). TGF-beta family members initiate their cellular action by binding to receptors with intrinsic serine/threonine kinase activity. Each member of the TGF-beta family binds to a characteristic combination of type I and type II receptors, both of which are needed for signaling. In the current model for TGF-beta receptor activation, a TGF-beta ligand first binds to the type II receptor (TbR-II), which occurs in the cell membrane in an oligomeric form with activated kinase. Thereafter, the type I receptor (TbR-I), which cannot bind ligand in the absence of TbR-II, is recruited into the complex to form a ligand/type II/type I ternary complex. TbR-II then phosphorylates TbR-I predominantly in a domain rich in glycine and serine residues (GS domain) in the juxtamembrane region, and thereby activates TbR-I. The activated type I receptor kinase then phosphorylates particular members of the Smad family of proteins that translocate to the nucleus where they modulate transcription of specific genes.
  • Bone Morphogenic Proteins (BMPS) are Key Regulatory Proteins in Determining Bone Mineral Density in Humans [0087]
  • A major advance in the understanding of bone formation was the identification of the bone morphogenic proteins (BMPs), also known as osteogenic proteins (OPs), which regulate cartilage and bone differentiation in vivo. BMPs/OPs induce endochondral bone differentiation through a cascade of events that include formation of cartilage, hypertrophy and calcification of the cartilage, vascular invasion, differentiation of osteoblasts, and formation of bone. As described above, the BMPs/OPs (BMP 2-14, and [0088] osteogenic protein 1 and −2, OP-1 and OP-2) see, e.g., GenBank P12643 (BMP-2); GenBank P12645 (BMP3); GenBank P55107 (BMP-3b, Growth/differentiation factor 10) (GDF-10)); GenBank P12644 (BMP4); GenBank P22003 (BMP5); GenBank P22004 (BMP6); GenBank P18075 (BMP7); GenBank P34820 (BMP8); GenBank Q9UK05 (BMP9); GenBank O95393 (BM10); GenBank O95390 (BMP11, Growth/differentiation factor 11 precursor (GDF-11)); GenBank O95972 (BM15)) are members of the TGF-beta super-family. The striking evolutionary conservation between members the BMP/OP sub-family suggests that they are critical in the normal development and function of animals. Moreover, the presence of multiple forms of BMPs/OPs raises an important question about the biological relevance of this apparent redundancy. In addition to postfetal chondrogenesis and osteogenesis, the BMPs/OPs play multiple roles in skeletogenesis (including the development of craniofacial and dental tissues) and in embryonic development and organogenesis of parenchymatous organs, including the kidney. It is now understood that nature relies on common (and few) molecular mechanisms tailored to provide the emergence of specialized tissues and organs. The BMP/OP super-family is an elegant example of nature parsimony in programming multiple specialized functions deploying molecular isoforms with minor variation in amino acid motifs within highly conserved carboxy-terminal regions.
  • BMPs are synthesized as large precursor proteins. Upon dimerization, the BMPs are proteolyically cleaved within the cell to yield carboxy-terminal mature proteins that are then secreted from the cell. BMPs, like other TGF-beta family members, initiate signal transduction by binding cooperatively to both type I and type II serine/threonine kinase receptors. Type I receptors for which BMPs may act as ligands include BMPR-IA (also known as ALK-3), BMPR-IB (also known as ALK-6), ALK-1, and ALK-2 (also known as ActR-1). Of the type II receptors, BMPs bind to BMP type II receptor (BMPR-II), Activin type II (ActR-II), and Activin type IIB (ActR-IIB). (See Balemans et al., supra, and references cited therein). Polynucleotide sequences and the encoded amino acid sequence of BMP type I receptor polypeptides are provided in the GenBank database, for example, GenBank NM[0089] 004329 (SEQ ID NO: 102 encoded by SEQ ID NO: 116); D89675 (SEQ ID NO: 103 encoded by SEQ ID NO: 117); NM001203 (SEQ ID NO: 104 encoded by SEQ ID NO: 118); S75359 (SEQ ID NO: 105 encoded by SEQ ID NO: 119); NM030849 (SEQ ID NO: 106 encoded by SEQ ID NO: 120); and D38082 (SEQ ID NO: 107 encoded by SEQ ID NO: 121). Other polypeptide sequences of type I receptors are provided in the GenBank database, for example, NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); and AAB33865 (SEQ ID NO: 110). Polynucleotide sequences and the encoded amino acid sequence of BMP type II receptor polypeptides are provided in the GenBank database and include, for example, U25110 (SEQ ID NO: 111 encoded by SEQ ID NO: 122); NM033346 (SEQ ID NO: 112 encoded by SEQ ID NO: 123); NM001204 (SEQ ID NO: 113 encoded by SEQ ID NO: 124); and Z48923 (SEQ ID NO: 114 encoded by SEQ ID NO: 125). Additional polypeptide sequences of type II receptors are also provided in the GenBank database, for example, CAA88759 (SEQ ID NO: 115).
  • BMPs, similar to other cystine-knot proteins, form a homodimer structure (Scheufler et al., [0090] J. Mol. Biol. 287:103-15 (1999)). According to evolutionary trace analysis performed on the BMP/TGF-β family, the BMP type I receptor binding site and type II receptor binding site were mapped to the surface of the BMP structure (Innis et al., Protein Eng. 13:839-47 (2000)). The location of the type I receptor binding site on BMP was later confirmed by the x-ray structure of BMP-2/BMP Receptor IA complex (Nickel et al., J. Joint Surg. Am. 83A(Suppl 1(Pt 1)):S7-S14 (2001)). The predicted type II receptor binding site is in good agreement with the x-ray structure of TGF-β3/TGF-β Type II receptor complex (Hart et al., Nat. Struct. Biol. 9:203-208 (2002)), which is highly similar to the BMP/BMP Receptor IIA system.
  • BMP Antagonism [0091]
  • The BMP and Activin sub-families are subject to significant post-translational regulation, such as by TGF-beta binding proteins. An intricate extracellular control system exists, whereby a high affinity antagonist is synthesized and exported, and subsequently complexes selectively with BMPs or activins to disrupt their biological activity (W. C. Smith (1999) TIG 15(1) 3-6). A number of these natural antagonists have been identified, and on the basis of sequence divergence, the antagonists appear to have evolved independently due to the lack of primary sequence conservation. Earlier studies of these antagonists highlighted a distinct preference for interacting and neutralizing BMP-2 and BMP-4. In vertebrates, antagonists include noggin, chordin, chordin-like, follistatin, FSRP, the DAN/Cerberus protein family, and sclerostin (SOST) (see Balemans et al., supra, and references cited therein). The mechanism of antagonism or inhibition seems to differ for the different antagonists (Iemura et al. (1998) [0092] Proc. Natl. Acad. Sci. USA 95 9337-9342).
  • The type I and type II receptor binding sites on the BMP antagonist noggin have also been mapped. Noggin binds to BMPs with high affinity (Zimmerman et al., 1996). A study of the noggin/BMP-7 complex structure revealed the binding interactions between the two proteins (Groppe et al., [0093] Nature 420:636-42 (2002)). Superposition of the noggin-BMP-7 structure onto a model of the BMP signaling complex showed that noggin binding effectively masks both pairs of binding epitopes (i.e., BMP Type I and Type II receptor binding sites) on BMP-7. The cysteine-rich scaffold sequence of noggin is preceded by an N-terminal segment of about 20 amino acid residues that are referred to as the “clip” (residues 28-48). The type I receptor-binding site is occluded by the N-terminal portion of the clip domain of Noggin, and the type II receptor binding site is occluded by the carboxy terminal portion of the clip domain. Two β-strands in the core region near the C-terminus of noggin also contact BMP-7 at the type II receptor binding site. This binding mode enables a noggin dimer to efficiently block all the receptor binding sites (two type I and two type II receptor binding sites) on a BMP dimer.
  • Novel TGF-Beta Binding-Proteins [0094]
  • As noted above, the present invention provides a novel class of TGF-beta binding-proteins that possess a nearly identical cysteine (disulfide) scaffold when compared to Human DAN, Human Gremlin, and Human Cerberus, and SCGF (U.S. Pat. No. 5,780,263) but almost no homology at the nucleotide level. (for background information, see generally Hsu, D. R., Economides, A. N., Wang, X., Eimon, P. M., Harland, R. M., “The Xenopus Dorsalizing Factor Gremlin Identifies a Novel Family of Secreted Proteins that Antagonize BMP Activities,” [0095] Molecular Cell 1:673-683, 1998).
  • Representative example of the novel class of nucleic acid molecules encoding TGF-beta binding-proteins are disclosed in SEQ ID NOS: 1, 5, 7, 9, 11, 13, 15, 100, and 101. The polynucleotides disclosed herein encode a polypeptide called Beer, which is also referred to herein as sclerostin or SOST. Representative members of this class of binding proteins should also be understood to include variants of the TGF-beta binding-protein (e.g., SEQ ID NOS: 5 and 7). As utilized herein, a “TGF-beta binding-protein variant gene” (e.g., an isolated nucleic acid molecule that encodes a TGF-beta binding protein variant) refers to nucleic acid molecules that encode a polypeptide having an amino acid sequence that is a modification of SEQ ID NOS: 2, 10, 12, 14, 16, 46, or 65. Such variants include naturally-occurring polymorphisms or allelic variants of TGF-beta binding-protein genes, as well as synthetic genes that contain conservative amino acid substitutions of these amino acid sequences. A variety of criteria known to those skilled in the art indicate whether amino acids at a particular position in a peptide or polypeptide are similar. For example, a similar amino acid or a conservative amino acid substitution is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain, which include amino acids with basic side chains (e.g., lysine, arginine, histidine); acidic side chains (e.g., aspartic acid, glutamic acid); uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, histidine); nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); beta-branched side chains (e.g., threonine, valine, isoleucine), and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan). Proline, which is considered more difficult to classify, shares properties with amino acids that have aliphatic side chains (e.g., Leu, Val, Ile, and Ala). In certain circumstances, substitution of glutamine for glutamic acid or asparagine for aspartic acid may be considered a similar substitution in that glutamine and asparagine are amide derivatives of glutamic acid and aspartic acid, respectively. [0096]
  • Additional variant forms of a TGF-beta binding-protein gene are nucleic acid molecules that contain insertions or deletions of the nucleotide sequences described herein. TGF-beta binding-protein variant genes can be identified by determining whether the genes hybridize with a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 1, 5, 7, 9, 11, 13, 15, 100, or 101 under stringent conditions. In addition, TGF-beta binding-protein variant genes should encode a protein having a cysteine backbone. [0097]
  • As an alternative, TGF-beta binding-protein variant genes can be identified by sequence comparison. As used herein, two amino acid sequences have “100% amino acid sequence identity” if the amino acid residues of the two amino acid sequences are the same when aligned for maximal correspondence. Similarly, two nucleotide sequences have “100% nucleotide sequence identity” if the nucleotide residues of the two nucleotide sequences are the same when aligned for maximal correspondence. Sequence comparisons can be performed using standard software programs such as those included in the LASERGENE bioinformatics computing suite, which is produced by DNASTAR (Madison, Wis.). Other methods for comparing two nucleotide or amino acid sequences by determining optimal alignment are well-known to those of skill in the art (see, for example, Peruski and Peruski, [0098] The Internet and the New Biology: Tools for Genomic and Molecular Research (ASM Press, Inc. 1997), Wu et al. (eds.), “Information Superhighway and Computer Databases of Nucleic Acids and Proteins,” in Methods in Gene Biotechnology, pages 123-151 (CRC Press, Inc. 1997), and Bishop (ed.), Guide to Human Genome Computing, 2nd Edition (Academic Press, Inc. 1998)).
  • A variant TGF-beta binding-protein should have at least a 50% amino acid sequence identity to SEQ ID NOS: 2, 6, 10, 12, 14, 16, 46, or 65 and preferably, greater than 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity. Alternatively, TGF-beta binding-protein variants can be identified by having at least a 70% nucleotide sequence identity to SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101. Moreover, the present invention contemplates TGF-beta binding-protein gene variants having greater than 75%, 80%, 85%, 90%, or 95% identity to SEQ ID NO: 1 or SEQ ID NO: 100. Regardless of the particular method used to identify a TGF-beta binding-protein variant gene or variant TGF-beta binding-protein, a variant TGF-beta binding-protein or a polypeptide encoded by a variant TGF-beta binding-protein gene can be functionally characterized by, for example, its ability to bind to and/or inhibit the signaling of a selected member of the TGF-beta family of proteins, or by its ability to bind specifically to an anti-TGF-beta binding-protein antibody. [0099]
  • The present invention includes functional fragments of TGF-beta binding-protein genes. Within the context of this invention, a “functional fragment” of a TGF-beta binding-protein gene refers to a nucleic acid molecule that encodes a portion of a TGF-beta binding-protein polypeptide which either (1) possesses the above-noted function activity, or (2) specifically binds with an anti-TGF-beta binding-protein antibody. For example, a functional fragment of a TGF-beta binding-protein gene described herein comprises a portion of the nucleotide sequence of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101. [0100]
  • 2. Isolation of the TGF-beta Binding-Protein Gene [0101]
  • DNA molecules encoding a TGF-beta binding-protein can be obtained by screening a human cDNA or genomic library using polynucleotide probes based upon, for example, SEQ ID NO: 1. For example, the first step in the preparation of a cDNA library is to isolate RNA using methods well-known to those of skill in the art. In general, RNA isolation techniques provide a method for breaking cells, a means of inhibiting RNase-directed degradation of RNA, and a method of separating RNA from DNA, protein, and polysaccharide contaminants. For example, total RNA can be isolated by freezing tissue in liquid nitrogen, grinding the frozen tissue with a mortar and pestle to lyse the cells, extracting the ground tissue with a solution of phenol/chloroform to remove proteins, and separating RNA from the remaining impurities by selective precipitation with lithium chloride (see, for example, Ausubel et al. (eds.), [0102] Short Protocols in Molecular Biology, 3rd Edition, pages 4-1 to 4-6 (John Wiley & Sons 1995) [“Ausubel (1995)”]; Wu et al., Methods in Gene Biotechnology, pages 33-41 (CRC Press, Inc. 1997) [“Wu (1997)”]). Alternatively, total RNA can be isolated by extracting ground tissue with guanidinium isothiocyanate, extracting with organic solvents, and separating RNA from contaminants using differential centrifugation (see, for example, Ausubel (1995) at pages 4-1 to 4-6; Wu (1997) at pages 33-41).
  • In order to construct a cDNA library, poly(A)[0103] + RNA is preferably isolated from a total RNA preparation. Poly(A)+ RNA can be isolated from total RNA by using the standard technique of oligo(dT)-cellulose chromatography (see, for example, Ausubel (1995) at pages 4-11 to 4-12). Double-stranded cDNA molecules may be synthesized from poly(A)+ RNA using techniques well-known to those in the art. (see, for example, Wu (1997) at pages 41-46). Moreover, commercially available kits can be used to synthesize double-stranded cDNA molecules (for example, Life Technologies, Inc. (Gaithersburg, Md.); CLONTECH Laboratories, Inc. (Palo Alto, Calif.); Promega Corporation (Madison, Wis.); and Stratagene Cloning Systems (La Jolla, Calif.)).
  • The basic approach for obtaining TGF-beta binding-protein cDNA clones can be modified by constructing a subtracted cDNA library that is enriched in TGF-binding-protein-specific cDNA molecules. Techniques for constructing subtracted libraries are well-known to those of skill in the art (see, for example, Sargent, “Isolation of Differentially Expressed Genes,” in [0104] Meth. Enzymol. 152:423, 1987; and Wu et al. (eds.), “Construction and Screening of Subtracted and Complete Expression cDNA Libraries,” in Methods in Gene Biotechnology, pages 29-65 (CRC Press, Inc. 1997)).
  • Various cloning vectors are appropriate for the construction of a cDNA library. For example, a cDNA library can be prepared in a vector derived from bacteriophage, such as a λgt10 vector (see, for example, Huynh et al., “Constructing and Screening cDNA Libraries in λgt10 and λgt11, ” in [0105] DNA Cloning: A Practical Approach Vol. I, Glover (ed.), page 49 (IRL Press, 1985); Wu (1997) at pages 47-52). Alternatively, double-stranded cDNA molecules can be inserted into a plasmid vector, such as a pBluescript vector (Stratagene Cloning Systems; La Jolla, Calif.), a LambdaGEM-4 (Promega Corp.; Madison, Wis.) or other commercially available vectors. Suitable cloning vectors also can be obtained from the American Type Culture Collection (Rockville, Md.).
  • In order to amplify the cloned cDNA molecules, the cDNA library is inserted into a prokaryotic host, using standard techniques. For example, a cDNA library can be introduced into competent [0106] E. coli DH5 cells, which can be obtained from Life Technologies, Inc. (Gaithersburg, Md.).
  • A human genomic DNA library can be prepared by means well-known in the art (see, for example, Ausubel (1995) at pages 5-1 to 5-6; Wu (1997) at pages 307-327). Genomic DNA can be isolated by lysing tissue with the detergent Sarkosyl, digesting the lysate with proteinase K, clearing insoluble debris from the lysate by centrifugation, precipitating nucleic acid from the lysate using isopropanol, and purifying resuspended DNA on a cesium chloride density gradient. [0107]
  • DNA fragments that are suitable for the production of a genomic library can be obtained by the random shearing of genomic DNA or by the partial digestion of genomic DNA with restriction endonucleases. Genomic DNA fragments can be inserted into a vector, such as a bacteriophage or cosmid vector, in accordance with conventional techniques, such as the use of restriction enzyme digestion to provide appropriate termini, the use of alkaline phosphatase treatment to avoid undesirable joining of DNA molecules, and ligation with appropriate ligases. Techniques for such manipulation are well-known in the art (see, for example, Ausubel (1995) at pages 5-1 to 5-6; Wu (1997) at pages 307-327). [0108]
  • Nucleic acid molecules that encode a TGF-beta binding-protein can also be obtained using the polymerase chain reaction (PCR) with oligonucleotide primers having nucleotide sequences that are based upon the nucleotide sequences of the human TGF-beta binding-protein gene, as described herein. General methods for screening libraries with PCR are provided by, for example, Yu et al., “Use of the Polymerase Chain Reaction to Screen Phage Libraries,” in [0109] Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, White (ed.), pages 211 -215 (Humana Press, Inc. 1993). Moreover, techniques for using PCR to isolate related genes are described by, for example, Preston, “Use of Degenerate Oligonucleotide Primers and the Polymerase Chain Reaction to Clone Gene Family Members,” in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, White (ed.), pages 317-337 (Humana Press, Inc. 1993).
  • Alternatively, human genomic libraries can be obtained from commercial sources such as Research Genetics (Huntsville, Ala.) and the American Type Culture Collection (Rockville, Md.). A library containing cDNA or genomic clones can be screened with one or more polynucleotide probes based upon SEQ ID NO: 1, using standard methods as described herein and known in the art (see, for example, Ausubel (1995) at pages 6-1 to 6-11). [0110]
  • Anti-TGF-beta binding-protein antibodies, produced as described herein, can also be used to isolate DNA sequences that encode a TGF-beta binding-protein from cDNA libraries. For example, the antibodies can be used to screen λgt11 expression libraries, or the antibodies can be used for immunoscreening following hybrid selection and translation (see, for example, Ausubel (1995) at pages 6-12 to 6-16; Margolis et al., “Screening λ expression libraries with antibody and protein probes,” in [0111] DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), pages 1- 14 (Oxford University Press 1995)).
  • The sequence of a TGF-beta binding-protein cDNA or TGF-beta binding-protein genomic fragment can be determined using standard methods. Moreover, the identification of genomic fragments containing a TGF-beta binding-protein promoter or regulatory element can be achieved using well-established techniques, such as deletion analysis (see generally Ausubel (1995), supra). [0112]
  • As an alternative, a TGF-beta binding-protein gene can be obtained by synthesizing DNA molecules using mutually priming long oligonucleotides and the nucleotide sequences described herein (see, for example, Ausubel (1995) at pages 8-8 to 8-9). Established techniques using the polymerase chain reaction provide the ability to synthesize DNA molecules at least two kilobases in length (Adang et al., [0113] Plant Molec. Biol. 21:1131, 1993; Bambot et al., PCR Methods and Applications 2:266, 1993; Dillon et al., “Use of the Polymerase Chain Reaction for the Rapid Construction of Synthetic Genes,” in Methods in Molecular Biology, Vol. 15: PCR Protocols: Current Methods and Applications, White (ed.), pages 263-268, (Humana Press, Inc. 1993); Holowachuk et al., PCR Methods Appl. 4:299, 1995).
  • 3. Production of TGF-Beta Binding-Protein Genes [0114]
  • Nucleic acid molecules encoding variant TGF-beta binding-protein genes can be obtained by screening various cDNA or genomic libraries with polynucleotide probes having nucleotide sequences based upon SEQ ID NO: 1, 5, 9, 11, 13, 15, 100, or 101 using procedures described herein. TGF-beta binding-protein gene variants can also be constructed synthetically. For example, a nucleic acid molecule can be devised that encodes a polypeptide having a conservative amino acid change, compared with the amino acid sequence of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65. That is, variants can be obtained that contain one or more amino acid substitutions of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65, in which an alkyl amino acid is substituted for an alkyl amino acid in a TGF-beta binding-protein amino acid sequence, an aromatic amino acid is substituted for an aromatic amino acid in a TGF-beta binding-protein amino acid sequence, a sulfur-containing amino acid is substituted for a sulfur-containing amino acid in a TGF-beta binding-protein amino acid sequence, a hydroxy-containing amino acid is substituted for a hydroxy-containing amino acid in a TGF-beta binding-protein amino acid sequence, an acidic amino acid is substituted for an acidic amino acid in a TGF-beta binding-protein amino acid sequence, a basic amino acid is substituted for a basic amino acid in a TGF-beta binding-protein amino acid sequence, or a dibasic monocarboxylic amino acid is substituted for a dibasic monocarboxylic amino acid in a TGF-beta binding-protein amino acid sequence. Among the common amino acids, for example, a “conservative amino acid substitution” is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine. In making such substitutions, it is important, when possible, to maintain the cysteine backbone outlined in FIG. 1. [0115]
  • Conservative amino acid changes in a TGF-beta binding-protein gene can be introduced by substituting nucleotides for the nucleotides recited in SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101. Such “conservative amino acid” variants can be obtained, for example, by oligonucleotide-directed mutagenesis, linker-scanning mutagenesis, mutagenesis using the polymerase chain reaction, and the like (see Ausubel (1995) at pages 8-10 to 8-22; McPherson (ed.), [0116] Directed Mutagenesis: A Practical Approach (IRL Press 1991)). The functional ability of such variants can be determined using a standard method, such as the assay described herein. Alternatively, a variant TGF-beta binding-protein polypeptide can be identified by the ability to specifically bind anti-TGF-beta binding-protein antibodies.
  • Routine deletion analyses of nucleic acid molecules can be performed to obtain “functional fragments” of a nucleic acid molecule that encodes a TGF-beta binding-protein polypeptide. As an illustration, DNA molecules having the nucleotide sequence of SEQ ID NO: 1 can be digested with Bal31 nuclease to obtain a series of nested deletions. The fragments are then inserted into expression vectors in proper reading frame, and the expressed polypeptides are isolated and tested for activity, or for the ability to bind anti-TGF-beta binding-protein antibodies. One alternative to exonuclease digestion is to use oligonucleotide-directed mutagenesis to introduce deletions or stop codons to specify production of a desired fragment. Alternatively, particular fragments of a TGF-beta binding-protein gene can be synthesized using the polymerase chain reaction. [0117]
  • Standard techniques for functional analysis of proteins are described by, for example, Treuter et al., [0118] Molec. Gen. Genet. 240:113, 1993; Content et al., “Expression and preliminary deletion analysis of the 42 kDa 2-5A synthetase induced by human interferon,” in Biological Interferon Systems, Proceedings of ISIR-TNO Meeting on Interferon Systems, Cantell (ed.), pages 65-72 (Nijhoff 1987); Herschman, “The EGF Receptor,” in Control of Animal Cell Proliferation, Vol. 1, Boynton et al., (eds.) pages 169-199 (Academic Press 1985); Coumailleau et al., J. Biol. Chem. 270:29270, 1995; Fukunaga et al., J. Biol. Chem. 270:25291, 1995; Yamaguchi et al., Biochem. Pharmacol. 50:1295, 1995; Meisel et al., Plant Molec. Biol. 30:1, 1996.
  • The present invention also contemplates functional fragments of a TGF-beta binding-protein gene that have conservative amino acid changes. [0119]
  • A TGF-beta binding-protein variant gene can be identified on the basis of structure by determining the level of identity with nucleotide and amino acid sequences of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101 and 2, 6, 10, 12, 14, 16, 46, or 65 as discussed above. An alternative approach to identifying a variant gene on the basis of structure is to determine whether a nucleic acid molecule encoding a potential variant TGF-beta binding-protein gene can hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 1, 5, 9, 11, 13, 15, 100, or 101, or a portion thereof of at least 15 or 20 nucleotides in length. As an illustration of stringent hybridization conditions, a nucleic acid molecule having a variant TGF-beta binding-protein sequence can bind with a fragment of a nucleic acid molecule having a sequence from SEQ ID NO: 1 in a buffer containing, for example, 5× SSPE (1× SSPE=180 mM sodium chloride, 10 mM sodium phosphate, 1 mM EDTA (pH 7.7), 5× Denhardt's solution (100× Denhardt's=2% (w/v) bovine serum albumin, 2% (w/v) Ficoll, 2% (w/v) polyvinylpyrrolidone) and 0.5% SDS incubated overnight at 55-60° C. Post-hybridization washes at high stringency are typically performed in 0.5×SSC (1×SSC=150 mM sodium chloride, 15 mM trisodium citrate) or in 0.5× SSPE at 55-60° C. [0120]
  • Regardless of the particular nucleotide sequence of a variant TGF-beta binding-protein gene, the gene encodes a polypeptide that can be characterized by its functional activity, or by the ability to bind specifically to an anti-TGF-beta binding-protein antibody. More specifically, variant TGF-beta binding-protein genes encode polypeptides which exhibit at least 50%, and preferably, greater than 60, 70, 80 or 90%, of the activity of polypeptides encoded by the human TGF-beta binding-protein gene described herein. [0121]
  • 4. Production of TGF-Beta Binding-Protein in Cultured Cells [0122]
  • To express a TGF-beta binding-protein gene, a nucleic acid molecule encoding the polypeptide must be operably linked to regulatory sequences that control transcriptional expression in an expression vector and then introduced into a host cell. In addition to transcriptional regulatory sequences, such as promoters and enhancers, expression vectors can include translational regulatory sequences and a marker gene that is suitable for selection of cells that carry the expression vector. Expression vectors that are suitable for production of a foreign protein in eukaryotic cells typically contain (1) prokaryotic DNA elements coding for a bacterial replication origin and an antibiotic resistance marker to provide for the growth and selection of the expression vector in a bacterial host; (2) eukaryotic DNA elements that control initiation of transcription, such as a promoter; and (3) DNA elements that control the processing of transcripts, such as a transcription termination/polyadenylation sequence. [0123]
  • TGF-beta binding-proteins of the present invention are preferably expressed in mammalian cells. Examples of mammalian host cells include African green monkey kidney cells (Vero; ATCC CRL 1587), human embryonic kidney cells (293-HEK; ATCC CRL 1573), baby hamster kidney cells (BHK-21; ATCC CRL 8544), canine kidney cells (MDCK; ATCC CCL 34), Chinese hamster ovary cells (CHO-K1; ATCC CCL61), rat pituitary cells (GH1; ATCC CCL82), HeLa S3 cells (ATCC CCL2.2), rat hepatoma cells (H-4-II-E; ATCC CRL 1548) SV40-transformed monkey kidney cells (COS-1; ATCC CRL 1650) and murine embryonic cells (NIH-3T3; ATCC CRL 1658). [0124]
  • For a mammalian host, the transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a particular gene which has a high level of expression. Suitable transcriptional and translational regulatory sequences also can be obtained from mammalian genes, such as actin, collagen, myosin, and metallothionein genes. [0125]
  • Transcriptional regulatory sequences include a promoter region sufficient to direct the initiation of RNA synthesis. Suitable eukaryotic promoters include the promoter of the mouse metallothionein I gene [Hamer et al., [0126] J. Molec. Appl. Genet. 1:273, 1982], the TK promoter of Herpes virus [McKnight, Cell 31:355, 1982], the SV40 early promoter [Benoist et al., Nature 290:304, 1981], the Rous sarcoma virus promoter [Gorman et al., Proc. Nat'l Acad. Sci. USA 79:6777, 1982], the cytomegalovirus promoter [Foecking et al., Gene 45:101, 1980], and the mouse mammary tumor virus promoter (see, generally, Etcheverry, “Expression of Engineered Proteins in Mammalian Cell Culture,” in Protein Engineering: Principles and Practice, Cleland et al. (eds.), pages 163-181 (John Wiley & Sons, Inc. 1996)).
  • Alternatively, a prokaryotic promoter, such as the bacteriophage T3 RNA polymerase promoter, can be used to control TGF-beta binding-protein gene expression in mammalian cells if the prokaryotic promoter is regulated by a eukaryotic promoter (Zhou et al., [0127] Mol. Cell. Biol. 10:4529, 1990; Kaufman et al., Nucleic Acids Res. 19:4485, 1991).
  • TGF-beta binding-protein genes may also be expressed in bacterial, yeast, insect, or plant cells. Suitable promoters that can be used to express TGF-beta binding-protein polypeptides in a prokaryotic host are well-known to those of skill in the art and include promoters capable of recognizing the T4, T3, Sp6 and T7 polymerases, the P[0128] R and PL promoters of bacteriophage lambda, the trp, recA, heat shock, lacUV5, tac, lpp-lacSpr, phoA, and lacZ promoters of E. coli, promoters of B. subtilis, the promoters of the bacteriophages of Bacillus, Streptomyces promoters, the int promoter of bacteriophage lambda, the bla promoter of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene. Prokaryotic promoters have been reviewed by Glick, J. Ind. Microbiol. 1:277, 1987, Watson et al., Molecular Biology of the Gene, 4th Ed. (Benjamin Cummins 1987), and by Ausubel et al. (1995).
  • Preferred prokaryotic hosts include [0129] E. coli and Bacillus subtilus. Suitable strains of E. coli include BL21(DE3), BL21(DE3)pLysS, BL21(DE3)pLysE, DH1, DH4I, DH5, DH5I, DH5IF′, DH5IMCR, DH10B, DH10B/p3, DH11S, C600, HB101, JM101, JM105, JM109, JM110, K38, RR1, Y1088, Y1089, CSH18, ER1451, and ER1647 (see, for example, Brown (Ed.), Molecular Biology Labfax (Academic Press 1991)). Suitable strains of Bacillus subtilus include BR151, YB886, MI119, MI120, and B170 (see, for example, Hardy, “Bacillus Cloning Methods,” in DNA Cloning: A Practical Approach, Glover (Ed.) (IRL Press 1985)).
  • Methods for expressing proteins in prokaryotic hosts are well-known to those of skill in the art (see, for example, Williams et al., “Expression of foreign proteins in [0130] E. coli using plasmid vectors and purification of specific polyclonal antibodies,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 15 (Oxford University Press 1995); Ward et al., “Genetic Manipulation and Expression of Antibodies,” in Monoclonal Antibodies: Principles and Applications, page 137 (Wiley-Liss, Inc. 1995); and Georgiou, “Expression of Proteins in Bacteria,” in Protein Engineering: Principles and Practice, Cleland et al. (eds.), page 101 (John Wiley & Sons, Inc. 1996)).
  • The baculovirus system provides an efficient means to introduce cloned TGF-beta binding-protein genes into insect cells. Suitable expression vectors are based upon the [0131] Autographa californica multiple nuclear polyhedrosis virus (AcMNPV), and contain well-known promoters such as Drosophila heat shock protein (hsp) 70 promoter, Autographa californica nuclear polyhedrosis virus immediate-early gene promoter (ie-1) and the delayed early 39K promoter, baculovirus p10 promoter, and the Drosophila metallothionein promoter. Suitable insect host cells include cell lines derived from IPLB-Sf-21, a Spodoptera frugiperda pupal ovarian cell line, such as Sf9 (ATCC CRL 1711), Sf21AE, and Sf21 (Invitrogen Corporation; San Diego, Calif.), as well as Drosophila Schneider-2 cells. Established techniques for producing recombinant proteins in baculovirus systems are provided by Bailey et al., “Manipulation of Baculovirus Vectors,” in Methods in Molecular Biology, Volume 7: Gene Transfer and Expression Protocols, Murray (ed.), pages 147-168 (The Humana Press, Inc. 1991), by Patel et al., “The baculovirus expression system,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al (eds.), pages 205-244 (Oxford University Press 1995), by Ausubel (1995) at pages 16-37 to 16-57, by Richardson (ed.), Baculovirus Expression Protocols (The Humana Press, Inc. 1995), and by Lucknow, “Insect Cell Expression Technology,” in Protein Engineering: Principles and Practice, Cleland et al. (eds.), pages 183-218 (John Wiley & Sons, Inc. 1996).
  • Promoters for expression in yeast include promoters from GAL1 (galactose), PGK (phosphoglycerate kinase), ADH (alcohol dehydrogenase), AOX1 (alcohol oxidase), HIS4 (histidinol dehydrogenase), and the like. Many yeast cloning vectors have been designed and are readily available. These vectors include YIp-based vectors, such as YIp5, YRp vectors, such as YRp17, YEp vectors such as YEp13 and YCp vectors, such as YCp19. One skilled in the art will appreciate that there are a wide variety of suitable vectors for expression in yeast cells. [0132]
  • Expression vectors can also be introduced into plant protoplasts, intact plant tissues, or isolated plant cells. General methods of culturing plant tissues are provided, for example, by Miki et al., “Procedures for Introducing Foreign DNA into Plants,” in [0133] Methods in Plant Molecular Biology and Biotechnology, Glick et al. (eds.), pages 67-88 (CRC Press, 1993).
  • An expression vector can be introduced into host cells using a variety of standard techniques including calcium phosphate transfection, liposome-mediated transfection, microprojectile-mediated delivery, electroporation, and the like. Preferably, the transfected cells are selected and propagated to provide recombinant host cells that comprise the expression vector stably integrated in the host cell genome. Techniques for introducing vectors into eukaryotic cells and techniques for selecting such stable transformants using a dominant selectable marker are described, for example, by Ausubel (1995) and- by Murray (ed.), [0134] Gene Transfer and Expression Protocols (Humana Press 1991). Methods for introducing expression vectors into bacterial, yeast, insect, and plant cells are also provided by Ausubel (1995).
  • General methods for expressing and recovering foreign protein produced by a mammalian cell system is provided by, for example, Etcheverry, “Expression of Engineered Proteins in Mammalian Cell Culture,” in [0135] Protein Engineering: Principles and Practice, Cleland et al. (eds.), pages 163 (Wiley-Liss, Inc. 1996). Standard techniques for recovering protein produced by a bacterial system is provided by, for example, Grisshammer et al., “Purification of over-produced proteins from E. coli cells,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), pages 59-92 (Oxford University Press 1995). Established methods for isolating recombinant proteins from a baculovirus system are described by Richardson (ed.), Baculovirus Expression Protocols (The Humana Press, Inc., 1995).
  • More generally, TGF-beta binding-protein can be isolated by standard techniques, such as affinity chromatography, size exclusion chromatography, ion exchange chromatography, HPLC and the like. Additional variations in TGF-beta binding-protein isolation and purification can be devised by those of skill in the art. For example, anti-TGF-beta binding-protein antibodies, obtained as described below, can be used to isolate large quantities of protein by immunoaffinity purification. [0136]
  • 5. Production of Antibodies to TGF-Beta Binding-Proteins [0137]
  • The present invention provides antibodies that specifically bind to sclerostin as described herein in detail. Antibodies to TGF-beta binding-protein can be obtained, for example, using the product of an expression vector as an antigen. Antibodies that specifically bind to sclerostin may also be prepared by using peptides derived from any one of the sclerostin polypeptide sequences provided herein (SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, and 65). Particularly useful anti-TGF-beta binding-protein antibodies “bind specifically” with TGF-beta binding-protein of Sequence ID Nos. 2, 6, 8, 10, 12, 14, 16, 46, or 65 but not to other TGF-beta binding-proteins such as Dan, Cerberus, SCGF, or Gremlin. Antibodies of the present invention (including fragments and derivatives thereof) may be a polyclonal or, especially a monoclonal antibody. The antibody may belong to any immunoglobulin class, and may be for example an IgG, (including isotypes of IgG, which for human antibodies are known in the art as IgG[0138] 1, IgG2, IgG3, IgG4); IgE; IgM; or IgA antibody. An antibody may be obtained from fowl or mammals, preferably, for example, from a murine, rat, human or other primate antibody. When desired the antibody may be an internalising antibody.
  • Polyclonal antibodies to recombinant TGF-beta binding-protein can be prepared using methods well-known to those of skill in the art (see, for example, Green et al., “Production of Polyclonal Antisera,” in [0139] Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press 1992); Williams et al., “Expression of foreign proteins in E. coli using plasmid vectors and purification of specific polyclonal antibodies,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 15 (Oxford University Press 1995)). Although polyclonal antibodies are typically raised in animals such as rats, mice, rabbits, goats, or sheep, an anti-TGF-beta binding-protein antibody of the present invention may also be derived from a subhuman primate antibody. General techniques for raising diagnostically and therapeutically useful antibodies in baboons may be found, for example, in Goldenberg et al., international patent publication No. WO 91/11465 (1991), and in Losman et al., Int. J. Cancer 46:310, 1990.
  • The antibody should comprise at least a variable region domain. The variable region domain may be of any size or amino acid composition and will generally comprise at least one hypervariable amino acid sequence responsible for antigen binding embedded in a framework sequence. In general terms the variable (V) region domain may be any suitable arrangement of immunoglobulin heavy (V[0140] H) and/or light (VL) chain variable domains. Thus for example the V region domain may be monomeric and be a VH or VL domain where these are capable of independently binding antigen with acceptable affinity. Alternatively the V region domain may be dimeric and contain VH-VH, VH-VL, or VL-VL, dimers in which the VH and VL chains are non-covalently associated (abbreviated hereinafter as Fv). Where desired, however, the chains may be covalently coupled either directly, for example via a disulphide bond between the two variable domains, or through a linker, for example a peptide linker, to form a single chain domain (abbreviated hereinafter as scFv).
  • The variable region domain may be any naturally occuring variable domain or an engineered version thereof. By engineered version is meant a variable region domain that has been created using recombinant DNA engineering techniques. Such engineered versions include those created for example from natural antibody variable regions by insertions, deletions or changes in or to the amino acid sequences of the natural antibodies. Particular examples of this type include those engineered variable region domains containing at least one CDR and optionally one or more framework amino acids from one antibody and the remainder of the variable region domain from a second antibody. [0141]
  • The variable region domain may be covalently attached at a C-terminal amino acid to at least one other antibody domain or a fragment thereof. Thus, for example where a V[0142] H domain is present in the variable region domain this may be linked to an immunoglobulin C H1 domain or a fragment thereof. Similarly a VL domain may be linked to a CK domain or a fragment thereof. In this way for example the antibody may be a Fab fragment wherein the antigen binding domain contains associated VH and VL domains covalently linked at their C-termini to a CH1 and CK domain respectively. The CH1 domain may be extended with further amino acids, for example to provide a hinge region domain as found in a Fab′ fragment, or to provide further domains, such as antibody CH2 and CH3 domains.
  • Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR). CDR peptides (“minimal recognition units”) can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al., [0143] Methods: A Companion to Methods in Enzymology 2:106, 1991; Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies,” in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al. (eds.), page 166 (Cambridge University Press 1995); and Ward et al., “Genetic Manipulation and Expression of Antibodies,” in Monoclonal Antibodies: Principles and Applications, Birch et al., (eds.), page 137 (Wiley-Liss, Inc. 1995)).
  • Antibodies for use in the invention may in general be monoclonal (prepared by conventional immunisation and cell fusion procedures) or in the case of fragments, derived therefrom using any suitable standard chemical such as reduction or enzymatic cleavage and/or digestion techniques, for example by treatment with pepsin. More specifically, monoclonal anti-TGF-beta binding-protein antibodies can be generated utilizing a variety of techniques. Rodent monoclonal antibodies to specific antigens may be obtained by methods known to those skilled in the art (see, for example, Kohler et al., [0144] Nature 256:495, 1975; and Coligan et al. (eds.), Current Protocols in Immunology, 1:2.5.1-2.6.7 (John Wiley & Sons 1991) [“Coligan”]; Picksley et al., “Production of monoclonal antibodies against proteins expressed in E. coli,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 93 (Oxford University Press 1995)).
  • Briefly, monoclonal antibodies can be obtained by injecting mice with a composition comprising a TGF-beta binding-protein gene product, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B-lymphocytes, fusing the B-lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures. [0145]
  • In addition, an anti-TGF-beta binding-protein antibody of the present invention may be derived from a human monoclonal antibody. Human monoclonal antibodies are obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. The transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., [0146] Nature Genet. 7:13, 1994; Lonberg et al., Nature 368:856, 1994; and Taylor et al., Int. Immun. 6:579, 1994.
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al., “Purification of Immunoglobulin G (IgG),” in [0147] Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)).
  • For particular uses, it may be desirable to prepare fragments of anti-TGF-beta binding-protein antibodies. Such antibody fragments can be obtained, for example, by proteolytic hydrolysis of the antibody. Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods. As an illustration, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)[0148] 2. This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab′ monovalent fragments. Optionally, the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages. As an alternative, an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an Fc fragment directly. These methods are described, for example, by Goldenberg, U.S. Pat. No. 4,331,647, Nisonoff et al.,Arch Biochem. Biophys. 89:230, 1960, Porter, Biochem. J. 73:119, 1959, Edelman et al., in Methods in Enzymology 1:422 (Academic Press 1967), and by Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4.
  • Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody. [0149]
  • Alternatively, the antibody may be a recombinant or engineered antibody obtained by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions. Such DNA is known and/or is readily available from DNA libraries including for example phage-antibody libraries (see Chiswell, D J and McCafferty, J. Tibtech. 10 80-84 (1992)) or where desired can be synthesised. Standard molecular biology and/or chemistry procedures may be used to sequence and manipulate the DNA, for example, to introduce codons to create cysteine residues, to modify, add or delete other amino acids or domains as desired. [0150]
  • One or more replicable expression vectors containing the DNA encoding a variable and/or constant region may be prepared and used to transform an appropriate cell line, e.g. a non-producing myeloma cell line, such as a mouse NSO line or a bacterial, such as [0151] E.coli, in which production of the antibody will occur. In order to obtain efficient transcription and translation, the DNA sequence in each vector should include appropriate regulatory sequences, particularly a promoter and leader sequence operably linked to a variable domain sequence. Particular methods for producing antibodies in this way are generally well known and routinely used. For example, basic molecular biology procedures are described by Maniatis et al (Molecular Cloning, Cold Spring Harbor Laboratory, New York, 1989); DNA sequencing can be performed as described in Sanger et al (Proc. Natl. Acad. Sci. USA 74: 5463, (1977)) and the Amersham International plc sequencing handbook; site directed mutagenesis can be carried out according to the method of Kramer et al. (Nucleic Acids Res. 12, 9441, (1984)); the Anglian Biotechnology Ltd handbook; Kunkel Proc. Natl. Acad. Sci. USA 82:488-92 (1985); Kunkel et al., Methods in Enzymol. 154:367-82 (1987). Additionally, numerous publications detail techniques suitable for the preparation of antibodies by manipulation of DNA, creation of expression vectors, and transformation of appropriate cells, for example as reviewed by Mountain A and Adair, J R in Biotechnology and Genetic Engineering Reviews (ed. Tombs, M P, 10, Chapter 1, 1992, Intercept, Andover, UK) and in International Patent Specification No. WO 91/09967.
  • In certain embodiments, the-antibody according to the invention may have one or more effector or reporter molecules attached to it and the invention extends to such modified proteins. A reporter molecule may be a detectable moiety or label such as an enzyme, cytotoxic agent or other reporter molecule, including a dye, radionuclide, luminescent group, fluorescent group, or biotin, or the like. The TGF-beta binding protein-specific immunoglobulin or fragment thereof may be radiolabeled for diagnostic or therapeutic applications. Techniques for radiolabeling of antibodies are known in the art. See, e.g., Adams 1998 In Vivo 12:11-21; Hiltunen 1993 [0152] Acta Oncol. 32:831-9. Therapeutic applications are described in greater detail below and may include use of the TGF-beta binding protein specific antibody (or fragment thereof) in conjunction with other therapeutic agents. The effector or reporter molecules may be attached to the antibody through any available amino acid side-chain, terminal amino acid or, where present carbohydrate functional group located in the antibody, provided that the attachment or the attachment process does not adversely affect the binding properties and the usefulness of the molecule. Particular functional groups include, for example any free amino, imino, thiol, hydroxyl, carboxyl or aldehyde group. Attachment of the antibody and the effector and/or reporter molecule(s) may be achieved via such groups and an appropriate functional group in the effector or reporter molecules. The linkage may be direct or indirect through spacing or bridging groups.
  • Effector molecules include, for example, antineoplastic agents, toxins (such as enzymatically active toxins of bacterial (such as [0153] P. aeruginosa exotoxin A) or plant origin and fragments thereof (e.g. ricin and fragments thereof; plant gelonin, bryodin from Bryonia dioica, or the like. See, e.g., Thrush et al., 1996 Annu. Rev. Immunol. 14:49-71; Frankel et al., 1996 Cancer Res. 56:926-32); biologically active proteins, for example enzymes; nucleic acids and fragments thereof such as. DNA, RNA and fragments thereof; naturally occurring and synthetic polymers (e.g., polysaccharides and polyalkylene polymers such as poly(ethylene glycol) and derivatives thereof); radionuclides, particularly radioiodide; and chelated metals. Suitable reporter groups include chelated metals, fluorescent compounds, or compounds that may be detected by NMR or ESR spectroscopy. Particularly useful effector groups are calichaemicin and derivatives thereof (see, for example, South African Patent Specifications Nos. 85/8794, 88/8127 and 90/2839).
  • Numerous other toxins, including chemotherapeutic agents, anti-mitotic agents, antibiotics, inducers of apoptosis (or “apoptogens”, see, e.g., Green and Reed, 1998, [0154] Science 281:1309-1312), or the like, are known to those familiar with the art, and the examples provided herein are intended to be illustrative without limiting the scope and spirit of the invention. Particular antineoplastic agents include cytotoxic and cytostatic agents, for example alkylating agents, such as nitrogen mustards (e.g., chlorambucil, melphalan, mechlorethamine, cyclophosphamide, or uracil mustard) and derivatives thereof, triethylenephosphoramide, triethylenethiophosphor-amide, busulphan, or cisplatin; antimetabolites, such as methotrexate, fluorouracil, floxuridine, cytarabine, mercaptopurine, thioguanine, fluoroacetic acid or fluorocitric acid, antibiotics, such as bleomycins (e.g., bleomycin sulphate), doxorubicin, daunorubicin, mitomycins (e.g., mitomycin C), actinomycins (e.g., dactinomycin) plicamycin, calichaemicin and derivatives thereof, or esperamicin and derivatives thereof; mitotic inhibitors, such as etoposide, vincristine or vinblastine and derivatives thereof, alkaloids, such as ellipticine; polyols such as taxicin-I or taxicin-II; hormones, such as androgens (e.g., dromostanolone or testolactone), progestins (e.g., megestrol acetate or medroxyprogesterone acetate), estrogens (e.g., dimethylstilbestrol diphosphate, polyestradiol phosphate or estramustine phosphate) or antiestrogens (e.g., tamoxifen); anthraquinones, such as mitoxantrone, ureas, such as hydroxyurea; hydrazines, such as procarbazine; or imidazoles, such as dacarbazine.
  • Chelated metals include chelates of di-or tripositive metals having a coordination number from 2 to 8 inclusive. Particular examples of such metals include technetium (Tc), rhenium (Re), cobalt (Co), copper (Cu), gold (Au), silver (Ag), lead (Pb), bismuth (Bi), indium (In), gallium (Ga), yttrium (Y), terbium (Tb), gadolinium (Gd), and scandium (Sc). In general the metal is preferably a radionuclide. Particular radionuclides include [0155] 99mTc, 186Re, 188Re, 58Co, 60Co, 67Cu, 195Au, 199Au, 110Ag, 203Pb, 206Bi, 207Bi, 111In, 67Ga, 68Ga, 88Y, 90Y, 160Tb, 153Gd, and 47Sc.
  • The chelated metal may be for example one of the above types of metal chelated with any suitable polydentate chelating agent, for example acyclic or cyclic polyamines, polyethers, (e.g., crown ethers and derivatives thereof); polyamides; porphyrins; and carbocyclic derivatives. In general, the type of chelating agent will depend on the metal in use. One particularly useful group of chelating agents in conjugates according to the invention, however, comprises acyclic and cyclic polyamines, especially. polyaminocarboxylic acids, for example diethylenetriaminepentaacetic acid and derivatives thereof, and macrocyclic amines, such as cyclic tri-aza and tetra-aza derivatives (for example, as described in International Patent Specification No. WO 92/22583), and polyamides, especially desferrioxamine and derivatives thereof. [0156]
  • When a thiol group in the antibody is used as the point of attachment this may be achieved through reaction with a thiol reactive group present in the effector or reporter molecule. Examples of such groups include an á-halocarboxylic acid or ester, such as iodoacetamide, an imide, such as maleimide, a vinyl sulphone, or a disulphide. These and other suitable linking procedures are generally and more particularly described in International Patent Specifications Nos. WO 93/06231, WO 92/22583, WO 90/091195, and WO 89/01476. [0157]
  • Assays for Selecting Molecules that Increase Bone Density [0158]
  • As discussed above, the present invention provides methods for selecting and/or isolating compounds that are capable of increasing bone density. For example, within one aspect of the present invention methods are provided for determining whether a selected molecule (e.g., a candidate agent) is capable of increasing bone mineral content, comprising the steps of (a) mixing (or contacting) a selected molecule with TGF-beta binding protein and a selected member of the TGF-beta family of proteins, (b) determining whether the selected molecule stimulates signaling by the TGF-beta family of proteins, or inhibits the binding of the TGF-beta binding protein to at least one member of the TGF-beta family of proteins. Within certain embodiments, the molecule enhances the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation. [0159]
  • Within other aspects of the invention, methods are provided for determining whether a selected molecule (candidate agent) is capable of increasing bone mineral content, comprising the steps of (a) exposing (contacting, mixing, combining) a selected molecule to cells which express TGF-beta binding-protein and (b) determining whether the expression (or activity) of TGF-beta binding-protein in the exposed cells decreases, or whether an activity of the TGF-beta binding protein decreases, and therefrom determining whether the compound is capable of increasing bone mineral content. Within one embodiment, the cells are selected from the group consisting of the spontaneously transformed or untransformed normal human bone from bone biopsies and rat parietal bone osteoblasts. Methods for detecting the level of expression of a TGF-beta binding protein may be accomplished in a wide variety of assay formats known in the art and described herein. Immunoassays may be used for detecting and quantifying the expression of a TGF-beta binding protein and include, for example, Countercurrent Immuno-Electrophoresis (CIEP), radioimmunoassays, radioimmunoprecipitations, Enzyme-Linked Immuno-Sorbent Assays (ELISA), immunoblot assays such as dot blot assays and Western blots, inhibition or competition assays, and sandwich assays (see U.S. Pat. Nos. 4,376,110 and 4,486,530; see also [0160] Antibodies: A Laboratory Manual, supra). Such immunoassays may use an antibody that is specific for a TGF-beta binding protein such as the anti-sclerostin antibodies described herein, or may use an antibody that is specific for a reporter molecule that is attached to the TGF-beta binding protein. The level of polypeptide expression may also be determined by quantifying the amount of TGF-beta binding protein that binds to a TGF-beta binding protein ligand. By way of example, binding of sclerostin in a sample to a BMP may be detected by surface plasmon resonance (SPR). Alternatively, the level of expression of mRNA encoding the specific TGF-beta binding protein may be quantified.
  • Representative embodiments of such assays are provided below in Examples 5 and 6. Briefly, a family member of the TGF-beta super-family or a TGF-beta binding protein is first bound to a solid phase, followed by addition of a candidate molecule. A labeled family member of the TGF-beta super-family or a TGF-beta binding protein is then added to the assay (i.e., the labeled polypeptide is the ligand for whichever polypeptide was bound to the solid phase), the solid phase washed, and the quantity of bound or labeled TGF-beta super-family member or TGF-beta binding protein on the solid support determined. Molecules which are suitable for use in increasing bone mineral content as described herein are those molecules which decrease the binding of TGF-beta binding protein to a member or members of the TGF-beta super-family in a statistically significant manner. Obviously, assays suitable for use within the present invention should not be limited to the embodiments described within Examples 2 and 3. In particular, numerous parameters may be altered, such as by binding TGF-beta to a solid phase, or by elimination of a solid phase entirely. [0161]
  • Within other aspects of the invention, methods are provided for determining whether a selected molecule is capable of increasing bone mineral content, comprising the steps of (a) exposing (contacting, mixing, combining) a selected molecule (candidate agent) to cells which express TGF-beta and (b) determining whether the activity of TGF-beta from said exposed cells is altered, and therefrom determining whether the compound is capable of increasing bone mineral content. Similar to the methods described herein, a wide variety of methods may be utilized to assess the changes of TGF-beta binding-protein expression due to a selected test compound. In one embodiment of the invention, the candidate agent is an antibody that binds to the TGF-beta binding protein sclerostin disclosed herein. [0162]
  • In a preferred embodiment of the invention, a method is provided for identifying an antibody that modulates a TGF-beta signaling pathway comprising contacting an antibody that specifically binds to a SOST polypeptide with a SOST peptide, including but not limited to the peptides disclosed herein, under conditions and for a time sufficient to permit formation of an antibody plus (+) SOST (antibody/SOST) complex and then detecting the level (e.g., quantifying the amount) of the SOST/antibody complex to determine the presence of an antibody that modulates a TGF-beta signaling pathway. The method may be performed using SPR or any number of different immunoassays known in the art and disclosed herein, including an ELISA, immunoblot, or the like. A TGF-beta signaling pathway includes a signaling pathway by which a BMP binds to a type I and a type II receptor on a cell to stimulate or induce the pathway that modulates bone mineral content. In certain preferred embodiments of the invention, an antibody that specifically binds to SOST stimulates or enhances the pathway for increasing bone mineral content. Such an antibody may be identified using the methods disclosed herein to detect binding of an antibody to SOST specific peptides. [0163]
  • The subject invention methods may also be used for identifying antibodies that impair, inhibit (including competitively inhibit), or prevent binding of a BMP to a SOST polypeptide by detecting whether an antibody binds to SOST peptides that are located in regions or portions of regions on SOST to which a BMP binds, such as peptides at the amino terminal end of SOST and peptides that include amino terminal amino acid residues and a portion of the core region (docking core) of SOST (e.g., SEQ ID NOS: 47-64, 66-73, and 92-95). The methods of the present invention may also be used to identify an antibody that impairs, prevents, or inhibits, formation of SOST homodimers. Such an antibody that binds specifically to SOST may be identified by detecting binding of the antibody to peptides that are derived from the core or the carboxy terminal region of SOST (e.g., SEQ ID NOS: 74-91 and 96-99). [0164]
  • Within another embodiment of the present invention, methods are provided for determining whether a selected molecule is capable of increasing bone mineral content, comprising the steps of (a) mixing or contacting a selected molecule (candidate agent) with a TGF-beta-binding-protein and a selected member of the TGF-beta family of proteins, (b) determining whether the selected molecule up-regulates the signaling of the TGF-beta family of proteins, or inhibits the binding of the TGF-beta binding-protein to the TGF-beta family of proteins. Within certain embodiments, the molecule enhances the ability of TGF-beta to function as a positive regulator of mesenchymal cell differentiation. [0165]
  • Similar to the above described methods, a wide variety of methods may be utilized to assess stimulation of TGF-beta due to a selected test compound. One such representative method is provided below in Example 6 (see also Durham et al., [0166] Endo. 136: 1374-1380.
  • Within yet other aspects of the present invention, methods are provided for determining whether a selected molecule (candidate agent) is capable of increasing bone mineral content, comprising the step of determining whether a selected molecule inhibits the binding of TGF-beta binding-protein to bone, or an analogue thereof. As utilized herein, it should be understood that bone or analogues thereof refers to hydroxyapatite, or a surface composed of a powdered form of bone, crushed bone or intact bone. Similar to the above described methods, a wide variety of methods may be utilized to assess the inhibition of TGF-beta binding-protein localization to bone matrix. One such representative method is provided below in Example 7 (see also Nicolas et al., [0167] Calcif. Tissue Int. 47:206-12 (1995)).
  • In one embodiment of the invention, an antibody or antigen-binding fragment thereof that specifically binds to a sclerostin polypeptide is capable of competitively inhibiting binding of a TGF-beta family member to the sclerostin polypeptide. The capability of the antibody or antibody fragment to impair or blocking binding of a TGF-beta family member, such as a BMP, to sclerostin may be determined according to any of the methods described herein. The antibody or fragment thereof that specifically binds to sclerostin may impair, block, or prevent binding of a TGF-beta family member to sclerostin by impairing sclerostin homodimer formation. An antibody that specifically binds to sclerostin may also be used to identify an activity of sclerostin by inhibiting or impairing sclerostin from binding to a BMP. Alternatively, the antibody or fragment thereof may be incorporated in a cell-based assay or in an animal model in which sclerostin has a defined activity to determine whether the antibody alters (increases or decreases in a statistically significant manner) that activity. An antibody or fragment thereof that specifically binds to sclerostin may be used to examine the effect of such an antibody in a signal transduction pathway and thereby modulate (stimulate or inhibit) the signaling pathway. Preferably, binding of an antibody to SOST results in a stimulation or induction of a signaling pathway. [0168]
  • While the methods recited herein may refer to the analysis of an individual test molecule, that the present invention should not be so limited. In particular, the selected molecule may be contained within a mixture of compounds. Hence, the recited methods may further comprise the step of isolating a molecule that inhibits the binding of TGF-beta binding-protein to a TGF-beta family member. [0169]
  • Candidate Molecules [0170]
  • A wide variety of molecules may be assayed for their ability to inhibit the binding of TGF-beta binding-protein to a TGF-beta family member. Representative examples discussed in more detail below include organic molecules (e.g., organic small molecules), proteins or peptides, and nucleic acid molecules. Although it should be evident from the discussion below that the candidate molecules described herein may be utilized in the assays described herein, it should also be readily apparent that such molecules can also be utilized in a variety of diagnostic and therapeutic settins. [0171]
  • 1. Organic Molecules [0172]
  • Numerous organic small molecules may be assayed for their ability to inhibit the binding of TGF-beta binding-protein to a TGF-beta family member. For example, within one embodiment of the invention suitable organic molecules may be selected from either a chemical library, wherein chemicals are assayed individually, or from combinatorial chemical libraries where multiple compounds are assayed at once, then deconvoluted to determine and isolate the most active compounds. [0173]
  • Representative examples of such combinatorial chemical libraries include those described by Agrafiotis et al., “System and method of automatically generating chemical compounds with desired properties,” U.S. Pat. No. 5,463,564; Armstrong, R. W., “Synthesis of combinatorial arrays of organic compounds through the use of multiple component combinatorial array syntheses,” WO 95/02566; Baldwin, J. J. et al., “Sulfonamide derivatives and their use,” WO 95/24186; Baldwin, J. J. et al., “Combinatorial dihydrobenzopyran library,” WO 95/30642; Brenner, S., “New kit for preparing combinatorial libraries,” WO 95/16918; Chenera, B. et al., “Preparation of library of resin-bound aromatic carbocyclic compounds,” WO 95/16712; Ellman, J. A., “Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support,” U.S. Pat. No. 5,288,514; Felder, E. et al., “Novel combinatorial compound libraries,” WO 95/16209; Lerner, R. et al., “Encoded combinatorial chemical libraries,” WO 93/20242; Pavia, M. R. et al., “A method for preparing and selecting pharmaceutically useful non-peptide compounds from a structurally diverse universal library,” WO 95/04277; Summerton, J. E. and D. D. Weller, “Morpholino-subunit combinatorial library and method,” U.S. Pat. No. 5,506,337; Holmes, C., “Methods for the Solid Phase Synthesis of Thiazolidinones, Metathiazanones, and Derivatives thereof,” WO 96/00148; Phillips, G. B. and G. P. Wei, “Solid-phase Synthesis of Benzimidazoles,” [0174] Tet. Letters 37:4887-90, 1996; Ruhland, B. et al., “Solid-supported Combinatorial Synthesis of Structurally Diverse β-Lactams,” J. Amer. Chem. Soc. 111:253-4, 1996; Look, G. C. et al., “The Indentification of Cyclooxygenase-1 Inhibitors from 4-Thiazolidinone Combinatorial Libraries,” Bioorg and Med. Chem. Letters 6:707-12, 1996.
  • 2. Proteins and Peptides [0175]
  • A wide range of proteins and peptides may likewise be utilized as candidate molecules for inhibitors of the binding of TGF-beta binding-protein to a TGF-beta family member. [0176]
  • a. Combinatorial Peptide Libraries [0177]
  • Peptide molecules which are putative inhibitors of the binding of TGF-beta binding-protein to a TGF-beta family member may be obtained through the screening of combinatorial peptide libraries. Such libraries may either be prepared by one of skill in the art (see e.g., U.S. Pat. Nos. 4,528,266 and 4,359,535, and Patent Cooperation Treaty Publication Nos. WO 92/15679, WO 92/15677, WO 90/07862, WO 90/02809, or purchased from commercially available sources (e.g., New England Biolabs Ph.D.™ Phage Display Peptide Library Kit). [0178]
  • b. Antibodies [0179]
  • The present invention provides antibodies that specifically bind to a sclerostin polypeptide methods for using such antibodies. The present invention also provides sclerostin polypeptide immunogens that may be used for generation and analysis of these antibodies. The antibodies may be useful to block or impair binding of a sclerostin polypeptide, which is a TGF-beta binding protein, to a ligand, particularly a bone morphogenic protein, and may also block or impair binding of the sclerostin polypeptide to one or more other ligands. [0180]
  • A molecule such as an antibody that inhibits the binding of the TGF-beta binding protein to one or more members of the TGF-beta family of proteins, including one or more bone morphogenic proteins (BMPs), should be understood to refer to, for example, a molecule that allows the activation of a TGF-beta family member or BMP, or allows binding of TGF-beta family members including one or more BMPs to their respective receptors by removing or preventing the TGF-beta member from binding to the TGF-binding-protein. [0181]
  • The present invention also provides peptide and polypeptide immunogens that may be used to generate and/or identify antibodies or fragments thereof that are capable of inhibiting, preventing, or impairing binding of the TGF-beta binding protein sclerostin to one or more BMPs. The present invention also provides peptide and polypeptide immunogens that may be used to generate and/or identify antibodies or fragments thereof that are capable of inhibiting, preventing, or impairing (e.g., decreasing in a statistically significant manner) the formation of sclerostin homodimers. The antibodies of the present invention are useful for increasing the mineral content and mineral density of bone, thereby ameliorating numerous conditions that result in the loss of bone mineral content, including for example, disease, genetic predisposition, accidents that result in the lack of use of bone (e.g., due to fracture), therapeutics that effect bone resorption or that kill bone forming cells, and normal aging. [0182]
  • Polypeptides or peptides useful for immunization and/or analysis of sclerostin-specific antibodies may also be selected by analyzing the primary, secondary, and tertiary structure of a TGF-beta binding protein according to methods known to those skilled in the art and described herein, in order to determine amino acid sequences more likely to generate an antigenic response in a host animal. See, e.g., Novotny, [0183] Mol. Immunol. 28:201-207 (1991); Berzofsky, Science 229:932-40 (1985)). Modeling and x-ray crystallography data may also be used to predict and/or identify which portions or regions of a TGF-beta binding protein interact with which portions of a TGF-beta binding protein ligand, such as a BMP. TGF-beta binding protein peptide immunogens may be designed and prepared that include amino acid sequences within or surrounding the portions or regions of interaction. These antibodies may be useful to block or impair binding of the TGF-beta binding protein to the same ligand and may also block or impair binding of the TGF-beta binding protein to one or more other ligands.
  • Antibodies or antigen binding fragments thereof contemplated by the present invention include antibodies that are capable of specifically binding to sclerostin and competitively inhibiting binding of a TGF-beta polypeptide, such as a BMP, to sclerostin. For example, the antibodies contemplated by the present invention competitively inhibit binding of the sclerostin polypeptide to the BMP Type I receptor site on a BMP, or to the BMP Type II receptor binding site, or may competitively inhibit binding of sclerostin to both the Type I and Type II receptor binding sites on a BMP. Without wishing to be bound by theory, when an anti-sclerostin antibody competitively inhibits binding of the Type I and/or Type II binding sites of the BMP polypeptide to sclerostin, thus blocking the antagonistic activity of sclerostin, the receptor binding sites on BMP are available to bind to the Type I and Type II receptors, thereby increasing bone mineralization. The binding interaction between a TGF-beta binding protein such as sclerostin and a TGF-beta polypeptide such as a BMP generally occurs when each of the ligand pairs forms a homodimer. Therefore instead of or in addition to using an antibody specific for sclerostin to block, impair, or prevent binding of sclerostin to a BMP by competitively inhibiting binding of sclerostin to BMP, a sclerostin specific antibody may be used to block or impair sclerostin homodimer formation. [0184]
  • By way of example, one dimer of human Noggin, which is a BMP antagonist that has the ability to bind a BMP with high affinity (Zimmerman et al., supra), was isolated in complex with one dimer of human BMP-7 and analyzed by multiwavelength anomalous diffraction (MAD) (Groppe et al., [0185] Nature 420:636-42 (2002)). As discussed herein, this study revealed that Noggin dimer may efficiently block all the receptor binding sites (two type I and two type II receptor binding sites) on a BMP dimer. The location of the amino acids of Noggin that contact BMP-7 may be useful in modeling the interaction between other TGF-beta binding proteins, such as sclerostin (SOST), and BMPs, and thus aiding the design of peptides that may be used as immunogens to generate antibodies that block or impair such an interaction.
  • In one embodiment of the present invention, an antibody, or an antigen-binding fragment thereof, that binds specifically to a SOST polypeptide competitively inhibits binding of the SOST polypeptide to at least one or both of a bone morphogenic protein (BMP) Type I Receptor binding site and a BMP Type II Receptor binding site that are located on a BMP. The epitopes on SOST to which these antibodies bind may include or be included within contiguous amino acid sequences that are located at the N-terminus of the SOST polypeptide (amino acids at about positions 1-56 of SEQ ID NO: 46). The polypeptides may also include a short linker peptide sequence that connects the N-terminal region to the core region, for example, polypeptides as provided in SEQ ID NO: 92 (human) and SEQ ID NO: 93 (rat). Shorter representative N-terminus peptide sequences of human SOST (e.g., SEQ ID NO: 46) include SEQ ID NOS: 47-51, and representative rat SOST (e.g., SEQ ID NO: 65) peptide sequences include SEQ ID NOS: 57-60. [0186]
  • Antibodies that specifically bind to a SOST polypeptide and block or competitively inhibit binding of the SOST polypeptide to a BMP, for example, by blocking or inhibiting binding to amino acids of a BMP corresponding to one or more of the Type I and Type II receptor binding sites may also specifically bind to peptides that comprise an amino acid sequence corresponding to the core region of SOST (amino acids at about positions 57-146 of SEQ ID NO: 46). Polypeptides that include the core region may also include additional amino acids extending at either or both the N-terminus and C-terminus, for example, to include cysteine residues that may be useful for conjugating the polypeptide to a carrier molecule. Representative core polypeptides of human and rat SOST, for example, comprise the amino acid sequences set forth in SEQ ID NO: 94 and SEQ ID NO: 95, respectively. Such antibodies may also bind shorter polypeptide sequences. Representative human SOST core peptide sequences are provided in SEQ ID NOS: 66-69 and representative rat SOST core sequences are provided in SEQ ID NOS: 70-73. [0187]
  • In another embodiment, antibodies that specifically bind to a SOST polypeptide impair (inhibit, prevent, or block, e.g., decrease in a statistically significant manner) formation of a SOST homodimer. Because the interaction between SOST and a BMP may involve a homodimer of SOST and a homodimer of the BMP, an antibody that prevents or impairs homodimer formation of SOST may thereby alter bone mineral density, preferably increasing bone mineral density. In one embodiment, antibodies that bind to the core region of SOST prevent homodimer formation. Such antibodies may also bind to peptides that comprise contiguous amino acid sequences corresponding the core region, for example, SEQ ID NOS: 74, 75, and 98 (human SOST) and SEQ ID NOS: 76 and 99 (rat SOST). Antibodies that bind to an epitope located on the C-terminal region of a SOST polypeptide (at about amino acid positions 147-190 of either SEQ ID NOS: 46 or 65) may also impair homodimer formation. Representative C-terminal polypeptides of human and rat SOST, for example, comprise the amino acid sequences set forth in SEQ ID NO: 96 and SEQ ID NO: 97, respectively. Such antibodies may also bind shorter polypeptide sequences. Representative human SOST C-terminal peptide sequences are provided in SEQ ID NOS: 78-81 and representative rat SOST C-terminal sequences are provided in SEQ ID NOS: 86-88. [0188]
  • The SOST polypeptides and peptides disclosed herein to which antibodies may specifically bind are useful as immunogens. These immunogens of the present invention may be used for immunizing an animal to generate a humoral immune response that results in production of antibodies that specifically bind to a Type I or Type II receptor binding site or both located on a BMP include peptides derived from the N-terminal region of SOST or that may prevent SOST homodimer formation. [0189]
  • Such SOST polypeptides and peptides that are useful as immunogens may also be used in methods for screening samples containing antibodies, for example, samples of purified antibodies, antisera, or cell culture supernatants or any other biological sample that may contain one or more antibodies specific for SOST. These peptides may also be used in methods for identifying and selecting from a biological sample one or more B cells that are producing an antibody that specifically binds to SOST (e.g., plaque forming assays and the like). The B cells may then be used as source of a SOST specific antibody-encoding polynucleotide that can be cloned and/or modified by recombinant molecular biology techniques known in the art and described herein. [0190]
  • A “biological sample” as used herein refers in certain embodiments to a sample containing at least one antibody specific for a SOST polypeptide, and a biological sample may be provided by obtaining a blood sample, biopsy specimen, tissue explant, organ culture, or any other tissue or cell preparation from a subject or a biological source. A sample may further refer to a tissue or cell preparation in which the morphological integrity or physical state has been disrupted, for example, by dissection, dissociation, solubilization, fractionation, homogenization, biochemical or chemical extraction, pulverization, lyophilization, sonication, or any other means for processing a sample derived from a subject or biological source. The subject or biological source may be a human or non-human animal, a primary cell culture (e.g., B cells immunized in vitro), or culture adapted cell line including but not limited to genetically engineered cell lines that may contain chromosomally integrated or episomal recombinant nucleic acid sequences, immortalized or immortalizable cell lines, somatic cell hybrid cell lines, differentiated or differentiatable cell lines, transformed cell lines, and the like. [0191]
  • SOST peptide immunogens may also be prepared by synthesizing a series of peptides that, in total, represent the entire polypeptide sequence of a SOST polypeptide and that each have a portion of the SOST amino acid sequence in common with another peptide in the series. This overlapping portion would preferably be at least four amino acids, and more preferably 5, 6, 7, 8, 9, or 10 amino acids. Each peptide may be used to immunize an animal, the sera collected from the animal, and tested in an assay to identify which animal is producing antibodies that impair or block binding of SOST to a TGF-beta protein. Antibodies are then prepared from such identified immunized animals according to methods known in the art and described herein. [0192]
  • Antibodies which inhibit the binding of TGF-beta binding-protein to a TGF-beta family member may readily be prepared given the disclosure provided herein. Particularly useful are anti-TGF-beta binding-protein antibodies that “specifically bind” TGF-beta binding-protein of SEQ ID NOS: 2, 6, 8, 10, 12, 14, 16, 46, or 65, but not to other TGF-beta binding-proteins such as Dan, Cerberus, SCGF, or Gremlin. Within the context of the present invention, antibodies are understood to include monoclonal antibodies, polyclonal antibodies, single chain, chimeric, CDR-grafted immunoglobulings, anti-idiotypic antibodies, and antibody fragments thereof (e.g., Fab, Fd, Fab′, and F(ab′)[0193] 2, FV variable regions, or complementarity determining regions). As discussed above, antibodies are understood to be specific against TGF-beta binding-protein, or against a specific TGF-beta family member, if they bind with a Ka of greater than or equal to 107 M−1, preferably greater than or equal to 108 M−1, and do not bind to other TGF-beta binding-proteins, or bind with a Ka of less than or equal to 106 M−1. Affinity of an antibody for its cognate antigen is also commonly expressed as a dissociation constant KD, and an anti-SOST antibody specifically binds to a TGF-beta family member if it binds with a KD of less than or equal to about 10−5 M, more preferably less than or equal to about 10−6 M, still more preferably less than or equal to 10−7 M, and still more preferably less than or equal to 10−8 M. Furthermore, antibodies of the present invention preferably block, impair, or inhibit (e.g., decrease with statistical significance) the binding of TGF-beta binding-protein to a TGF-beta family member. The affinity of a monoclonal antibody or binding partner, as well as inhibition of binding can be readily determined by one of ordinary skill in the art (see Scatchard, Ann. N.Y. Acad. Sci. 51:660-672, 1949). Affinity may also be determined by surface plasmon resonance (SPR; BIAcore, Biosensor, Piscataway, N.J.). For surface plasmon resonance, target molecules are immobilized on a solid phase and exposed to ligands in a mobile phase running along a flow cell. If ligand binding to the immobilized target occurs, the local refractive index changes, leading to a change in SPR angle, which can be monitored in real time by detecting changes in the intensity of the reflected light. The rates of change of the SPR signal can be analyzed to yield apparent rate constants for the association and dissociation phases of the binding reaction. The ratio of these values gives the apparent equilibrium constant (affinity) (see, e.g., Wolff et al., Cancer Res. 53:2560-65 (1993)).
  • An antibody according to the present invention may belong to any immunoglobulin class, for example IgG, IgE, IgM, IgD, or IgA, and may be any one of the different isotypes that may comprise a class (such as IgG1, IgG2, IgG3, and IgG4 of the human IgG class). It may be obtained from or derived from an animal, for example, fowl (e.g., chicken) and mammals, which includes but is not limited to a mouse, rat, hamster, rabbit, or other rodent, a cow, horse, sheep, goat, camel, human, or other primate. The antibody may be an internalising antibody. [0194]
  • Methods well known in the art may be used to generate antibodies, polyclonal antisera, or monoclonal antibodies that are specific for a TGF-beta binding protein such as SOST. Antibodies also may be produced as genetically engineered immunoglobulins (Ig) or Ig fragments designed to have desirable properties. For example, by way of illustration and not limitation, antibodies may include a recombinant IgG that is a chimeric fusion protein having at least one variable (V) region domain from a first mammalian species and at least one constant region domain from a second, distinct mammalian species. Most commonly, a chimeric antibody has murine variable region sequences and human constant region sequences. Such a murine/human chimeric immunoglobulin may be “humanized” by grafting the complementarity determining regions (CDRs) derived from a murine antibody, which confer binding specificity for an antigen, into human-derived V region framework regions and human-derived constant regions. Fragments of these molecules may be generated by proteolytic digestion, or optionally, by proteolytic digestion followed by mild reduction of disulfide bonds and alkylation. Alternatively, such fragments may also be generated by recombinant genetic engineering techniques. [0195]
  • Certain preferred antibodies are those antibodies that inhibit or block a TGF-beta binding protein activity within an in vitro assay, as described herein. Binding properties of an antibody to a TGF-beta binding protein may generally be assessed using immunodetection methods including, for example, an enzyme-linked immunosorbent assay (ELISA), immunoprecipitation, immunoblotting, countercurrent immunoelectrophoresis, radioimmunoassays, dot blot assays, inhibition or competition assays, and the like, which may be readily performed by those having ordinary skill in the art (see, e.g., U.S. Pat. Nos. 4,376,110 and 4,486,530; Harlow et al., [0196] Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory (1988)).
  • An immunogen may be comprised of cells expressing a TGF-beta binding protein, purified or partially purified TGF-beta binding polypeptides, or variants or fragments (i.e., peptides) thereof, or peptides derived from a TGF-beta binding protein. Such peptides may be generated by proteolytic cleavage of a larger polypeptide, by recombinant molecular methodologies, or may be chemically synthesized. For instance, nucleic acid sequences encoding TGF-beta binding proteins are provided herein, such that those skilled in the art may routinely prepare TGF-beta binding proteins for use as immunogens. Peptides may be chemically synthesized by methods as described herein and known in the art. Alternatively, peptides may be generated by proteolytic cleavage of a TGF-beta binding protein, and individual peptides isolated by methods known in the art such as polyacrylamide gel electrophoresis or any number of liquid chromatography or other separation methods. Peptides useful as immunogens typically may have an amino acid sequence of at least 4 or 5 consecutive amino acids from a TGF-beta binding protein amino acid sequence such as those described herein, and preferably have at least 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 18, 19 or 20 consecutive amino acids of a TGF-beta binding protein. Certain other preferred peptide immunogens comprise at least 6 but no more than 12 or more consecutive amino acids of a TGF-beta binding protein sequence, and other preferred peptide immunogens comprise at least 21 but no more than 50 consecutive amino acids of a SOST polypeptide. Other preferred peptide immunogens comprise 21-25, 26-30, 31-35, 36-40, 41-50, or any whole integer number of amino acids between and including 21 and 100 consecutive amino acids, and between 100 and 190 consecutive amino acids of a TGF-beta binding protein sequence. [0197]
  • As disclosed herein, polyclonal antibodies may be readily generated by one of ordinary skill in the art from a variety of warm-blooded animals such as horses, cows, various fowl, rabbits, mice, sheep, goats, baboons, or rats. Typically, the TGF-beta binding-protein or unique peptide thereof of 13-20 amino acids or as described herein (preferably conjugated to keyhole limpet hemocyanin by cross-linking with glutaraldehyde) is used to immunize the animal through intraperitoneal, intramuscular, intraocular, intradermal, or subcutaneous injections, along with an adjuvant such as Freund's complete or incomplete adjuvant, or the Ribi Adjuvant System (Corixa Corporation, Seattle, Was.). See also, e.g., Harlow et al., supra. In general, after the first injection, animals receive one or more booster immunizations according to a preferred schedule that may vary according to, inter alia, the antigen, the adjuvant (if any), and/or the particular animal species. The immune response may be monitored by periodically bleeding the animal and preparing and analyzing sera in an immunoassay, such as an ELISA or Ouchterlony diffusion assay, or the like, to determine the specific antibody titer. Particularly preferred polyclonal antisera will give a detectable signal on one of these assays, such as an ELISA, that is preferably at least three times greater than background. Once the titer of the animal has reached a plateau in terms of its reactivity to the protein, larger quantities of antisera may be readily obtained either by weekly bleedings, or by exsanguinating the animal. [0198]
  • Polyclonal antibodies that bind specifically to the TGF-beta binding protein or peptide may then be purified from such antisera, for example, by affinity chromatography using protein A. Alternatively, affinity chromatography may be performed wherein the TGF-beta binding protein or peptide or an antibody specific for an Ig constant region of the particular immunized animal species is immobilized on a suitable solid support. [0199]
  • Antibodies for use in the invention include monoclonal antibodies that are prepared by conventional immunization and cell fusion procedures as described herein an known in the art. Monoclonal antibodies may be readily generated using conventional techniques (see, e.g., Kohler et al., [0200] Nature 256:495, 1975; Coligan et al. (eds.), Current Protocols in Immunology, 1:2.5.1-2.6.7 (John Wiley & Sons 1991) [“Coligan”]; U.S. Pat. Nos. RE 32,011, 4,902,614, 4,543,439, and 4,411,993 which are incorporated herein by reference; see also Monoclonal Antibodies, Hybridomas: A New Dimension in Biological Analyses, Plenum Press, Kennett, McKearn, and Bechtol (eds.), 1980, and Antibodies: A Laboratory Manual, Harlow and Lane (eds.), Cold Spring Harbor Laboratory Press, 1988, which are also incorporated herein by reference; Picksley et al., “Production of monoclonal antibodies against proteins expressed in E. coli,” in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 93 (Oxford University Press 1995)). Antibody fragments may be derived therefrom using any suitable standard technique such as proteolytic digestion, or optionally, by proteolytic digestion (for example, using papain or pepsin) followed by mild reduction of disulfide bonds and alkylation. Alternatively, such fragments may also be generated by recombinant genetic engineering techniques.
  • Briefly, within one embodiment a subject animal such as a rat or mouse or hamster is immunized with TGF-beta binding-protein or a portion of a region thereof, including peptides within a region, as described herein. The protein may be admixed with an adjuvant such as Freund's complete or incomplete adjuvant or Ribi adjuvant in order to increase the resultant immune response. Between one and three weeks after the initial immunization the animal may be reimmunized with another booster immunization, and tested for reactivity to the protein using assays described herein. Once the animal has reached a plateau in its reactivity to the injected protein, it is sacrificed, and organs which contain large numbers of B cells such as the spleen and lymph nodes are harvested. The harvested spleen and/or lymph node cell suspensions are fused with a suitable myeloma cell that is drug-sensitized in order to create a “hybridoma” which secretes monoclonal antibody. Suitable myeloma lines include, for example, NS-0, SP20, NS-1 (ATCC No. TIB 18), and P3X63-Ag 8.653 (ATCC No. CRL 1580). [0201]
  • The lymphoid (e.g., spleen) cells and the myeloma cells may be combined for a few minutes with a membrane fusion-promoting agent, such as polyethylene glycol or a nonionic detergent, and then plated at low density on a selective medium that supports the growth of hybridoma cells but not unfused myeloma cells. Following the fusion, the cells may be placed into culture plates containing a suitable medium, such as RPMI 1640, or DMEM (Dulbecco's Modified Eagles Medium) (JRH Biosciences, Lenexa, Kans.), as well as additional ingredients, such as fetal bovine serum (FBS, i.e., from Hyclone, Logan, Utah, or JRH Biosciences). Additionally, the medium should contain a reagent which selectively allows for the growth of fused spleen and myeloma cells such as HAT (hypoxanthine, aminopterin, and thymidine) (Sigma Chemical Co., St. Louis, Mo.). After about seven days, the resulting fused cells or hybridomas may be screened in order to determine the presence of antibodies which are reactive with TGF-beta binding-protein (depending on the antigen used), and which block, impair, or inhibit the binding of TGF-beta binding-protein to a TGF-beta family member. Hybridomas that produce monoclonal antibodies that specifically bind to sclerostin or a variant thereof are preferred. [0202]
  • A wide variety of assays may be utilized to determine the presence of antibodies which are reactive against the proteins of the present invention, including for example countercurrent immuno-electrophoresis, radioimmunoassays, radioimmunoprecipitations, enzyme-linked immuno-sorbent assays (ELISA), dot blot assays, western blots, immunoprecipitation, inhibition or competition assays, and sandwich assays (see U.S. Pat. Nos. 4,376,110 and 4,486,530; see also [0203] Antibodies: A Laboratory Manual, Harlow and Lane (eds.), Cold Spring Harbor Laboratory Press, 1988). The hybridomas are cloned, for example, by limited dilution cloning or by soft agar plaque isolation, and reassayed. Thus, a hybridoma producing antibodies reactive against the desired protein may be isolated.
  • The monoclonal antibodies from the hybridoma cultures may be isolated from the supernatants of hybridoma cultures. An alternative method for production of a murine monoclonal antibody is to inject the hybridoma cells into the peritoneal cavity of a syngeneic mouse, for example, a mouse that has been treated (e.g., pristane-primed) to promote formation of ascites fluid containing the monoclonal antibody. Monoclonal antibodies can be isolated and purified by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al., “Purification of Immunoglobulin G (IgG),” in [0204] Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)). Monoclonal antibodies may be purified by affinity chromatography using an appropriate ligand selected based on particular properties of the antibody (e.g., heavy or light chain isotype, binding specificity, etc.). Examples of a suitable ligand, immobilized on a solid support, include Protein A, Protein G, an anti-constant region (light chain or heavy chain) antibody, an anti-idiotype antibody, and a TGF-beta binding protein, or fragment or variant thereof.
  • In addition, an anti-TGF-beta binding-protein antibody of the present invention may be a human monoclonal antibody. Human monoclonal antibodies may be generated by any number of techniques with which those having ordinary skill in the art will be familiar. Such methods include, but are not limited to, Epstein Barr Virus (EBV) transformation of human peripheral blood cells (e.g., containing B lymphocytes), in vitro immunization of human B cells, fusion of spleen cells from immunized transgenic mice carrying inserted human immunoglobulin genes, isolation from human immunoglobulin V region phage libraries, or other procedures as known in the art and based on the disclosure herein. For example, human monoclonal antibodies may be obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge. Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., [0205] Nature Genet. 7:13, 1994; Lonberg et al., Nature 368:856, 1994; Taylor et al., Int. Immun. 6:579, 1994; U.S. Pat. No. 5,877,397; Bruggemann et al., 1997 Curr. Opin. Biotechnol. 8:455-58; Jakobovits et al., 1995 Ann. N. Y. Acad. Sci. 764:525-35. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. (See also Bruggemann et al., Curr. Opin. Biotechnol. 8:455-58 (1997)). For example, human immunoglobulin transgenes may be mini-gene constructs, or transloci on yeast artificial chromosomes, which undergo B cell-specific DNA rearrangement and hypermutation in the mouse lymphoid tissue. Human monoclonal antibodies may be obtained by immunizing the transgenic mice, which may then produce human antibodies specific for the antigen. Lymphoid cells of the immunized transgenic mice can be used to produce human antibody-secreting hybridomas according to the methods described herein. Polyclonal sera containing human antibodies may also be obtained from the blood of the immunized animals.
  • Another method for generating human TGF-beta binding protein specific monoclonal antibodies includes immortalizing human peripheral blood cells by EBV transformation. See, e.g., U.S. Pat. No. 4,464,456. Such an immortalized B cell line (or lymphoblastoid cell line) producing a monoclonal antibody that specifically binds to a TGF-beta binding protein (or a variant or fragment thereof) can be identified by immunodetection methods as provided herein, for example, an ELISA, and then isolated by standard cloning techniques. The stability of the lymphoblastoid cell line producing an anti-TGF-beta binding protein antibody may be improved by fusing the transformed cell line with a murine myeloma to produce a mouse-human hybrid cell line according to methods known in the art (see, e.g., Glasky et al., [0206] Hybridoma 8:377-89 (1989)). Still another method to generate human monoclonal antibodies is in vitro immunization, which includes priming human splenic B cells with antigen, followed by fusion of primed B cells with a heterohybrid fusion partner. See, e.g., Boerner et al., 1991 J Immunol. 147:86-95.
  • In certain embodiments, a B cell that is producing an anti-SOST antibody is selected and the light chain and heavy chain variable regions are cloned from the B cell according to molecular biology techniques known in the art (WO 92/02551; U.S. Pat. No. 5,627,052; Babcook et al., [0207] Proc. Natl. Acad. Sci. USA 93:7843-48 (1996)) and described herein. Preferably B cells from an immunized animal are isolated from the spleen, lymph node, or peripheral blood sample by selecting a cell that is producing an antibody that specifically binds to SOST. B cells may also be isolated from humans, for example, from a peripheral blood sample. Methods for detecting single B cells that are producing an antibody with the desired specificity are well known in the art, for example, by plaque formation, fluorescence-activated cell sorting, in vitro stimulation followed by detection of specific antibody, and the like. Methods for selection of specific antibody producing B cells include, for example, preparing a single cell suspension of B cells in soft agar that contains SOST or a peptide fragment thereof. Binding of the specific antibody produced by the B cell to the antigen results in the formation of a complex, which may be visible as an immunoprecipitate. After the B cells producing the specific antibody are selected, the specific antibody genes may be cloned by isolating and amplifying DNA or mRNA according to methods known in the art and described herein.
  • For particular uses, fragments of anti-TGF-beta binding protein antibodies may be desired. Antibody fragments, F(ab′)[0208] 2, Fab, Fab′, Fv, Fe, Fd, retain the antigen binding site of the whole antibody and therefore bind to the same epitope. These antigen-binding fragments derived from an antibody can be obtained, for example, by proteolytic hydrolysis of the antibody, for example, pepsin or papain digestion of whole antibodies according to conventional methods. As an illustration, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)2. This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab′ monovalent fragments. Optionally, the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages. As an alternative, an enzymatic cleavage using papain produces two monovalent Fab fragments and an Fc fragment directly. These methods are described, for example, by Goldenberg, U.S. Pat. No. 4,331,647, Nisonoff et al., Arch. Biochem. Biophys. 89:230, 1960; Porter, Biochem. J. 73:119, 1959; Edelman et al., in Methods in Enzymology 1:422 (Academic Press 1967); and by Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4. Other methods for cleaving antibodies, such as separating heavy chains to form monovalent light-heavy chain fragments (Fd), further cleaving of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody.
  • An antibody fragment may also be any synthetic or genetically engineered protein that acts like an antibody in that it binds to a specific antigen to form a complex. For example, antibody fragments include isolated fragments consisting of the light chain variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (scFv proteins), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region. The antibody of the present invention preferably comprises at least one variable region domain. The variable region domain may be of any size or amino acid composition and will generally comprise at least one hypervariable amino acid sequence responsible for antigen binding and which is adjacent to or in frame with one or more framework sequences. In general terms, the variable (V) region domain may be any suitable arrangement of immunoglobulin heavy (V[0209] H) and/or light (VL) chain variable domains. Thus, for example, the V region domain may be monomeric and be a VH or VL domain, which is capable of independently binding antigen with acceptable affinity. Alternatively, the V region domain may be dimeric and contain VH-VH, VH-VL, or VL-VL, dimers. Preferably, the V region dimer comprises at least one VH and at least one VL chain that are non-covalently associated (hereinafter referred to as Fv). If desired, the chains may be covalently coupled either directly, for example via a disulphide bond between the two variable domains, or through a linker, for example a peptide linker, to form a single chain Fv (scFv).
  • The variable region domain may be any naturally occurring variable domain or an engineered version thereof. By engineered version is meant a variable region domain that has been created using recombinant DNA engineering techniques. Such engineered versions include those created, for example, from a specific antibody variable region by insertions, deletions, or changes in or to the amino acid sequences of the specific antibody. Particular examples include engineered variable region domains containing at least one CDR and optionally one or more framework amino acids from a first antibody and the remainder of the variable region domain from a second antibody. [0210]
  • The variable region domain may be covalently attached at a C-terminal amino acid to at least one other antibody domain or a fragment thereof. Thus, for example, a V[0211] H, domain that is present in the variable region domain may be linked to an immunoglobulin C H1 domain, or a fragment thereof. Similarly a VL domain may be linked to a CK domain or a fragment thereof. In this way, for example, the antibody may be a Fab fragment wherein the antigen binding domain contains associated VH and VL domains covalently linked at their C-termini to a CH1 and CK domain, respectively. The CH1 domain may be extended with further amino acids, for example to provide a hinge region or a portion of a hinge region domain as found in a Fab′ fragment, or to provide further domains, such as antibody CH2 and CH3 domains.
  • Another form of an antibody fragment is a peptide comprising for a single complementarity-determining region (CDR). CDR peptides (“minimal recognition units”) can be obtained by constructing polynucleotides that encode the CDR of an antibody of interest. Such polynucleotides are prepared, for example, by using the polymerase chain reaction to synthesize the variable region using mRNA of antibody-producing cells as a template (see, for example, Larrick et al., [0212] Methods: A Companion to Methods in Enzymology 2:106, 1991; Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies,” in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al. (eds.), page 166 (Cambridge University Press 1995); and Ward et al., “Genetic Manipulation and Expression of Antibodies,” in Monoclonal Antibodies: Principles and Applications, Birch et al., (eds.), page 137 (Wiley-Liss, Inc. 1995)).
  • Alternatively, the antibody may be a recombinant or engineered antibody obtained by the use of recombinant DNA techniques involving the manipulation and re-expression of DNA encoding antibody variable and/or constant regions. Such DNA is known and/or is readily available from DNA libraries including for example phage-antibody libraries (see Chiswell and McCafferty, [0213] Tibtech. 10:80-84 (1992)) or if desired can be synthesized. Standard molecular biology and/or chemistry procedures may be used to sequence and manipulate the DNA, for example, to introduce codons to create cysteine residues, or to modify, add or delete other amino acids or domains as desired.
  • Chimeric antibodies, specific for a TGF-beta binding protein, and which include humanized antibodies, may also be generated according to the present invention. A chimeric antibody has at least one constant region domain derived from a first mammalian species and at least one variable region domain derived from a second, distinct mammalian species (see, e.g., Morrison et al., [0214] Proc. Natl. Acad. Sci. USA, 81:6851-55 (1984)). In preferred embodiments, a chimeric antibody may be constructed by cloning the polynucleotide sequence that encodes at least one variable region domain derived from a non-human monoclonal antibody, such as the variable region derived from a murine, rat, or hamster monoclonal antibody, into a vector containing a nucleotide sequence that encodes at least one human constant region (see, e.g., Shin et al., Methods Enzymol. 178:459-76 (1989); Walls et al., Nucleic Acids Res. 21:2921-29 (1993)). By way of example, the polynucleotide sequence encoding the light chain variable region of a murine monoclonal -antibody may be inserted into a vector containing a nucleotide sequence encoding the human kappa light chain constant region sequence. In a separate vector, the polynucleotide sequence encoding the heavy chain variable region of the monoclonal antibody may be cloned in frame with sequences encoding a human IgG constant region, for example, the human IgG1 constant region. The particular human constant region selected may depend upon the effector functions desired for the particular antibody (e.g. complement fixing, binding to a particular Fc receptor, etc.). Preferably, the constructed vectors will be transfected into eukaryotic cells for stable expression of the chimeric antibody. Another method known in the art for generating chimeric antibodies is homologous recombination (e.g., U.S. Pat. No. 5,482,856).
  • A non-human/human chimeric antibody may be further genetically engineered to create a “humanized” antibody. Such a humanized antibody may comprise a plurality of CDRs derived from an immunoglobulin of a non-human mammalian species, at least one human variable framework region, and at least one human immunoglobulin constant region. Useful strategies for designing humanized antibodies may include, for example by way of illustration and not limitation, identification of human variable framework regions that are most homologous to the non-human framework regions of the chimeric antibody. Without wishing to be bound by theory, such a strategy may increase the likelihood that the humanized antibody will retain specific binding affinity for a TGF-beta binding protein, which in some preferred embodiments may be substantially the same affinity for a TGF-beta binding protein or variant or fragment thereof, and in certain other preferred embodiments may be a greater affinity for TGF-beta binding protein. See, e.g., Jones et al., 1986 [0215] Nature 321:522-25; Riechmann et al., 1988 Nature 332:323-27. Designing such a humanized antibody may therefore include determining CDR loop conformations and structural determinants of the non-human variable regions, for example, by computer modeling, and then comparing the CDR loops and determinants to known human CDR loop structures and determinants. See, e.g., Padlan et al., 1995 FASEB 9:133-39; Chothia et al., 1989 Nature, 342:377-383. Computer modeling may also be used to compare human structural templates selected by sequence homology with the non-human variable regions. See, e.g., Bajorath et al., 1995 Ther. Immunol. 2:95-103; EP-0578515-A3. If humanization of the non-human CDRs results in a decrease in binding affinity, computer modeling may aid in identifying specific amino acid residues that could be changed by site-directed or other mutagenesis techniques to partially, completely or supra-optimally (i.e., increase to a level greater than that of the non-humanized antibody) restore affinity. Those having ordinary skill in the art are familiar with these techniques, and will readily appreciate numerous variations and modifications to such design strategies.
  • One such method for preparing a humanized antibody is called veneering. As used herein, the terms “veneered FRs” and “recombinantly veneered FRs” refer to the selective replacement of FR residues from, e.g., a rodent heavy or light chain V region, with human FR residues in order to provide a xenogeneic molecule comprising an antigen-binding site that retains substantially all of the native FR polypeptide folding structure. Veneering techniques are based on the understanding that the ligand binding characteristics of an antigen-binding site are determined primarily by the structure and relative disposition of the heavy and light chain CDR sets within the antigen-binding surface. Davies et al., [0216] Ann. Rev. Biochem. 59:439-73, 1990. Thus, antigen binding specificity can be preserved in a humanized antibody only wherein the CDR structures, their interaction with each other, and their interaction with the rest of the V region domains are carefully maintained. By using veneering techniques, exterior (e.g., solvent-accessible) FR residues that are readily encountered by the immune system are selectively replaced with human residues to provide a hybrid molecule that comprises either a weakly immunogenic, or substantially non-immunogenic veneered surface.
  • The process of veneering makes use of the available sequence data for human antibody variable domains compiled by Kabat et al., in [0217] Sequences of Proteins of Immunological Interest, 4th ed., (U.S. Dept. of Health and Human Services, U.S. Government Printing Office, 1987), updates to the Kabat database, and other accessible U.S. and foreign databases (both nucleic acid and protein). Solvent accessibilities of V region amino acids can be deduced from the known three-dimensional structure for human and murine antibody fragments. Initially, the FRs of the variable domains of an antibody molecule of interest are compared with corresponding FR sequences of human variable domains obtained from the above-identified sources. The most homologous human V regions are then compared residue by residue to corresponding murine amino acids. The residues in the murine FR that differ from the human counterpart are replaced by the residues present in the human moiety using recombinant techniques well known in the art. Residue switching is only carried out with moieties which are at least partially exposed (solvent accessible), and care is exercised in the replacement of amino acid residues that may have a significant effect on the tertiary structure of V region domains, such as proline, glycine, and charged amino acids.
  • In this manner, the resultant “veneered” antigen-binding sites are thus designed to retain the rodent CDR residues, the residues substantially adjacent to the CDRs, the residues identified as buried or mostly buried (solvent inaccessible), the residues believed to participate in non-covalent (e.g., electrostatic and hydrophobic) contacts between heavy and light chain domains, and the residues from conserved structural regions of the FRs which are believed to influence the “canonical” tertiary structures of the CDR loops. These design criteria are then used to prepare recombinant nucleotide sequences that combine the CDRs of both the heavy and light chain of a antigen-binding site into human-appearing FRs that can be used to transfect mammalian cells for the expression of recombinant human antibodies that exhibit the antigen specificity of the rodent antibody molecule. [0218]
  • An additional method for selecting antibodies that specifically bind to a TGF-beta binding protein or variant or fragment thereof is by phage display. See, e.g., Winter et al., 1994 [0219] Annu. Rev. Immunol. 12:433-55; Burton et al., 1994 Adv. Immunol. 57:191-280. Human or murine immunoglobulin variable region gene combinatorial libraries may be created in phage vectors that can be screened to select Ig fragments (Fab, Fv, sFv, or multimers thereof) that bind specifically to TGF-beta binding protein or variant or fragment thereof. See, e.g., U.S. Pat. No. 5,223,409; William D. Huse et al., “Generation of a Large Combinational Library of the Immunoglobulin Repertoire in Phage Lambda,” Science 246:1275-1281, December 1989; see also L. Sastry et al., “Cloning of the Immunological Repertoire in Escherichia coli for Generation of Monoclonal Catalytic Antibodies: Construction of a Heavy Chain Variable Region-Specific cDNA Library,” Proc. Natl. Acad. Sci. USA 86:5728-5732, August 1989; see also Michelle Alting-Mees et al., “Monoclonal Antibody Expression Libraries: A Rapid Alternative to Hybridomas,” Strategies in Molecular Biology 3:1-9, January 1990; Kang et al., 1991 Proc. Natl. Acad. Sci. USA 88:4363-66; Hoogenboom et al., 1992 J. Molec. Biol. 227:381-388; Schlebusch et al., 1997 Hybridoma 16:47-52 and references cited therein). A commercial system is available from Stratagene (La Jolla, Calif.) which enables the production of antibodies through recombinant techniques. Briefly, mRNA is isolated from a B cell population, and utilized to create heavy and light chain immunoglobulin cDNA expression libraries in the λ ImmunoZap(H) and λImmunoZap(L) vectors. Positive plaques may subsequently be converted to a non-lytic plasmid which allows high level expression of monoclonal antibody fragments from E. coli. Alternatively, a library containing a plurality of polynucleotide sequences encoding Ig variable region fragments may be inserted into the genome of a filamentous bacteriophage, such as M13 or a variant thereof, in frame with the sequence encoding a phage coat protein. A fusion protein may be a fusion of the coat protein with the light chain variable region domain and/or with the heavy chain variable region domain. According to certain embodiments, immunoglobulin Fab fragments may also be displayed on a phage particle (see, e.g., U.S. Pat. No. 5,698,426). These vectors may be screened individually or co-expressed to form Fab fragments or antibodies (see Huse et al., supra; see also Sastry et al., supra).
  • Similarly, portions or fragments, such as Fab and Fv fragments, of antibodies may also be constructed utilizing conventional enzymatic digestion or recombinant DNA techniques to incorporate the variable regions of a gene which encodes a specifically binding antibody. Within one embodiment, the genes which encode the variable region from a hybridoma producing a monoclonal antibody of interest are amplified using nucleotide primers for the variable region. These primers may be synthesized by one of ordinary skill in the art, or may be purchased from commercially available sources. Stratagene (La Jolla, Calif.) sells primers for mouse and human variable regions including, among others, primers for V[0220] Ha, VHb, VHc, VHd, CH1, VL and CL regions. These primers may be utilized to amplify heavy or light chain variable regions, which may then be inserted into vectors such as ImmunoZAP™ H or ImmunoZAP™ L (Stratagene), respectively. These vectors may then be introduced into E. coli, yeast, or mammalian-based systems for expression. Utilizing these techniques, large amounts of a single-chain protein containing a fusion of the VH and VL domains may be produced (see Bird et al., Science 242:423-426, 1988). In addition, such techniques may be utilized to change a “murine” antibody to a “human” antibody, without altering the binding specificity of the antibody.
  • In certain particular embodiments of the invention, combinatorial phage libraries may also be used for humanization of non-human variable regions. See, e.g., Rosok et al., 1996 [0221] J. Biol. Chem. 271:22611-18; Rader et al., 1998 Proc. Natl. Acad. Sci. USA 95:8910-15. A phage library may be screened to select an Ig variable region fragment of interest by immunodetection methods known in the art and described herein, and the DNA sequence of the inserted immunoglobulin gene in the phage so selected may be determined by standard techniques. See, Sambrook et al., 2001 Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press. The selected Ig-encoding sequence may then be cloned into another suitable vector for expression of the Ig fragment or, optionally, may be cloned into a vector containing Ig constant regions, for expression of whole immunoglobulin chains.
  • In certain other embodiments, the invention contemplates SOST-specific antibodies that are multimeric antibody fragments. Useful methodologies are described generally, for example in Hayden et al. 1997, [0222] Curr Opin. Immunol. 9:201-12; Coloma et al., 1997 Nat. Biotechnol. 15:159-63). For example, multimeric antibody fragments may be created by phage techniques to form miniantibodies (U.S. Pat. No. 5,910,573) or diabodies (Holliger et al., 1997, Cancer Immunol. Immunother. 45:128-130).
  • In certain embodiments of the invention, an antibody specific for SOST may be an antibody that is expressed as an intracellular protein. Such intracellular antibodies are also referred to as intrabodies and may comprise an Fab fragment, or preferably comprise a scFv fragment (see, e.g., Lecerf et al., [0223] Proc. Natl. Acad. Sci. USA 98:4764-49 (2001). The framework regions flanking the CDR regions can be modified to improve expression levels and solubility of an intrabody in an intracellular reducing environment (see, e.g., Worn et al., J. Biol. Chem. 275:2795-803 (2000). An intrabody may be directed to a particular cellular location or organelle, for example by constructing a vector that comprises a polynucleotide sequence encoding the variable regions of an intrabody that may be operatively fused to a polynucleotide sequence that encodes a particular target antigen within the cell (see, e.g., Graus-Porta et al., Mol. Cell Biol. 15:1182-91 (1995); Lener et al., Eur. J. Biochem. 267:1196-205 (2000)). An intrabody may be introduced into a cell by a variety of techniques available to the skilled artisan including via a gene therapy vector, or a lipid mixture (e.g., Provectin™ manufactured by Imgenex Corporation, San Diego, Calif.), or according to photochemical internalization methods.
  • Introducing amino acid mutations into an immunoglobulin molecule specific for a TGF-beta binding protein may be useful to increase the specificity or affinity for TGF-beta binding protein or to alter an effector function. Immunoglobulins with higher affinity for TGF-beta binding protein may be generated by site-directed mutagenesis of particular residues. Computer assisted three-dimensional molecular modeling may be employed to identify the amino acid residues to be changed, in order to improve affinity for the TGF-beta binding protein. See, e.g., Mountain et al., 1992, [0224] Biotechnol. Genet Eng. Rev. 10: 1-142. Alternatively, combinatorial libraries of CDRs may be generated in M13 phage and screened for immunoglobulin fragments with improved affinity. See, e.g., Glaser et al., 1992, J. Immunol. 149:3903-3913; Barbas et al., 1994 Proc. Natl. Acad. Sci. USA 91:3809-13; U.S. Pat. No. 5,792,456.
  • Effector functions may also be altered by site-directed mutagenesis. See, e.g., Duncan et al., 1988 [0225] Nature 332:563-64; Morgan et al., 1995 Immunology 86:319-24; Eghtedarzedeh-Kondri et al., 1997 Biotechniques 23:830-34. For example, mutation of the glycosylation site on the Fc portion of the immunoglobulin may alter the ability of the immunoglobulin to fix complement. See, e.g., Wright et al., 1997 Trends Biotechnol. 15:26-32. Other mutations in the constant region domains may alter the ability of the immunoglobulin to fix complement, or to effect antibody-dependent cellular cytotoxicity. See, e.g., Duncan et al., 1988 Nature 332:563-64; Morgan et al., 1995 Immunology 86:319-24; Sensel et al., 1997 Mol. Immunol. 34:1019-29.
  • According to certain embodiments, non-human, human, or humanized heavy chain and light chain variable regions of any of the Ig molecules described herein may be constructed as single chain Fv (scFv) polypeptide fragments (single chain antibodies). See, e.g., Bird et al., 1988 [0226] Science 242:423-426; Huston et al., 1988 Proc. Natl. Acad. Sci. USA 85:5879-5883. Multi-functional scFv fusion proteins may be generated by linking a polynucleotide sequence encoding an scFv polypeptide in-frame with at least one polynucleotide sequence encoding any of a variety of known effector proteins. These methods are known in the art, and are disclosed, for example, in EP-B1-0318554, U.S. Pat. No. 5,132,405, U.S. Pat. No. 5,091,513, and U.S. Pat. No. 5,476,786. By way of example, effector proteins may include immunoglobulin constant region sequences. See, e.g., Hollenbaugh et al., 1995 J. Immunol. Methods 188:1-7. Other examples of effector proteins are enzymes. As a non-limiting example, such an enzyme may provide a biological activity for therapeutic purposes (see, e.g., Siemers et al., 1997 Bioconjug. Chem. 8:510-19), or may provide a detectable activity, such as horseradish peroxidase-catalyzed conversion of any of a number of well-known substrates into a detectable product, for diagnostic uses. Still other examples of scFv fusion proteins include Ig-toxin fusions, or immunotoxins, wherein the scFv polypeptide is linked to a toxin.
  • The scFv or any antibody fragment described herein may, in certain embodiments, be fused to peptide or polypeptide domains that permits detection of specific binding between the fusion protein and antigen (e.g., a TGF-beta binding protein). For example, the fusion polypeptide domain may be an affinity tag polypeptide for detecting binding of the scFv fusion protein to a TGF-beta binding protein by any of a variety of techniques with which those skilled in the art will be familiar. Examples of a peptide tag, include avidin, streptavidin or His (e.g., polyhistidine). Detection techniques may also include, for example, binding of an avidin or streptavidin fusion protein to biotin or to a biotin mimetic sequence (see, e.g., Luo et al., 1998 [0227] J. Biotechnol. 65:225 and references cited therein), direct covalent modification of a fusion protein with a detectable moiety (e.g., a labeling moiety), non-covalent binding of the fusion protein to a specific labeled reporter molecule, enzymatic modification of a detectable substrate by a fusion protein that includes a portion having enzyme activity, or immobilization (covalent or non-covalent) of the fusion protein on a solid-phase support. Other useful affinity polypeptides for construction of scFv fusion proteins may include streptavidin fusion proteins, as disclosed, for example, in WO 89/03422, U.S. Pat. No. 5,489,528, U.S. Pat. No. 5,672,691, WO 93/24631, U.S. Pat. No. 5,168,049, U.S. Pat. No. 5,272,254; avidin fusion proteins (see, e.g., EP 511,747); an enzyme such as glutathione-S-transferase; and Staphylococcus aureus protein A polypeptide.
  • The polynucleotides encoding an antibody or fragment thereof that specifically bind a TGF-beta binding protein, as described herein, may be propagated and expressed according to any of a variety of well-known procedures for nucleic acid excision, ligation, transformation, and transfection using any number of known expression vectors. Thus, in certain embodiments expression of an antibody fragment may be preferred in a prokaryotic host, such as [0228] Escherichia coli (see, e.g., Pluckthun et al., 1989 Methods Enzymol. 178:497-515). In certain other embodiments, expression of the antibody or a fragment thereof may be preferred in a eukaryotic host cell, including yeast (e.g., Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Pichia pastoris), animal cells (including mammalian cells) or plant cells. Examples of suitable animal cells include, but are not limited to, myeloma (such as a mouse NSO line), COS, CHO, or hybridoma cells. Examples of plant cells include tobacco, corn, soybean, and rice cells.
  • Once suitable antibodies have been obtained, they may be isolated or purified by many techniques well known to those of ordinary skill in the art (see [0229] Antibodies: A Laboratory Manual, Harlow and Lane (eds.), Cold Spring Harbor Laboratory Press, 1988). Suitable techniques include peptide or protein affinity columns (including use of anti-constant region antibodies attached to the column matrix), HPLC or RP-HPLC, purification on protein A or protein G columns, or any combination of these techniques.
  • c. Mutant TGF-Beta Binding-Proteins [0230]
  • As described herein and below in the Examples (e.g., Examples 8 and 9), altered versions of TGF-beta binding-protein which compete with native TGF-beta binding-protein's ability to block the activity of a particular TGF-beta family member should lead to increased bone density. Thus, mutants of TGF-beta binding-protein which bind to the TGF-beta family member but do not inhibit the function of the TGF-beta family member would meet the criteria. The mutant versions must effectively compete with the endogenous inhibitory functions of TGF-beta binding-protein. [0231]
  • d. Production of Proteins [0232]
  • Polypeptides described herein include the TGF binding protein sclerostin and variants thereof and antibodies or fragments thereof that specifically bind to sclerostin. The polynucleotides that encode these polypeptides include derivatives of the genes that are substantially similar to the genes and isolated nucleic acid molecules, and, when appropriate, the proteins (including peptides and polypeptides) that are encoded by the genes and their derivatives. As used herein, a nucleotide sequence is deemed to be “substantially similar” if (a) the nucleotide sequence is derived from the coding region of the above-described genes and nucleic acid molecules and includes, for example, portions of the sequence or allelic variations of the sequences discussed above, or alternatively, encodes a molecule which inhibits the binding of TGF-beta binding-protein to a member of the TGF-beta family; (b) the nucleotide sequence is capable of hybridization to nucleotide sequences of the present invention under moderate, high or very high stringency (see Sambrook et al., [0233] Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, NY, 1989); and/or (c) the DNA sequences are degenerate as a result of the genetic code to the DNA sequences defined in (a) or (b). Further, the nucleic acid molecule disclosed herein includes both complementary and non-complementary sequences, provided the sequences otherwise meet the criteria set forth herein. Within the context of the present invention, high stringency means standard hybridization conditions (e.g., 5× SSPE, 0.5% SDS at 65° C., or the equivalent).
  • The structure of the proteins encoded by the nucleic acid molecules described herein may be predicted from the primary translation products using the hydrophobicity plot function of, for example, P/C Gene or Intelligenetics Suite (Intelligenetics, Mountain View, Calif.), or according to the methods described by Kyte and Doolittle ([0234] J. Mol. Biol. 157:105-132, 1982).
  • Proteins of the present invention may be prepared in the form of acidic or basic salts, or in neutral form. In addition, individual amino acid residues may be modified by oxidation or reduction. Furthermore, various substitutions, deletions, or additions may be made to the amino acid or nucleic acid sequences, the net effect of which is to retain or further enhance or decrease the biological activity of the mutant or wild-type protein. Moreover, due to degeneracy in the genetic code, for example, there may be considerable variation in nucleotide sequences encoding the same amino acid sequence. [0235]
  • Other derivatives of the proteins disclosed herein include conjugates of the proteins along with other proteins or polypeptides. This may be accomplished, for example, by the synthesis of N-terminal or C-terminal fusion proteins which may be added to facilitate purification or identification of proteins (see U.S. Pat. No. 4,851,341, see also, Hopp et al., [0236] Bio/Technology 6:1204, 1988.) Alternatively, fusion proteins such as Flag®/TGF-beta binding-protein be constructed in order to assist in the identification, expression, and analysis of the protein.
  • Proteins of the present invention may be constructed using a wide variety of techniques described herein. Further, mutations may be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a derivative having the desired amino acid insertion, substitution, or deletion. [0237]
  • Alternatively, oligonucleotide-directed site-specific (or segment specific) mutagenesis procedures may be employed to provide an altered gene or nucleic acid molecule having particular codons altered according to the substitution, deletion, or insertion required. Exemplary methods of making the alterations set forth above are disclosed by Walder et al. ([0238] Gene 42:133, 1986); Bauer et al. (Gene 37:73, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods, Plenum Press, 1981); and Sambrook et al. (supra). Deletion or truncation derivatives of proteins (e.g., a soluble extracellular portion) may also be constructed by utilizing convenient restriction endonuclease sites adjacent to the desired deletion. Subsequent to restriction, overhangs may be filled in and the DNA religated. Exemplary methods of making the alterations set forth above are disclosed by Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, 1989).
  • Mutations which are made in the nucleic acid molecules of the present invention preferably preserve the reading frame of the coding sequences. Furthermore, the mutations will preferably not create complementary regions that when transcribed could hybridize to produce secondary mRNA structures, such as loops or hairpins, that would adversely affect translation of the mRNA. Although a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutants screened for gain or loss or retention of biological activity. Alternatively, mutations may be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a derivative having the desired amino acid insertion, substitution, or deletion. [0239]
  • Nucleic acid molecules which encode proteins of the present invention may also be constructed utilizing techniques such as PCR mutagenesis, chemical mutagenesis (Drinkwater and Klinedinst, [0240] PNAS 83:3402-3406, 1986), by forced nucleotide misincorporation (e.g., Liao and Wise Gene 88:107-111, 1990), or by use of randomly mutagenized oligonucleotides (Horwitz et al., Genome 3:112-117, 1989).
  • The present invention also provides for the manipulation and expression of the above described genes and nucleic acid molecules by culturing host cells containing a vector capable of expressing the above-described genes. Such vectors or vector constructs include either synthetic or cDNA-derived nucleic acid molecules encoding the desired protein, which are operably linked to suitable transcriptional or translational regulatory elements. Suitable regulatory elements may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, insect, or plant genes. Selection of appropriate regulatory elements is dependent on the host cell chosen, and may be readily accomplished by one of ordinary skill in the art. Examples of regulatory elements include a transcriptional promoter and enhancer or RNA polymerase binding sequence, a transcriptional terminator, and a ribosomal binding sequence, including a translation initiation signal. [0241]
  • Nucleic acid molecules that encode any of the proteins described above may be readily expressed by a wide variety of prokaryotic and eukaryotic host cells, including bacterial, mammalian, yeast or other fungi, viral, insect, or plant cells. Methods for transforming or transfecting such cells to express foreign DNA are well known in the art (see, e.g., Itakura et al., U.S. Pat. No. 4,704,362; Hinnen et al., [0242] Proc. Natl. Acad. Sci. USA 75:1929-1933, 1978; Murray et al., U.S. Pat. No. 4,801,542; Upshall et al., U.S. Pat. No. 4,935,349; Hagen et al., U.S. Pat. No. 4,784,950; Axel et al., U.S. Pat. No. 4,399,216; Goeddel et al., U.S. Pat. No. 4,766,075; and Sambrook et al. Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, 1989; for plant cells see Czako and Marton, Plant Physiol. 104:1067-1071, 1994; and Paszkowski et al., Biotech. 24:387-392, 1992).
  • Bacterial host cells suitable for carrying out the present invention include [0243] E. coli, B. subtilis, Salmonella typhimurium, and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, as well as many other bacterial species well known to one of ordinary skill in the art and described herein. A representative example of a bacterial host cell includes E. coli DH5α (Stratagene, La Jolla, Calif.).
  • Bacterial expression vectors preferably comprise a promoter which functions in the host cell, one or more selectable phenotypic markers, and a bacterial origin of replication. Representative promoters include the β-lactamase (penicillinase) and lactose promoter system (see Chang et al., [0244] Nature 275:615, 1978), the T7 RNA polymerase promoter (Studier et al., Meth. Enzymol. 185:60-89, 1990), the lambda promoter (Elvin et al., Gene 87:123-126, 1990), the trp promoter (Nichols and Yanofsky, Meth. in Enzymology 101:155, 1983), and the tac promoter (Russell et al., Gene 20:231, 1982). Representative selectable markers include various antibiotic resistance markers such as the kanamycin or ampicillin resistance genes. Many plasmids suitable for transforming host cells are well known in the art, including among others, pBR322 (see Bolivar et al., Gene 2:95, 1977), the pUC plasmids pUC18, pUC19, pUC118, pUC119 (see Messing, Meth. in Enzymology 101:20-77, 1983 and Vieira and Messing, Gene 19:259-268, 1982), and pNH8A, pNH16a, pNH18a, and Bluescript M13 (Stratagene, La Jolla, Calif.).
  • Yeast and fungi host cells suitable for carrying out the present invention include, among others, [0245] Saccharomyces pombe, Saccharomyces cerevisiae, the genera Pichia or Kluyveromyces and various species of the genus Aspergillus (McKnight et al., U.S. Pat. No. 4,935,349). Suitable expression vectors for yeast and fungi include, among others, YCp50 (ATCC No. 37419) for yeast, and the amdS cloning vector pV3 (Turnbull, Bio/Technology 7:169, 1989), YRp7 (Struhl et al., Proc. Natl. Acad. Sci. USA 76:1035-1039, 1978), YEp13 (Broach et al., Gene 8:121-133, 1979), pJDB249 and pJDB219 (Beggs, Nature 275:104-108, 1978) and derivatives thereof.
  • Preferred promoters for use in yeast include promoters from yeast glycolytic genes (Hitzeman et al., [0246] J. Biol. Chem. 255:12073-12080, 1980; Alber and Kawasaki, J. Mol. Appl. Genet. 1:419-434, 1982) or alcohol dehydrogenase genes (Young et al., in Genetic Engineering of Microorganisms for Chemicals, Hollaender et al. (eds.), p. 355, Plenum, New York, 1982; Ammerer, Meth. Enzymol. 101:192-201, 1983). Examples of useful promoters for fungi vectors include those derived from Aspergillus nidulans glycolytic genes, such as the adh3 promoter (McKnight et al., EMBO J 4:2093-2099, 1985). The expression units may also include a transcriptional terminator. An example of a suitable terminator is the adh3 terminator (McKnight et al., supra, 1985).
  • As with bacterial vectors, the yeast vectors will generally include a selectable marker, which may be one of any number of genes that exhibit a dominant phenotype for which a phenotypic assay exists to enable transformants to be selected. Preferred selectable markers are those that complement host cell auxotrophy, provide antibiotic resistance, or enable a cell to utilize specific carbon sources, and include leu2 (Broach et al., ibid.), ura3 (Botstein et al., [0247] Gene 8:17, 1979), or his3 (Struhl et al., ibid.). Another suitable selectable marker is the cat gene, which confers chloramphenicol resistance on yeast cells.
  • Techniques for transforming fungi are well known in the literature and have been described, for instance, by Beggs (ibid.), Hinnen et al. ([0248] Proc. Natl. Acad. Sci. USA 75:1929-1933, 1978), Yelton et al. (Proc. Natl Acad. Sci. USA 81:1740-1747, 1984), and Russell (Nature 301:167-169, 1983). The genotype of the host cell may contain a genetic defect that is complemented by the selectable marker present on the expression vector. Choice of a particular host and selectable marker is well within the level of ordinary skill in the art.
  • Protocols for the transformation of yeast are also well known to those of ordinary skill in the art. For example, transformation may be readily accomplished either by preparation of spheroplasts of yeast with DNA (see Hinnen et al., [0249] PNAS USA 75:1929, 1978) or by treatment with alkaline salts such as LiCl (see Itoh et al., J. Bacteriology 153:163, 1983). Transformation of fungi may also be carried out using polyethylene glycol as described by Cullen et al. (Bio/Technology 5:369, 1987).
  • Viral vectors include those that comprise a promoter that directs the expression of an isolated nucleic acid molecule that encodes a desired protein as described above. A wide variety of promoters may be utilized within the context of the present invention, including for example, promoters such as MoMLV LTR, RSV LTR, Friend MuLV LTR, adenoviral promoter (Ohno et al., [0250] Science 265:781-784, 1994), neomycin phosphotransferase promoter/enhancer, late parvovirus promoter (Koering et al., Hum. Gene Therap. 5:457-463, 1994), Herpes TK promoter, SV40 promoter, metallothionein IIa gene enhancer/promoter, cytomegalovirus immediate early promoter, and the cytomegalovirus immediate late promoter. Within particularly preferred embodiments of the invention, the promoter is a tissue-specific promoter (see e.g., WO 91/02805; EP 0,415,731; and WO 90/07936). Representative examples of suitable tissue specific promoters include neural specific enolase promoter, platelet derived growth factor beta promoter, bone morphogenic protein promoter, human alpha1-chimaerin promoter, synapsin I promoter and synapsin II promoter. In addition to the above-noted promoters, other viral-specific promoters (e.g., retroviral promoters (including those noted above, as well as others such as HIV promoters), hepatitis, herpes (e.g., EBV), and bacterial, fungal or parasitic (e.g., malarial)-specific promoters may be utilized in order to target a specific cell or tissue which is infected with a virus, bacteria, fungus, or parasite.
  • Mammalian cells suitable for carrying out the present invention include, among others COS, CHO, SaOS, osteosarcomas, KS483, MG-63, primary osteoblasts, and human or mammalian bone marrow stroma. Mammalian expression vectors for use in carrying out the present invention will include a promoter capable of directing the transcription of a cloned gene, nucleic acid molecule, or cDNA. Preferred promoters include viral promoters and cellular promoters. Bone specific promoters include the promoter for bone sialo-protein and the promoter for osteocalcin. Viral promoters include the cytomegalovirus immediate early promoter (Boshart et al., [0251] Cell 41:521-530, 1985), cytomegalovirus immediate late promoter, SV40 promoter (Subramani et al., Mol. Cell. Biol. 1:854-864, 1981), MMTV LTR, RSV LTR, metallothionein-1, adenovirus E1a. Cellular promoters include the mouse metallothionein-1 promoter (Palmiter et al., U.S. Pat. No. 4,579,821), a mouse Vκ promoter (Bergman et al., Proc. Natl Acad. Sci. USA 81:7041-7045, 1983; Grant et al., Nucleic Acids Res. 15:5496, 1987) and a mouse VH promoter (Loh et al., Cell 33:85-93, 1983). The choice of promoter will depend, at least in part, upon the level of expression desired or the recipient cell line to be transfected.
  • Such expression vectors may also contain a set of RNA splice sites located downstream from the promoter and upstream from the DNA sequence encoding the peptide or protein of interest. Preferred RNA splice sites may be obtained from adenovirus and/or immunoglobulin genes. Also contained in the expression vectors is a polyadenylation signal located downstream of the coding sequence of interest. Suitable polyadenylation signals include the early or late polyadenylation signals from SV40 (Kaufman and Sharp, ibid.), the polyadenylation signal from the [0252] Adenovirus 5 E1B region and the human growth hormone gene terminator (DeNoto et al., Nucleic Acids Res. 9:3719-3730, 1981). The expression vectors may include a noncoding viral leader sequence, such as the Adenovirus 2 tripartite leader, located between the promoter and the RNA splice sites. Preferred vectors may also include enhancer sequences, such as the SV40 enhancer. Expression vectors may also include sequences encoding the adenovirus VA RNAs. Suitable expression vectors can be obtained from commercial sources (e.g., Stratagene, La Jolla, Calif.).
  • Vector constructs comprising cloned DNA sequences can be introduced into cultured mammalian cells by, for example, calcium phosphate-mediated transfection (Wigler et al., [0253] Cell 14:725, 1978; Corsaro and Pearson, Somatic Cell Genetics 7:603, 1981; Graham and Van der Eb, Virology 52:456, 1973), electroporation (Neumann et al., EMBO J. 1:841-845, 1982), or DEAE-dextran mediated transfection (Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley and Sons, Inc., NY, 1987). To identify cells that have stably transfected with the vector or have integrated the cloned DNA, a selectable marker is generally introduced into the cells along with the gene or cDNA of interest. Preferred selectable markers for use in cultured mammalian cells include genes that confer resistance to drugs, such as neomycin, hygromycin, and methotrexate. The selectable marker may be an amplifiable selectable marker. Preferred amplifiable selectable markers are the DHFR gene and the neomycin resistance gene. Selectable markers are reviewed by Thilly (Mammalian Cell Technology, Butterworth Publishers, Stoneham, Mass., which is incorporated herein by reference).
  • Mammalian cells containing a suitable vector are allowed to grow for a period of time, typically 1-2 days, to begin expressing the DNA sequence(s) of interest. Drug selection is then applied to select for growth of cells that are expressing the selectable marker in a stable fashion. For cells that have been transfected with an amplifiable, selectable marker, the drug concentration may be increased in a stepwise manner to select for increased copy number of the cloned sequences, thereby increasing expression levels. Cells expressing the introduced sequences are selected and screened for production of the protein of interest in the desired form or at the desired level. Cells that satisfy these criteria can then be cloned and scaled up for production. [0254]
  • Protocols for the transfection of mammalian cells are well known to those of ordinary skill in the art. Representative methods include calcium phosphate mediated transfection, electroporation, lipofection, retroviral, adenoviral and protoplast fusion-mediated transfection (see Sambrook et al., supra). Naked vector constructs can also be taken up by muscular cells or other suitable cells subsequent to injection into the muscle of a mammal (or other animals). [0255]
  • Methods for using insect host cells and plant host cells for production of polypeptides are known in the art and described herein. Numerous insect host cells known in the art can also be useful within the present invention. For example, the use of baculoviruses as vectors for expressing heterologous DNA sequences in insect cells has been reviewed by Atkinson et al. ([0256] Pestic. Sci. 28:215-224,1990). Numerous vectors and plant host cells known in the art can also be useful within the present invention, for example, the use of Agrobacterium rhizogenes as vectors for expressing genes and nucleic acid molecules in plant cells (see review by Sinkar et al., J. Biosci. (Bangalore 11:47-58, 1987).
  • Within related aspects of the present invention, proteins of the present invention may be expressed in a transgenic animal whose germ cells and somatic cells contain a gene which encodes the desired protein and which is operably linked to a promoter effective for the expression of the gene. Alternatively, in a similar manner transgenic animals may be prepared that lack the desired gene (e.g., “knock-out” mice). Such transgenics may be prepared in a variety of non-human animals, including mice, rats, rabbits, sheep, dogs, goats, and pigs (see Hammer et al., [0257] Nature 315:680-683, 1985, Palmiter et al., Science 222:809-814, 1983, Brinster et al., Proc. Natl Acad. Sci. USA 82:4438-4442, 1985, Palmiter and Brinster, Cell 41:343-345, 1985, and U.S. Pat. Nos. 5,175,383, 5,087,571, 4,736,866, 5,387,742, 5,347,075, 5,221,778, and 5,175,384). Briefly, an expression vector, including a nucleic acid molecule to be expressed together with appropriately positioned expression control sequences, is introduced into pronuclei of fertilized eggs, for example, by microinjection. Integration of the injected DNA is detected by blot analysis of DNA from tissue samples. It is preferred that the introduced DNA be incorporated into the germ line of the animal so that it is passed on to the animal's progeny. Tissue-specific expression may be achieved through the use of a tissue-specific promoter, or through the use of an inducible promoter, such as the metallothionein gene promoter (Palmiter et al., 1983, supra), which allows regulated expression of the transgene.
  • Proteins can be isolated by, among other methods, culturing suitable host and vector systems to produce the recombinant translation products as described herein. Supernatants from such cell lines, or protein inclusions, or whole cells from which the protein is not excreted into the supernatant, can then be treated by a variety of purification procedures in order to isolate the desired proteins. For example, the supernatant may be first concentrated using commercially available protein concentration filters, such as an Amicon or Millipore Pellicon ultrafiltration unit. Following concentration, the concentrate may be applied to a suitable purification matrix such as, for example, an anti-protein antibody (e.g., an antibody that specifically binds to the polypeptide to be isolated) bound to a suitable support. Alternatively, anion or cation exchange resins may be employed in order to purify the protein. As a further alternative, one or more reverse-phase high performance liquid chromatography (RP-HPLC) steps may be employed to further purify the protein. Other methods of isolating the proteins of the present invention are well known in the art. [0258]
  • The purity of an isolated polypeptide may be determined by techniques known in the art and described herein, such as gel electrophoresis and chromatography methods. Preferably, such isolated polypeptides are at least about 90% pure, more preferably at least about 95% pure, and most preferably at least about 99% pure. Within certain specific embodiments, a protein is deemed to be “isolated” within the context of the present invention if no other undesired protein is detected pursuant to SDS-PAGE analysis followed by Coomassie blue staining. Within other embodiments, the desired protein can be isolated such that no other undesired protein is detected pursuant to SDS-PAGE analysis followed by silver staining. [0259]
  • 3. Nucleic Acid Molecules [0260]
  • Within other aspects of the invention, nucleic acid molecules are provided which are capable of inhibiting TGF-beta binding-protein binding to a member of the TGF-beta family. For example, within one embodiment antisense oligonucleotide molecules are provided which specifically inhibit expression of TGF-beta binding-protein nucleic acid sequences (see generally, Hirashima et al. in [0261] Molecular Biology of RNA: New Perspectives (M. Inouye and B. S. Dudock, eds., 1987 Academic Press, San Diego, p. 401); Oligonucleotides: Antisense Inhibitors of Gene Expression (J. S. Cohen, ed., 1989 MacMillan Press, London); Stein and Cheng, Science 261:1004-1012, 1993; WO 95/10607; U.S. Pat. No. 5,359,051; WO 92/06693; and EP-A2-612844). Briefly, such molecules are constructed such that they are complementary to, and able to form Watson-Crick base pairs with, a region of transcribed TGF-beta binding-protein mRNA sequence. The resultant double-stranded nucleic acid interferes with subsequent processing of the mRNA, thereby preventing protein synthesis (see Example 10).
  • Within other aspects of the invention, ribozymes are provided which are capable of inhibiting the TGF-beta binding-protein binding to a member of the TGF-beta family. As used herein, “ribozymes” are intended to include RNA molecules that contain anti-sense sequences -for specific recognition, and an RNA-cleaving enzymatic activity. The catalytic strand cleaves a specific site in a target RNA at greater than stoichiometric concentration. A wide variety of ribozymes may be utilized within the context of the present invention, including for example, the hammerhead ribozyme (for example, as described by Forster and Symons, [0262] Cell 48:211-220, 1987; Haseloff and Gerlach, Nature 328:596-600, 1988; Walbot and Bruening, Nature 334:196, 1988; Haseloff and Gerlach, Nature 334:585, 1988); the hairpin ribozyme (for example, as described by Haseloff et al., U.S. Pat. No. 5,254,678, issued Oct. 19, 1993 and Hempel et al., European Patent Publication No. 0 360 257, published Mar. 26, 1990); and Tetrahymena ribosomal RNA-based ribozymes (see Cech et al., U.S. Pat. No. 4,987,071). Ribozymes of the present invention typically consist of RNA, but may also be composed of DNA, nucleic acid analogs (e.g., phosphorothioates), or chimerics thereof (e.g., DNA/RNA/RNA).
  • 4. Labels [0263]
  • The gene product or any of the candidate molecules described above and below, may be labeled with a variety of compounds, including for example, fluorescent molecules, toxins, and radionuclides. Representative examples of fluorescent molecules include fluorescein, Phycobili proteins, such as phycoerythrin, rhodamine, Texas red and luciferase. Representative examples of toxins include ricin, abrin diphtheria toxin, cholera toxin, gelonin, pokeweed antiviral protein, tritin, Shigella toxin, and Pseudomonas exotoxin A. Representative examples of radionuclides include Cu-64, Ga-67, Ga-68, Zr-89, Ru-97, Tc-99m, Rh-105, Pd-109, In-111, I-123, I-125, I-131, Re-186, Re-188, Au-198, Au-199, Pb-203, At-211, Pb-212 and Bi-212. In addition, the antibodies described above may also be labeled or conjugated to one partner of a ligand binding pair. Representative examples include avidin-biotin, streptavidin-biotin, and riboflavin-riboflavin binding protein. [0264]
  • Methods for conjugating or labeling the molecules described herein with the representative labels set forth above may be readily accomplished by one of ordinary skill in the art (see Trichothecene Antibody Conjugate, U.S. Pat. No. 4,744,981; Antibody Conjugate, U.S. Pat. No. 5,106,951; Fluorogenic Materials and Labeling Techniques, U.S. Pat. No. 4,018,884; Metal Radionuclide Labeled Proteins for Diagnosis and Therapy, U.S. Pat. No. 4,897,255; and Metal Radionuclide Chelating Compounds for Improved Chelation Kinetics, U.S. Pat. No. 4,988,496; see also Inman, [0265] Methods In Enzymology, Vol. 34, Affinity Techniques, Enzyme Purification: Part B, Jakoby and Wilchek (eds.), Academic Press, New York, p. 30, 1974; see also Wilchek and Bayer, “The Avidin-Biotin Complex in Bioanalytical Applications,” Anal. Biochem. 171:1-32, 1988).
  • Pharmaceutical Compositions [0266]
  • As noted above, the present invention also provides a variety of pharmaceutical compositions, comprising one of the above-described molecules which inhibits the TGF-beta binding-protein binding to a member of the TGF-beta family along with a pharmaceutically or physiologically acceptable carrier, excipients or diluents. Generally, such carriers should be nontoxic to recipients at the dosages and concentrations employed. Ordinarily, the preparation of such compositions entails combining the therapeutic agent with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, maltose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients. Neutral buffered saline or saline mixed with nonspecific serum albumin are exemplary appropriate diluents. [0267]
  • The pharmaceutical compositions of the present invention may be prepared for administration by a variety of different routes. In general, the type of carrier is selected based on the mode of administration. Pharmaceutical compositions may be formulated for any appropriate manner of administration, including, for example, topical, oral, nasal, intrathecal, rectal, vaginal, sublingual or parenteral administration, including subcutaneous, intravenous, intramuscular, intrasternal, intracavernous, intrameatal, or intraurethral injection or infusion. A pharmaceutical composition (e.g., for oral administration or delivery by injection) may be in the form of a liquid (e.g., an elixir, syrup, solution, emulsion or suspension). A liquid pharmaceutical composition may include, for example, one or more of the following: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils that may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents; antioxidants; chelating agents; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. A parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. The use of physiological saline is preferred, and an injectable pharmaceutical composition is preferably sterile. [0268]
  • The compositions described herein may be formulated for sustained release (i.e., a formulation such as a capsule or sponge that effects a slow release of compound following administration). Such compositions may generally be prepared using well known technology and administered by, for example, oral, rectal or subcutaneous implantation, or by implantation at the desired target site. Sustained-release formulations may contain an agent dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Carriers for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release. The amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented. Illustrative carriers useful in this regard include microparticles of poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose, dextran and the like. Other illustrative delayed-release carriers include supramolecular biovectors, which comprise a non-liquid hydrophilic core (e.g., a cross-linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphilic compound, such as a phospholipid (see e.g., U.S. Pat. No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638). [0269]
  • In another illustrative embodiment, biodegradable microspheres (e.g., polylactate polyglycolate) are employed as carriers for the compositions of this invention. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344, 5,407,609 and 5,942,252. Modified hepatitis B core protein carrier systems, such as described in WO/99 40934, and references cited therein, will also be useful for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which are capable of inducing a class I-restricted cytotoxic T lymphocyte responses in a host. [0270]
  • In another illustrative embodiment, calcium phosphate core particles are employed as carriers or as controlled release matrices for the compositions of this invention. Exemplary calcium phosphate particles are disclosed, for example, in published patent application No. WO/0046147. [0271]
  • For pharmaceutical compositions comprising a polynucleotide encoding an anti-SOST antibody and/or modulating agent (such that the polypeptide and/or modulating agent is generated in situ), the polynucleotide may be present within any of a variety of delivery systems known to those of ordinary skill in the art, including nucleic acid, and bacterial, viral and mammalian expression systems. Techniques for incorporating DNA into such expression systems are well known to those of ordinary skill in the art. The DNA may also be “naked,” as described, for example, in Ulmer et al., [0272] Science 259:1745-1749, 1993 and reviewed by Cohen, Science 259:1691-1692, 1993. The uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.
  • The development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular administration and formulation, is well known in the art, some of which are briefly discussed below for general purposes of illustration. [0273]
  • In certain applications, the pharmaceutical compositions disclosed herein may be delivered via oral administration to an animal. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. [0274]
  • In certain circumstances it will be desirable to deliver the pharmaceutical compositions disclosed herein parenterally, intravenously, intramuscularly, or even intraperitoneally. Such approaches are well known to the skilled artisan, some of which are further described, for example, in U.S. Pat. No. 5,543,158; U.S. Pat. No. 5,641,515 and U.S. Pat. No. 5,399,363. In certain embodiments, solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally will contain a preservative to prevent the growth of microorganisms. [0275]
  • Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see U.S. Pat. No. 5,466,468). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants. The prevention of the action of microorganisms can be facilitated by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. [0276]
  • In one embodiment, for parenteral administration in an aqueous solution, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, [0277] Remington's Pharmaceutical Sciences, 15th ed., pp. 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. Moreover, for human administration, preparations will of course preferably meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologics standards.
  • In another embodiment of the invention, the compositions disclosed herein may be formulated in a neutral or salt form. Illustrative pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. [0278]
  • The carriers can further comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. The phrase “pharmaceutically-acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human. [0279]
  • In certain embodiments, liposomes, nanocapsules, microparticles, lipid particles, vesicles, and the like, are used for the introduction of the compositions of the present invention into suitable host cells/organisms. In particular, the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like. Alternatively, compositions of the present invention can be bound, either covalently or non-covalently, to the surface of such carrier vehicles. [0280]
  • The formation and use of liposome and liposome-like preparations as potential drug carriers is generally known to those of skill in the art (see for example, Lasic, [0281] Trends Biotechnol. 16(7):307-21, 1998; Takakura, Nippon Rinsho 56(3):691-95, 1998; Chandran et al., Indian J. Exp. Biol. 35(8):801-09, 1997; Margalit, Crit. Rev. Ther. Drug Carrier Syst. 12(2-3):233-61, 1995; U.S. Pat. No. 5,567,434; U.S. Pat. No. 5,552,157; U.S. Pat. No. 5,565,213; U.S. Pat. No. 5,738,868 and U.S. Pat. No. 5,795,587, each specifically incorporated herein by reference in its entirety).
  • Liposomes have been used successfully with a number of cell types that are normally difficult to transfect by other procedures, including T cell suspensions, primary hepatocyte cultures and PC 12 cells (Renneisen et al., [0282] J. Biol. Chem. 265(27):16337-42, 1990; Muller et al., DNA Cell Biol. 9(3):221-29, 1990). In addition, liposomes are free of the DNA length constraints that are typical of viral-based delivery systems. Liposomes have been used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals. Furthermore, he use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery.
  • In certain embodiments, liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles (MLVs). [0283]
  • Alternatively, in other embodiments, the invention provides for pharmaceutically-acceptable nanocapsule formulations of the compositions of the present invention. Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guerrero et al., [0284] Drug Dev. Ind. Pharm. 24(12):1113-28, 1998). To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 μm) may be designed using polymers able to be degraded in vivo. Such particles can be made as described, for example, by Couvreur et al., Crit. Rev. Ther. Drug Carrier Syst. 5(1):1-20, 1988; zur Muhlen et al., Eur. J. Pharm. Biopharm. 45(2):149-55, 1998; Zambaux et al., J. Controlled Release 50(1-3):31-40, 1998; and U.S. Pat. No. 5,145,684.
  • In addition, pharmaceutical compositions of the present invention may be placed within containers, along with packaging material that provides instructions regarding the use of such pharmaceutical compositions. Generally, such instructions will include a tangible expression describing the reagent concentration, as well as within certain embodiments, relative amounts of excipient ingredients or diluents (e.g., water, saline or PBS) which may be necessary to reconstitute the pharmaceutical composition. [0285]
  • Methods of Treatment [0286]
  • The present invention also provides methods for increasing the mineral content and mineral density of bone. Briefly, numerous conditions result in the loss of bone mineral content, including for example, disease, genetic predisposition, accidents which result in the lack of use of bone (e.g., due to fracture), therapeutics which effect bone resorption, or which kill bone forming cells and normal aging. Through use of the molecules described herein which inhibit the TGF-beta binding-protein binding to a TGF-beta family member such conditions may be treated or prevented. As utilized herein, it should be understood that bone mineral content has been increased if bone mineral content has been increased in a statistically significant manner (e.g., greater than one-half standard deviation), at a selected site. [0287]
  • A wide variety of conditions that result in loss of bone mineral content may be treated with the molecules described herein. Patients with such conditions may be identified through clinical diagnosis utilizing well known techniques (see, e.g., Harrison's Principles of Internal Medicine, McGraw-Hill, Inc.). Representative examples of diseases that may be treated included dysplasias, wherein there is abnormal growth or development of bone. Representative examples of such conditions include achondroplasia, cleidocranial dysostosis, enchondromatosis, fibrous dysplasia, Gaucher's Disease, hypophosphatemic rickets, Marfan's Syndrome, multiple hereditary exotoses, neurofibromatosis, osteogenesis imperfecta, osteopetrosis, osteopoikilosis, sclerotic lesions, fractures, periodontal disease, pseudoarthrosis, and pyogenic osteomyelitis. [0288]
  • Other conditions which may be treated or prevented include a wide variety of causes of osteopenia (i.e., a condition that causes greater than one standard deviation of bone mineral content or density below peak skeletal mineral content at youth). Representative examples of such conditions include anemic states, conditions caused by steroids, conditions caused by heparin, bone marrow disorders, scurvy, malnutrition, calcium deficiency, idiopathic osteoporosis, congenital osteopenia or osteoporosis, alcoholism, chronic liver disease, senility, postmenopausal state, oligomenorrhea, amenorrhea, pregnancy, diabetes mellitus, hyperthyroidism, Cushing's disease, acromegaly, hypogonadism, immobilization or disuse, reflex sympathetic dystrophy syndrome, transient regional osteoporosis, and osteomalacia. [0289]
  • Within one aspect of the present invention, bone mineral content or density may be increased by administering to a warm-blooded animal a therapeutically effective amount of a molecule that inhibits binding of the TGF-beta binding-protein to a TGF-beta family member. Examples of warm-blooded animals that may be treated include both vertebrates and mammals, including for example humans, horses, cows, pigs, sheep, dogs, cats, rats and mice. Representative examples of therapeutic molecules include ribozymes, ribozyme genes, antisense oligonucleotides, and antibodies (e.g., humanized antibodies or any other antibody described herein). [0290]
  • Within other aspects of the present invention, methods are provided for increasing bone density, comprising the steps of introducing into cells which home to bone, a vector that directs the expression of a molecule which inhibits binding of the TGF-beta binding-protein to a member of the TGF-beta family, and administering the vector-containing cells to a warm-blooded animal. Briefly, cells that home to bone may be obtained directly from the bone of patients (e.g., cells obtained from the bone marrow such as CD34+, osteoblasts, osteocytes, and the like), from peripheral blood, or from cultures. [0291]
  • A vector that directs the expression of a molecule that inhibits the binding of TGF-beta binding-protein to a member of the TGF-beta family may be introduced into cells. Representative examples of suitable vectors include viral vectors such as herpes viral vectors (e.g., U.S. Pat. No. 5,288,641), adenoviral vectors (e.g., WO 94/26914, WO 93/9191; Kolls et al., [0292] PNAS 91(1):215-219, 1994; Kass-Eisler et al., PNAS 90(24):11498-502, 1993; Guzman et al., Circulation 88(6):2838-48, 1993; Guzman et al., Cir. Res. 73(6):1202-1207, 1993; Zabner et al., Cell 75(2):207-216, 1993; Li et al., Hum Gene Ther. 4(4):403-409, 1993; Caillaud et al., Eur. J. Neurosci. 5(10:1287-1291, 1993; Vincent et al., Nat. Genet. 5(2):130-134, 1993; Jaffe et al., Nat. Genet. 1(5):372-378, 1992; and Levrero et al., Gene 101(2):195-202, 1991), adeno-associated viral vectors (WO 95/13365; Flotte et al., PNAS 90(22):10613-10617, 1993), baculovirus vectors, parvovirus vectors (Koering et al., Hum. Gene Therap. 5:457-463, 1994), pox virus vectors (Panicali and Paoletti, PNAS 79:4927-4931, 1982; and Ozaki et al., Biochem. Biophys. Res. Comm. 193(2):653-660, 1993), and retroviruses (e.g., EP 0,415,731; WO 90/07936; WO 91/0285, WO 94/03622; WO 93/25698; WO 93/25234; U.S. Pat. No. 5,219,740; WO 93/11230; WO 93/10218). Viral vectors may likewise be constructed which contain a mixture of different elements (e.g., promoters, envelope sequences, and the like) from different viruses, or non-viral sources. Within various embodiments, either the viral vector itself, or a viral particle which contains the viral vector may be utilized in the methods and compositions described below. Within other embodiments of the invention, nucleic acid molecules which encode a molecule which inhibits binding of the TGF-beta binding-protein to a member of the TGF-beta family may be administered by a variety of techniques, including, for example, administration of asialoosomucoid (ASOR) conjugated with poly-L-lysine DNA complexes (Cristano et al., PNAS 92122-92126, 1993), DNA linked to killed adenovirus (Curiel et al., Hum. Gene Ther. 3(2):147-154, 1992), cytofectin-mediated introduction (DMRIE-DOPE, Vical, Calif.), direct DNA injection (Acsadi et al., Nature 352:815-818, 1991); DNA ligand (Wu et al., J. of Biol. Chem. 264:16985-16987, 1989); lipofection (Felgner et al., Proc. Natl. Acad. Sci. USA 84:7413-7417, 1989); liposomes (Pickering et al., Circ. 89(l):13-21, 1994; and Wang et al., PNAS 84:7851-7855, 1987); microprojectile bombardment (Williams et al., PNAS 88:2726-2730, 1991); and direct delivery of nucleic acids which encode the protein itself either alone (Vile and Hart, Cancer Res. 53: 3860-3864, 1993), or utilizing PEG-nucleic acid complexes. Representative examples of molecules that may be expressed by the vectors of present invention include ribozymes and antisense molecules, each of which are discussed in more detail above.
  • Determination of increased bone mineral content may be determined directly through the use of X-rays (e.g., Dual Energy X-ray Absorptometry or “DEXA”), or by inference through bone turnover markers (such as osteoblast specific alkaline phosphatase, osteocalcin, [0293] type 1 procollagen C′ propeptide (PICP), and total alkaline phosphatase; see Comier, C., Curr. Opin. in Rheu. 7:243, 1995), or by markers of bone resorption (pyridinoline, deoxypryridinoline, N-telopeptide, urinary hydroxyproline, plasma tartrate-resistant acid phosphatases and galactosyl hydroxylysine; see Comier, supra). The amount of bone mass may also be calculated from body weights or by other methods known in the art (see Guinness-Hey, Metab. Bone Dis. and Relat. Res. 5:177-181, 1984).
  • As will be evident to one of skill in the art, the amount and frequency of administration will depend, of course, on such factors as the nature and severity of the indication being treated, the desired response, the condition of the patient, and so forth. Typically, the compositions may be administered by a variety of techniques, as noted above. [0294]
  • The following examples are offered by way of illustration and not by way of limitation. [0295]
  • EXAMPLES Example 1
  • Sclerosteosis Maps to the Long Arm of Human Chromosome 17 [0296]
  • Genetic mapping of the defect responsible for sclerosteosis in humans localized the gene responsible for this disorder to the region of human chromosome 17 that encodes a novel TGF-beta binding-protein family member. In sclerosteosis, skeletal bone displays a substantial increase in mineral density relative to that of unafflicted individuals. Bone in the head displays overgrowth as well. Sclerosteosis patients are generally healthy although they may exhibit variable degrees of syndactyly at birth and variable degrees of cranial compression and nerve compression in the skull. [0297]
  • Linkage analysis of the gene defect associated with sclerosteosis was conducted by applying the homozygosity mapping method to DNA samples collected from 24 South African Afrikaaner families in which the disease occurred. (Sheffield et al., 1994, [0298] Human Molecular Genetics 3:1331-1335. “Identification of a Bardet-Biedl syndrome locus on chromosome 3 and evaluation of an efficient approach to homozygosity mapping”). The Afrikaaner population of South Africa is genetically homogeneous; the population is descended from a small number of founders who colonized the area several centuries ago, and it has been isolated by geographic and social barriers since the founding. Sclerosteosis is rare everywhere in the world outside the Afrikaaner community, which suggests that a mutation in the gene was present in the founding population and has since increased in numbers along with the increase in the population. The use of homozygosity mapping is based on the assumption that DNA mapping markers adjacent to a recessive mutation are likely to be homozygous in affected individuals from consanguineous families and isolated populations.
  • A set of 371 microsatellite markers (Research Genetics, Set 6) from the autosomal chromosomes was selected to type pools of DNA from sclerosteosis patient samples. The DNA samples for this analysis came from 29 sclerosteosis patients in 24 families, 59 unaffected family members and a set of unrelated control individuals from the same population. The pools consisted of 4-6 individuals, either affected individuals, affected individuals from consanguineous families, parents and unaffected siblings, or unrelated controls. In the pools of unrelated individuals and in most of the pools with affected individuals or family members analysis of the markers showed several allele sizes for each marker. One marker, D17S1299, showed an indication of homozygosity: one band in several of the pools of affected individuals. [0299]
  • All 24 sclerosteosis families were typed with a total of 19 markers in the region of D17S1299 (at 17q12-q21). Affected individuals from every family were shown to be homozygous in this region, and 25 of the 29 individuals were homozygous for a core haplotype; they each had the same alleles between D17S1787 and D17S930. The other four individuals had one chromosome which matched this haplotype and a second which did not. In sum, the data compellingly suggested that this 3 megabase region contained the sclerosteosis mutation. Sequence analysis of most of the exons in this 3 megabase region identified a nonsense mutation in the novel TGF-beta binding-protein coding sequence (C>T mutation at position 117 of Sequence ID No. 1 results in a stop codon). This mutation was shown to be unique to sclerosteosis patients and carriers of Afrikaaner descent. The identity of the gene was further confirmed by identifying a mutation in its intron (A>T mutation at position +3 of the intron) which results in improper mRNA processing in a single, unrelated patient with diagnosed sclerosteosis. [0300]
  • Example 2
  • Tissue-Specificity of TGF-Beta Binding-Protein Expression [0301]
  • A. Human Beer Gene Expression by RT-PCR: [0302]
  • First-strand cDNA was prepared from the following total RNA samples using a commercially available kit (“Superscript Preamplification System for First-Strand cDNA Synthesis”, Life Technologies, Rockville, Md.): human brain, human liver, human spleen, human thymus, human placenta, human skeletal muscle, human thyroid, human pituitary, human osteoblast (NHOst from Clonetics Corp., San Diego, Calif.), human osteosarcoma cell line (Saos-2, ATCC# HTB-85), human bone, human bone marrow, human cartilage, vervet monkey bone, saccharomyces cerevisiae, and human peripheral blood monocytes. All RNA samples were purchased from a commercial source (Clontech, Palo Alto, Calif.), except the following which were prepared in-house: human osteoblast, human osteosarcoma cell line, human bone, human cartilage and vervet monkey bone. These in-house RNA samples were prepared using a commercially available kit (“TRI Reagent”, Molecular Research Center, Inc., Cincinnati, Ohio). [0303]
  • PCR was performed on these samples, and additionally on a human genomic sample as a control. The sense Beer oligonucleotide primer had the [0304] sequence 5′-CCGGAGCTGGAGAACAACAAG-3′ (SEQ ID NO: 19). The antisense Beer oligonucleotide primer had the sequence 5′-GCACTGGCCGGAGCACACC-3′ (SEQ ID NO: 20). In addition, PCR was performed using primers for the human beta-actin gene, as a control. The sense beta-actin oligonucleotide primer had the sequence 5′-AGGCCAACCGCGAGAAGATGA CC-3′ (SEQ ID NO: 21). The antisense beta-actin oligonucleotide primer had the sequence 5′-GAAGT CCAGGGCGACGTAGCA-3′ (SEQ ID NO: 22). PCR was performed using standard conditions in 25 ul reactions, with an annealing temperature of 61 degrees Celsius. Thirty-two cycles of PCR were performed with the Beer primers and twenty-four cycles were performed with the beta-actin primers.
  • Following amplification, 12 ul from each reaction were analyzed by agarose gel electrophoresis and ethidium bromide staining. See FIG. 2A. [0305]
  • B. RNA In-situ Hybridization of Mouse Embryo Sections: [0306]
  • The full length mouse Beer cDNA (Sequence ID No. 11) was cloned into the pCR2.1 vector (Invitrogen, Carlsbad, Calif.) in the antisense and sense direction using the manufacturer's protocol. [0307] 35S-alpha-GTP-labeled cRNA sense and antisense transcripts were synthesized using in-vitro transcription reagents supplied by Ambion, Inc (Austin, Tex.). In-situ hybridization was performed according to the protocols of Lyons et al. (J. Cell Biol. 111:2427-2436, 1990).
  • The mouse Beer cRNA probe detected a specific message expressed in the neural tube, limb buds, blood vessels and ossifying cartilages of developing mouse embryos. Panel A in FIG. 3 shows expression in the apical ectodermal ridge (aer) of the limb (l) bud, blood vessels (bv) and the neural tube (nt). Panel B shows expression in the 4[0308] th ventricle of the brain (4). Panel C shows expression in the mandible (ma) cervical vertebrae (cv), occipital bone (oc), palate (pa) and a blood vessel (bv). Panel D shows expression in the ribs (r) and a heart valve (va). Panel A is a transverse section of 10.5 dpc embryo. Panel B is a sagittal section of 12.5 dpc embryo and panels C and D are sagittal sections of 15.5 dpc embryos.
  • ba=branchial arch, h=heart, te=telencephalon (forebrain), b=brain, f=frontonasal mass, g=gut, h=heart, j=jaw, li=liver, lu=lung, ot=otic vesicle, ao=, sc=spinal cord, skm=skeletal muscle, ns=nasal sinus, th=thymus, to=tongue, fl=forelimb, di=diaphragm [0309]
  • Example 3
  • Expression and Purification of Recombinant Beer Protein [0310]
  • A. Expression in COS-1 Cells: [0311]
  • The DNA sequence encoding the full length human Beer protein was amplified using the following PCR oligonucleotide primers: The 5′ oligonucleotide primer had the [0312] sequence 5′-AAGCTTGGTACCATGCAGCTCCCAC-3′ (SEQ ID NO: 23) and contained a HindIII restriction enzyme site (in bold) followed by 19 nucleotides of the Beer gene starting 6 base pairs prior to the presumed amino terminal start codon (ATG). The 3′ oligonucleotide primer had the sequence 5′-AAGCTTCTACTTGTCATCGTCGTCCT TGTAGTCGTAGGCGTTCTCCAGCT-3′ (SEQ ID NO: 24) and contained a HindIII restriction enzyme site (in bold) followed by a reverse complement stop codon (CTA) followed by the reverse complement of the FLAG epitope (underlined, Sigma-Aldrich Co., St. Louis, Mo.) flanked by the reverse complement of nucleotides coding for the carboxy terminal 5 amino acids of the Beer. The PCR product was TA cloned (“Original TA Cloning Kit”, Invitrogen, Carlsbad, Calif.) and individual clones were screened by DNA sequencing. A sequence-verified clone was then digested by HindIII and purified on a 1.5% agarose gel using a commercially available reagents (“QIAquick Gel Extraction Kit”, Qiagen Inc., Valencia, Calif.). This fragment was then ligated to HindIII digested, phosphatase-treated pcDNA3.1 (Invitrogen, Carlsbad, Calif.) plasmid with T4 DNA ligase. DH10B E. coli were transformed and plated on LB, 100 μg/ml ampicillin plates. Colonies bearing the desired recombinant in the proper orientation were identified by a PCR-based screen, using a 5′ primer corresponding to the T7 promoter/priming site in pcDNA3.1 and a 3′ primer with the sequence 5′-GCACTGGCCGGAGCACACC-3′ (SEQ ID NO: 25) that corresponds to the reverse complement of internal BEER sequence. The sequence of the cloned fragment was confirmed by DNA sequencing.
  • COS-1 cells (ATCC# CRL-1650) were used for transfection. 50 μg of the expression plasmid pcDNA-Beer-Flag was transfected using a commercially available kit following protocols supplied by the manufacturer (“DEAE-Dextran Transfection Kit”, Sigma Chemical Co., St. Louis, Mo.). The final media following transfection was DMEM (Life Technologies, Rockville, Md.) containing 0.1% Fetal Bovine Serum. After 4 days in culture, the media was removed. Expression of recombinant BEER was analyzed by SDS-PAGE and Western Blot using anti-FLAG® M2 monoclonal antibody (Sigma-Aldrich Co., St. Louis, Mo.). Purification of recombinant BEER protein was performed using an anti-FLAG M2 affinity column (“Mammalian Transient Expression System”, Sigma-Aldrich Co., St. Louis, Mo.). The column profile was analyzed via SDS-PAGE and Western Blot using anti-FLAG M2 monoclonal antibody. [0313]
  • B. Expression in SF9 Insect Cells: [0314]
  • The human Beer gene sequence was amplified using PCR with standard conditions and the following primers: [0315]
    Sense primer: 5′-GTCGTCGGATCCATGGGGTGGCAGGCGTTCAAGAATGAT-3′ (SEQ ID NO:26)
    Antisense primer: 5′-GTCGTCAAGCTTCTACTTGTCATCGTCCTTGTAGTCGTAGGCGTTCTCCAGCTCGGC-3′ (SEQ ID NO:27)
  • The resulting cDNA contained the coding region of Beer with two modifications. The N-terminal secretion signal was removed and a FLAG epitope tag (Sigma) was fused in frame to the C-terminal end of the insert. BamH1 and HindIII cloning sites were added and the gene was subcloned into pMelBac vector (Invitrogen) for transfer into a baculoviral expression vector using standard methods. [0316]
  • Recombinant baculoviruses expressing Beer protein were made using the Bac-N-Blue transfection kit (Invitrogen) and purified according to the manufacturers instructions. [0317]
  • SF9 cells (Invitrogen) were maintained in TNM_FH media (Invitrogen) containing 10% fetal calf serum. For protein expression, SF9 cultures in spinner flasks were infected at an MOI of greater than 10. Samples of the media and cells were taken daily for five days, and Beer expression monitored by western blot using an anti-FLAG M2 monoclonal antibody (Sigma) or an anti-Beer rabbit polyclonal antiserum. [0318]
  • After five days the baculovirus-infected SF9 cells were harvested by centrifugation and cell associated protein was extracted from the cell pellet using a high salt extraction buffer (1.5 M NaCl, 50 mM Tris pH 7.5). The extract (20 ml per 300 ml culture) was clarified by centrifugation, dialyzed three times against four liters of Tris buffered saline (150 mM NaCl, 50 mM Tris pH 7.5), and clarified by centrifugation again. This high salt fraction was applied to Hitrap Heparin (Pharmacia; 5 ml bed volume), washed extensively with HEPES buffered saline (25 mM HEPES 7.5, 150 mM Nacl) and bound proteins were eluted with a gradient from 150 mM NaCl to 1200 mM NaCl. Beer elution was observed at aproximately 800 mM NaCl. Beer containing fractions were supplemented to 10% glycerol and 1 mM DTT and frozen at −80 degrees C. [0319]
  • Example 4
  • Preparation and Testing of Polyclonal Antibodies to Beer, Gremlin, and Dan [0320]
  • A. Preparation of Antigen: [0321]
  • The DNA sequences of Human Beer, Human Gremlin, and Human Dan were amplified using standard PCR methods with the following oligonucleotide primers: [0322]
    H. Beer
    Sense: (SEQ ID NO:28)
    5′-GACTTGGATCCCAGGGGTGGCAGGCGTTC-3′
    Antisense (SEQ ID NO:29)
    5′-AGCATAAGCTTCTAGTAGGCGTTCTCCAG-3′
    H. Gremlin
    Sense: (SEQ ID NO:30)
    5′-GACTTGGATCCGAAGGGAAAAAGAAAGGG-3′
    Antisense: (SEQ ID NO:31)
    5′-AGCATAAGCTTTTAATCCAAATCGATGGA-3′
    H. Dan
    Sense: (SEQ ID NO:32)
    5′-ACTACGAGCTCGGCCCCACCACCCATCAACAAG-3′
    Antisense: (SEQ ID NO:33)
    5′-ACTTAGAAGCTTTCAGTCCTCAGCCCCCTCTTCC-3′
  • In each case the listed primers amplified the entire coding region minus the secretion signal sequence. These include restriction sites for subcloning into the bacterial expression vector pQE-30 (Qiagen Inc., Valencia, Calif.) at sites BamHI/HindIII for Beer and Gremlin, and sites SacI/HindIII for Dan. pQE30 contains a coding sequence for a 6× His tag at the 5′ end of the cloning region. The completed constructs were transformed into [0323] E. coli strain M-15/pRep (Qiagen Inc) and individual clones verified by sequencing. Protein expression in M-15/pRep and purification (6× His affinity tag binding to Ni-NTA coupled to Sepharose) were performed as described by the manufacturer (Qiagen, The QIAexpressionist).
  • The [0324] E. coli-derived Beer protein was recovered in significant quantity using solubilization in 6M guanidine and dialyzed to 2-4M to prevent precipitation during storage. Gremlin and Dan protein were recovered in higher quantity with solubilization in 6M guanidine and a post purification guanidine concentration of 0.5M.
  • B. Production and Testing of Polyclonal Antibodies: [0325]
  • Polyclonal antibodies to each of the three antigens were produced in rabbit and in chicken hosts using standard protocols (R & R Antibody, Stanwood, Wash.; standard protocol for rabbit immunization and antisera recovery; Short Protocols in Molecular Biology. 2nd edition. 1992. 11.37-11.41. Contributors Helen M. Cooper and Yvonne Paterson; chicken antisera was generated with Strategic Biosolutions, Ramona, Calif.). [0326]
  • Rabbit antisera and chicken egg Igy fraction were screened for activity via Western blot. Each of the three antigens was separated by PAGE and transferred to 0.45 um nitrocellulose (Novex, San Diego, Calif.). The membrane was cut into strips with each strip containing approximately 75 ng of antigen. The strips were blocked in 3% Blotting Grade Block (Bio-Rad Laboratories, Hercules, Calif.) and washed 3 times in 1× Tris buffer saline (TBS)/0.02% TWEEN buffer. The primary antibody (preimmunization bleeds, rabbit antisera or chicken egg IgY in dilutions ranging from 1:100 to 1:10,000 in blocking buffer) was incubated with the strips for one hour with gentle rocking. A second series of three [0327] washes 1× TBS/0.02% TWEEN was followed by an one hour incubation with the secondary antibody (peroxidase conjugated donkey anti-rabbit, Amersham Life Science, Piscataway, N.J.; or peroxidase conjugated donkey anti-chicken, Jackson ImmunoResearch, West Grove, Pa.). A final cycle of 3× washes of 1× TBS/0.02% TWEEN was performed and the strips were developed with Lumi-Light Western Blotting Substrate (Roche Molecular Biochemicals, Mannheim, Germany).
  • C. Antibody Cross-Reactivity Test: [0328]
  • Following the protocol described in the previous section, nitrocellulose strips of Beer, Gremlin or Dan were incubated with dilutions (1:5000 and 1:10,000) of their respective rabbit antisera or chicken egg IgY as well as to antisera or chicken egg Igy (dilutions 1:1000 and 1:5000) made to the remaining two antigens. The increased levels of nonmatching antibodies was performed to detect low affinity binding by those antibodies that may be seen only at increased concentration. The protocol and duration of development is the same for all three binding events using the protocol described above. There was no antigen cross-reactivity observed for any of the antigens tested. [0329]
  • Example 5
  • Interaction of Beer with TGF-Beta Super-Family Proteins [0330]
  • The interaction of Beer with proteins from different phylogenetic arms of the TGF-β superfamily were studied using immunoprecipitation methods. Purified TGFβ-1, TGFβ-2, TGFβ-3, BMP-4, BMP-5, BMP-6 and GDNF were obtained from commerical sources (R&D systems; Minneapolis, Minn.). A representative protocol is as follows. Partially purified Beer was dialyzed into HEPES buffered saline (25 mM HEPES 7.5, 150 mM NaCl). Immunoprecipitations were done in 300 ul of IP buffer (150 mM NaCl, 25 mM Tris pH 7.5, 1 mM EDTA, 1.4 mM β-mercaptoethanol, 0.5 % triton ×100, and 10% glycerol). 30 ng recombinant human BMP-5 protein (R&D systems) was applied to 15 ul of FLAG affinity matrix (Sigma; St Louis Mo.)) in the presence and absence of 500 ng FLAG epitope-tagged Beer. The proteins were incubated for 4 hours@ 4° C. and then the affinity matrix-associated proteins were washed 5 times in IP buffer (1 ml per wash). The bound proteins were eluted from the affinity matrix in 60 microliters of 1× SDS PAGE sample buffer. The proteins were resolved by SDS PAGE and Beer associated BMP-5 was detected by western blot using anti-BMP-5 antiserum (Research Diagnostics, Inc) (see FIG. 5). [0331]
  • BEER Ligand Binding Assay: [0332]
  • FLAG-Beer protein (20 ng) is added to 100 ul PBS/0.2% BSA and adsorbed into each well of 96 well microtiter plate previously coated with anti-FLAG monoclonal antibody (Sigma; St Louis Mo.) and blocked with 10% BSA in PBS. This is conducted at room temperature for 60 minutes. This protein solution is removed and the wells are washed to remove unbound protein. BMP-5 is added to each well in concentrations ranging from 10 pM to 500 nM in PBS/0.2% BSA and incubated for 2 hours at room temperature. The binding solution is removed and the plate washed with three times with 200 ul volumes of PBS/0.2% BSA. BMP-5 levels are then detected using BMP-5 anti-serum via ELISA (F. M. Ausubel et al (1998) Current Protocols in Mol Biol. Vol 2 11.2.1-11.2.22). Specific binding is calculated by subtracting non-specific binding from total binding and analyzed by the LIGAND program (Munson and Podbard, Anal. Biochem., 107, p220-239, (1980). [0333]
  • In a variation of this method, Beer is engineered and expressed as a human Fc fusion protein. Likewise the ligand BMP is engineered and expressed as mouse Fc fusion. These proteins are incubated together and the assay conducted as described by Mellor et al using homogeneous time resolved fluorescence detection (G. W. Mellor et al., [0334] J of Biomol Screening, 3(2) 91-99, 1998).
  • Example 6
  • Screening Assay for Inhibition of TGF-Beta Binding-Protein Binding to TGF-Beta Family Members [0335]
  • The assay described above is replicated with two exceptions. First, BMP concentration is held fixed at the Kd determined previously. Second, a collection of antagonist candidates is added at a fixed concentration (20 uM in the case of the small organic molecule collections and 1 uM in antibody studies). These candidate molecules (antagonists) of TGF-beta binding-protein binding include organic compounds derived from commercial or internal collections representing diverse chemical structures. These compounds are prepared as stock solutions in DMSO and are added to assay wells at ≦1% of final volume under the standard assay conditions. These are incubated for 2 hours at room temperature with the BMP and Beer, the solution removed and the bound BMP is quantitated as described. Agents that inhibit 40% of the BMP binding observed in the absence of compound or antibody are considered antagonists of this interaction. These are further evaluated as potential inhibitors based on titration studies to determine their inhibition constants and their influence on TGF-beta binding-protein binding affinity. Comparable specificity control assays may also be conducted to establish the selectivity profile for the identified antagonist through studies using assays dependent on the BMP ligand action (e.g. BMP/BMP receptor competition study). [0336]
  • Example 7
  • Inhibition of TGF-Beta Binding-Protein Localization to Bone Matrix [0337]
  • Evaluation of inhibition of localization to bone matrix (hydroxyapatite) is conducted using modifications to the method of Nicolas (Nicolas, V. [0338] Calcif Tissue Int 57:206, 1995). Briefly, 125I-labelled TGF-beta binding-protein is prepared as described by Nicolas (supra). Hydroxyapatite is added to each well of a 96 well microtiter plate equipped with a polypropylene filtration membrane (Polyfiltroninc, Weymouth Mass.). TGF-beta binding-protein is added to 0.2% albumin in PBS buffer. The wells containing matrix are washed 3 times with this buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3M NaOH and quantitated.
  • Inhibitor identification is conducted via incubation of TGF-beta binding-protein with test molecules and applying the mixture to the matrix as described above. The matrix is washed 3 times with 0.2% albumin in PBS buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3 M NaOH and quantitated. Agents that inhibit 40% of the TGF-beta binding-protein binding observed in the absence of compound or antibody are considered bone localization inhibitors. These inhibitors are further characterized through dose response studies to determine their inhibition constants and their influence on TGF-beta binding-protein binding affinity. [0339]
  • Example 8
  • Construction of TGF-Beta Binding-Protein Mutant [0340]
  • A. Mutagenesis: [0341]
  • A full-length TGF-beta binding-protein cDNA in pBluescript SK serves as a template for mutagenesis. Briefly, appropriate primers (see the discussion provided above) are utilized to generate the DNA fragment by polymerase chain reaction using Vent DNA polymerase (New England Biolabs, Beverly, Mass.). The polymerase chain reaction is run for 23 cycles in buffers provided by the manufacturer using a 57° C. annealing temperature. The product is then exposed to two restriction enzymes and after isolation using agarose gel electrophoresis, ligated back into pRBP4-503 from which the matching sequence has been removed by enzymatic digestion. Integrity of the mutant is verified by DNA sequencing. [0342]
  • B. Mammalian Cell Expression and Isolation of Mutant TGF-Beta Binding-Protein: [0343]
  • The mutant TGF-beta binding-protein cDNAs are transferred into the pcDNA3.1 mammalian expression vector described in EXAMPLE 3. After verifying the sequence, the resultant constructs are transfected into COS-1 cells, and secreted protein is purified as described in EXAMPLE 3. [0344]
  • Example 9
  • Animal Models-I Generation of Transgenic Mice Overexpressing the Beer Gene [0345]
  • The ˜200 kilobase (kb) BAC clone 15G5, isolated from the CITB mouse genomic DNA library (distributed by Research Genetics, Huntsville, Ala.) was used to determine the complete sequence of the mouse Beer gene and its 5′ and 3′ flanking regions. A 41 kb SalI fragment, containing the entire gene body, plus ˜17 kb of 5′ flanking and ˜20 kb of 3′ flanking sequence was sub-cloned into the BamHI site of the SuperCosI cosmid vector (Stratagene, La Jolla, Calif.) and propagated in the [0346] E. coli strain DH10B. From this cosmid construct, a 35 kb MluI—AviII restriction fragment (Sequence No. 6), including the entire mouse Beer gene, as well as 17 kb and 14 kb of 5′ and 3′ flanking sequence, respectively, was then gel purified, using conventional means, and used for microinjection of mouse zygotes (DNX Transgenics; U.S. Pat. No. 4,873,191). Founder animals in which the cloned DNA fragment was integrated randomly into the genome were obtained at a frequency of 5-30% of live-born pups. The presence of the transgene was ascertained by performing Southern blot analysis of genomic DNA extracted from a small amount of mouse tissue, such as the tip of a tail. DNA was extracted using the following protocol: tissue was digested overnight at 55° C. in a lysis buffer containing 200 mM NaCl, 100 mM Tris pH8.5, 5 mM EDTA, 0.2% SDS and 0.5 mg/ml Proteinase K. The following day, the DNA was extracted once with phenol/chloroform (50:50), once with chloroform/isoamylalcohol (24:1) and precipitated with ethanol. Upon resuspension in TE (10 mM Tris pH7.5, 1 mM EDTA) 8-10 ug of each DNA sample were digested with a restriction endonuclease, such as EcoRI, subjected to gel electrophoresis and transferred to a charged nylon membrane, such as HyBondN+ (Amersham, Arlington Heights, Ill.). The resulting filter was then hybridized with a radioactively labelled fragment of DNA deriving from the mouse Beer gene locus, and able to recognize both a fragment from the endogenous gene locus and a fragment of a different size deriving from the transgene. Founder animals were bred to normal non-transgenic mice to generate sufficient numbers of transgenic and non-transgenic progeny in which to determine the effects of Beer gene overexpression. For these studies, animals at various ages (for example, 1 day, 3 weeks, 6 weeks, 4 months) are subjected to a number of different assays designed to ascertain gross skeletal formation, bone mineral density, bone mineral content, osteoclast and osteoblast activity, extent of endochondral ossification, cartilage formation, etc. The transcriptional activity from the transgene may be determined by extracting RNA from various tissues, and using an RT-PCR assay which takes advantage of single nucleotide polymorphisms between the mouse strain from which the transgene is derived (129 Sv/J) and the strain of mice used for DNA microinjection [(C57BL5/J×SJL/J)F2].
  • Animal Models—II [0347]
  • Disruption of the Mouse Beer Gene By Homologous Recombination [0348]
  • Homologous recombination in embryonic stem (ES) cells can be used to inactivate the endogenous mouse Beer gene and subsequently generate animals carrying the loss-of-function mutation. A reporter gene, such as the [0349] E. coli β-galactosidase gene, was engineered into the targeting vector so that its expression is controlled by the endogenous Beer gene's promoter and translational initiation signal. In this way, the spatial and temporal patterns of Beer gene expression can be determined in animals carrying a targeted allele.
  • The targeting vector was constructed by first cloning the drug-selectable phosphoglycerate kinase (PGK) promoter driven neomycin-resistance gene (neo) cassette from pGT-N29 (New England Biolabs, Beverly, Mass.) into the cloning vector pSP72 (Promega, Madson, Wis.). PCR was used to flank the PGKneo cassette with bacteriophage P1 loxP sites, which are recognition sites for the P1 Cre recombinase (Hoess et al., PNAS USA, 79:3398, 1982). This allows subsequent removal of the neo-resistance marker in targeted ES cells or ES cell-derived animals (U.S. Pat. No. 4,959,317). The PCR primers were comprised of the 34 nucleotide (ntd) loxP sequence, 15-25 ntd complementary to the 5′ and 3′ ends of the PGKneo cassette, as well as restriction enzyme recognition sites (BamHI in the sense primer and EcoRI in the anti-sense primer) for cloning into pSP72. The sequence of the sense primer was 5′-AATCTGGATCCATAACTTCGTATAGCATACATTATACGAAGTTATCTGCAG GATTCGAGGGCCCCT-3′ (SEQ ID NO: 34); sequence of the anti-sense primer was 5′-AATCTGAATTCCACCGGTGTTAATTAAATAACTTCGT ATAATGTATGCTATACGAAGTTATAGATCTAGAG TCAGCTTCTGA-3′ (SEQ ID NO: 35). [0350]
  • The next step was to clone a 3.6 kb XhoI-HindIII fragment, containing the [0351] E. coli β-galactosidase gene and SV40 polyadenylation signal from pSVβ (Clontech, Palo Alto, Calif.) into the pSP72-PGKneo plasmid. The “short arm” of homology from the mouse Beer gene locus was generated by amplifying a 2.4 kb fragment from the BAC clone 15G5. The 3′ end of the fragment coincided with the translational initiation site of the Beer gene, and the anti-sense primer used in the PCR also included 30 ntd complementary to the 5′ end of the β-galactosidase gene so that its coding region could be fused to the Beer initiation site in-frame. The approach taken for introducing the “short arm” into the pSP72-βgal-PGKneo plasmid was to linearize the plasmid at a site upstream of the β-gal gene and then to co-transform this fragment with the “short arm” PCR product and to select for plasmids in which the PCR product was integrated by homologous recombination. The sense primer for the “short arm” amplification included 30 ntd complementary to the pSP72 vector to allow for this recombination event. The sequence of the sense primer was 5′-ATTTAGGTGACACT ATAGAACTCGAGCAGCTGAAGCTTAACCACATGGTGGCTCACAACCAT-3′ (SEQ ID NO: 36) and the sequence of the anti-sense primer was 5′-AACGACGGCCAGTGAATCCGTA ATCATGGTCATGCTGCCAGGTGGAGGAGGGCA-3′ (SEQ ID NO: 37).
  • The “long arm” from the Beer gene locus was generated by amplifying a 6.1 kb fragment from BAC clone 15G5 with primers which also introduce the rare-cutting restriction enzyme sites SgrAI, FseI, AscI and PacI. Specifically, the sequence of the sense primer was 5′-ATTACCACCGGTGACACCCGCTTCCTGACAG-3′ (SEQ ID NO: 38); the sequence of the anti-sense primer was 5′-ATTACTTAATTAAACATGGCGCGCCAT ATGGCCGGCCCCTAATTGCGGCGCATCGTTAATT-3′ (SEQ ID NO: 39). The resulting PCR product was cloned into the TA vector (Invitrogen, Carlsbad, Calif.) as an intermediate step. [0352]
  • The mouse Beer gene targeting construct also included a second selectable marker, the herpes simplex virus I thymidine kinase gene (HSVTK) under the control of rous sarcoma virus long terminal repeat element (RSV LTR). Expression of this gene renders mammalian cells sensitive (and inviable) to gancyclovir; it is therefore a convenient way to select against neomycin-resistant cells in which the construct has integrated by a non-homologous event (U.S. Pat. No. 5,464,764). The RSVLTR-HSVTK cassette was amplified from pPS1337 using primers that allow subsequent cloning into the FseI and AscI sites of the “long arm”-TA vector plasmid. For this PCR, the sequence of the sense primer was 5′-ATTACGGCCGGCCGCAAAGGAATTCAAGA TCTGA-3′ (SEQ ID NO: 40); the sequence of the anti-sense primer was 5′-ATTACGGCGCGCCCCTC ACAGGCCGCACCCAGCT-3′ (SEQ ID NO: 41). [0353]
  • The final step in the construction of the targeting vector involved cloning the 8.8 kb SgrAI-AscI fragment containing the “long arm” and RSVLTR-HSVTK gene into the SgrAI and AscI sites of the pSP72-“short arm”-βgal-PGKneo plasmid. This targeting vector was linearized by digestion with either AscI or PacI before electroporation into ES cells. [0354]
  • Example 10
  • Antisense-Mediated Beer Inactivation [0355]
  • 17-nucleotide antisense oligonucleotides are prepared in an overlapping format, in such a way that the 5′ end of the first oligonucleotide overlaps the translation initiating AUG of the Beer transcript, and the 5′ ends of successive oligonucleotides occur in 5 nucleotide increments moving in the 5′ direction (up to 50 nucleotides away), relative to the Beer AUG. Corresponding control oligonucleotides are designed and prepared using equivalent base composition but redistributed in sequence to inhibit any significant hybridization to the coding mRNA. Reagent delivery to the test cellular system is conducted through cationic lipid delivery (P. L. Felgner, [0356] Proc. Natl. Acad. Sci. USA 84:7413, 1987). 2 ug of antisense oligonucleotide is added to 100 ul of reduced serum media (Opti-MEM I reduced serum media; Life Technologies, Gaithersburg Md.) and this is mixed with Lipofectin reagent (6 ul) (Life Technologies, Gaithersburg Md.) in the 100 ul of reduced serum media. These are mixed, allowed to complex for 30 minutes at room temperature and the mixture is added to previously seeded MC3T3E21 or KS483 cells. These cells are cultured and the mRNA recovered. Beer mRNA is monitored using RT-PCR in conjunction with Beer specific primers. In addition, separate experimental wells are collected and protein levels characterized through western blot methods described in Example 4. The cells are harvested, resuspended in lysis buffer (50 mM Tris pH 7.5, 20 mM NaCl, 1 mM EDTA, 1% SDS) and the soluble protein collected. This material is applied to 10-20 % gradient denaturing SDS PAGE. The separated proteins are transferred to nitrocellulose and the western blot conducted as above using the antibody reagents described. In parallel, the control oligonucleotides are added to identical cultures and experimental operations are repeated. Decrease in Beer mRNA or protein levels are considered significant if the treatment with the antisense oligonucleotide results in a 50% change in either instance compared to the control scrambled oligonucleotide. This methodology enables selective gene inactivation and subsequent phenotype characterization of the mineralized nodules in the tissue culture model.
  • Example 11
  • Modeling of Sclerostin Core Region [0357]
  • Homology recognition techniques (e.g., PSI-BLAST (Altschul et al., [0358] Nucleic Acids Res. 25:3389-402 (1997)), FUGUE (Shi et al., J. Mol. Biol. 310:243-57 (2001)) suggested that the core region of SOST (SOST_Core) adopts a cystine-knot fold. FUGUE is a sensitive method for detecting homology between sequences and structures. Human Chorionic Gonadotropin β (hCG-β), for which an experimentally determined 3D structure is known, was identified by FUGUE (Shi et al., supra) as the closest homologue of SOST_Core. Therefore, hCG-β was used as the structural template to build 3D models for SOST_Core.
  • An alignment of SOST_Core and its close homologues is shown in FIG. 7. Among the homologues shown in the alignment, only hCG-β (CGHB) had known 3D structure. The sequence identity between SOST_Core and hCG-β was approximately 25%. Eight CYS residues were conserved throughout the family, emphasizing the overall structural similarity between SOST_Core and hCG-β. Three pairs of cystines (1-5, 3-7, 4-8) formed disulfide bonds (shown with solid lines in FIG. 7) in a “knot” configuration, which was characteristic to the cystine-knot fold. An extra disulfide bond (2-6), shown as a dotted line in FIG. 7, was unique to this family and distinguished the family of proteins from other cystine-knot families (e.g., TGF-β, BMP). [0359]
  • SOST_Core was modeled using PDB (Berman et al., [0360] Acta Crystallogr. D. Biol. Crystallogr. 58(Pt 6 Pt1):899-907 (2002)) entry 1HCN, the 3D structure of hCG-β (Wu et al., Structure 2:545-58 (1994)), as the structural template. Models were calculated with MODELER (Sali & Blundell, J. Mol. Biol. 234:779-815 (1993)). A snapshot of the best model is shown in FIG. 8.
  • Most of the cystine-knot proteins form dimers because of the lack of hydrophobic core in a monomer (Scheufler et al., supra; Schlunegger and Grutter, [0361] J. Mol. Biol. 231:445-58 (1993)); Wu et al., supra). SOST likely follows the same rule and forms a homodimer to increase its stability. Constructing a model for the dimerized SOST_Core region presented several challenges because (1) the sequence similarity between SOST_Core and hCG-β was low (25%); (2) instead of a homodimer, hCG-β formed a heterodimer with hCG-α; and (3) a number of different relative conformations of monomers have been observed in dimerized cystine-knot proteins from different families (e.g., PDGF, TGF-β, Neurotrophin, IL-17F, Gonadotropin), which suggested that the dimer conformation of SOST could deviate significantly from the hCG-α/β heterodimer conformation. In constructing the model, hCG-α was replaced with hCG-β from the heterodimer structure (1HCN) using structure superimposition techniques combined with manual adjustment, and then a SOST_Core homodimer model was built according to the pseudo hCG-β homodimer structure. The final model is shown in FIG. 9.
  • Example 12
  • Modeling SOST-BMP Interaction [0362]
  • This example describes protein modeling of type I and type II receptor binding sites on BMP that are involved with interaction between BMP and SOST. [0363]
  • Competition studies demonstrated that SOST competed with both type I and type II receptors for binding to BMP. In an ELISA-based competition assay, BMP-6 selectively interacted with the sclerostin-coated surface (300 ng/well) with high affinity (K[0364] D=3.4 nM). Increasing amounts of BMP receptor IA (FC fusion construct) competed with sclerostin for binding to BMP-6 (11 nM) (IC50=114 nM). A 10-fold molar excess of the BMP receptor was sufficient to reduce binding of sclerostin to BMP-6 by approximately 50%. This competition was also observed with a BMP receptor II-FC fusion protein (IC50=36 nM) and DAN (IC50=43 nM). Specificity of the assay was shown by lack of competition for binding to BMP-6 between sclerostin and a rActivin R1B-FC fusion protein, a TGF-β receptor family member that did not bind BMP.
  • The type I and type II receptor binding sites on a BMP polypeptide have been mapped and were spatially separated (Scheufler et al., supra; Innis et al., supra; Nickel et al., supra; Hart et al. supra). Noggin, another BMP antagonist that binds to BMP with high affinity, contacts BMP at both type I and type II receptor binding sites via the N-terminal portion of Noggin (Groppe et al., supra). The two β-strands in the core region near the C-terminal also contact BMP at the type II receptor binding site. [0365]
  • A manually tuned alignment of Noggin and SOST indicated that the two polypeptides shared sequence similarity between the N-terminal portions of the proteins and between the core regions. An amino acid sequence alignment is presented in FIG. 10. The cysteine residues that form the characteristic cys-knot were conserved between Noggin and SOST. The overall sequence identity was 24%, and the sequence identity within the N-terminal binding region (alignment positions 1-45) was 33%. Two residues in the Noggin N-terminal binding region, namely Leu (L) at alignment position 21 and Glu (E) at position 23, were reported to play important roles in BMP binding (Groppe et al., supra). Both residues were conserved in SOST as well. The sequence similarity within the core region (alignment positions 131-228) was about 20%, but the cys-knot scaffold was maintained and a sufficient number of key residues was conserved, supporting homology between Noggin and SOST. [0366]
  • The Noggin structure was compared to SOST also to understand how two SOST monomers dimerize. As shown in FIG. 11, the Noggin structure suggested that the linker between the N-terminal region and the core region not only played a role in connecting the two regions, but also formed part of the dimerization interface between two Noggin monomers. One major difference between Noggin and SOST was that the linker between the N-terminal region and the core region was much shorter in SOST. [0367]
  • The C-terminal region of SOST may play a role in SOST dimerization. The sequence of Noggin ended with the core region, while SOST had an extra C-terminal region. In the Noggin structure a disulfide bond connected the C-termini of two Noggin monomers. Thus, the C-terminal region of SOST started close to the interface of two monomers and could contribute to dimerization. In addition, secondary structure prediction showed that some portions of the C-terminal region of SOST had a tendency to form helices. This region in SOST may be responsible for the dimerization activity, possibly through helix-helix packing, which mimicked the function of the longer linker in Noggin. Another difference between the structure of Noggin and SOST was the amino acid insertion in the SOST core region at alignment positions 169-185 (see FIG. 10). This insertion extended a β-hairpin, which pointed towards the dimerization interface in the Noggin structure (shown in FIG. 11 as a loop region in the middle of the monomers and above the C-terminal Cys residue). This elongated β-hairpin could also contribute to SOST dimerization. [0368]
  • Example 13
  • Design and Preparation of SOST Peptide Immunogens [0369]
  • This Example describes the design of SOST peptide immunogens that are used for immunizing animals and generating antibodies that block interactions between BMP and SOST and prevent dimer formation of SOST monomers. [0370]
  • BMP Binding Fragments [0371]
  • The overall similarity between SOST and Noggin and the similarity between the N-terminal regions of the two polypeptides suggest that SOST may interact with BMP in a similar manner to Noggin. That is, the N-terminal region of SOST may interact with both the type I and type II receptor binding sites on BMP, and a portion of the core region (amino acid alignment positions 190-220 in FIG. 10) may interact with the type II receptor binding site such that antibodies specific for these SOST regions may block or impair binding of BMP to SOST. [0372]
  • The amino acid sequences of these SOST polypeptide fragments for rat and human SOST are provided as follows. [0373]
  • SOST_N_Linker: The N-terminal region (includes the short linker that connects to the core region) [0374]
    Human: QGWQAFKNDATEIIPELGEYPEPPPELENNKTMNRAENGGRPPHHPFETKDVSEYS (SEQ ID NO:92)
    Rat: QGWQAFKNDATEIIPGLREYPEPPQELENNQTMNRAENGGRPPHHPYDTKDVSEYS (SEQ ID NO:93)
  • SOST_Core_Bind: Portion of the core region that is likely to contact BMP at its type II receptor binding site (extended slightly at both termini to include the CYS residue anchors): [0375]
    Human:
    CIPDRYRAQRVQLLCPGGEAPRARKVRLVASC (SEQ ID NO:94)
    Rat:
    CIPDRYRAQRVQLLCPGGAAPRSRKVRLVASC (SEQ ID NO:95)
  • SOST Dimerization Fragments [0376]
  • The C-terminal region of SOST is likely to be involved in the formation of SOST homodimers (see Example 12). The elongated β-hairpin may also play a role in homodimer formation. Antibodies that specifically bind to such regions may prevent or impair dimerization of SOST monomers, which may in turn interfere with interaction between SOST and BMP. Polypeptide fragments in rat and human SOST corresponding to these regions are as follows. [0377]
  • SOST_C: the C-terminal region [0378]
    (SEQ ID NO:96)
    Human: LTRFHNQSELKDFGTEAARPQKGRKPRPRARSAKANQAELENAY
    (SEQ ID NO:97)
    Rat: LTRFHNQSELKDFGPETARPQKGRKPRPRARGAKANQAELENAY
  • SOST_Core_Dimer: Portion of the core region that is likely involved in SOST dimerization (extended slightly at both termini to include the Cys residue anchors): [0379]
    Human: CGPARLLPNAIGRGKWWRPSGPDFRC (SEQ ID NO:98)
    Rat: CGPARLLPNAIGRVKWWRPNGPDFRC (SEQ ID NO:99)
  • BMP Binding Fragment at SOST N-Terminus [0380]
  • The key N-terminal binding region of SOST (alignment positions 1-35 in FIG. 10) was modeled on the basis of the Noggin/BMP-7 complex structure (Protein Data Bank Entry No: 1M4U) and the amino acid sequence alignment (see FIG. 10) to identify amino acid residues of the SOST N-terminus that likely interact with BMP. The model of SOST is presented in FIG. 12. In the comparative model, phenylalanine (Phe, F) at alignment position 8 (see arrow and accompanying text) in the SOST sequence projects into a hydrophobic pocket on the surface of the BMP dimer. The same “knob-into-hole” feature has been observed in the BMP and type I receptor complex structure (Nickel et al., supra), where Phe85 of the receptor fits into the same pocket, which is a key feature in ligand-type I receptor recognition for TGF-β superfamily members (including, for example, TGF-β family, BMP family, and the like). According to the model, a proline (Pro) directed turn is also conserved, which allows the N-terminal binding fragment to thread along the BMP dimer surface, traveling from type I receptor binding site to type II receptor binding site on the other side of the complex. Also conserved is another Pro-directed turn near the carboxy end of the binding fragment, which then connects to the linker region. Extensive contacts between SOST and BMP are evident in FIG. 12. [0381]
  • Peptide Immunogens [0382]
  • Peptides were designed to encompass the SOST N-terminal region predicted to make contact with BMP proteins. The peptide sequences are presented below. For immunizing animals, the peptide sequences were designed to overlap, and an additional cysteine was added to the C-terminal end to facilitate crosslinking to KLH. The peptides were then used for immunization. The peptide sequences of the immunogens are as follows. [0383]
    Human SOST:
    QGWQAFKNDATEIIPELGEY (SEQ ID NO:47)
    TEIIPELGEYPEPPPELENN (SEQ ID NO:48)
    PEPPPELENNKTMNRAENGG (SEQ ID NO:49)
    KTMNRAENGGRPPHHPFETK (SEQ ID NO:50)
    RPPHHPFETKDVSEYS (SEQ ID NO:51)
    Human SOST Peptides with Additional Cys:
    QGWQAFKNDATEIIPELGEY-C (SEQ ID NO:52)
    TEIIPELGEYPEPPPELENN-C (SEQ ID NO:53)
    PEPPPELENNKTMNRAENGG-C (SEQ ID NO:54)
    KTMNRAENGGRPPHHPFETK-C (SEQ ID NO:55)
    RPPHHPFETKDVSEYS-C (SEQ ID NO:56)
    Rat SOST:
    QGWQAFKNDATEIIPGLREYPEPP (SEQ ID NO:57)
    PEPPQELENNQTMNRAENGG (SEQ ID NO:58)
    ENGGRPPHHPYDTKDVSEYS (SEQ ID NO:59)
    TEIIPGLREYPEPPQELENN (SEQ ID NO:60)
    Rat SOST Peptides with Additional Cys:
    QGWQAFKNDATEIIPGLREYPEPP-C (SEQ ID NO:61)
    PEPPQELENNQTMNRAENGG-C (SEQ ID NO:62)
    ENGGRPPHHPYDTKDVSEYS-C (SEQ ID NO:63)
    TEIIPGLREYPEPPQELENN-C (SEQ ID NO:64)
  • The following peptides were designed to contain the amino acid portion of core region that was predicted to make contact with BMP proteins. Cysteine was added at the C-terminal end of each peptide for conjugation to KLH, and the conjugated peptides were used for immunization. In the Docking Core N-terminal Peptide an internal cysteine was changed to a serine to avoid double conjugation to KLH. [0384]
    For Human SOST:
    Amino acid sequence without Cys residues added:
    Docking_Core_N-terminal Peptide: IPDRYRAQRVQLLCPGGEAP (SEQ ID NO:66)
    Docking_Core_Cterm_Peptide: QLLCPGGEAPRARKVRLVAS (SEQ ID NO:67)
    Docking_Core_N-terminal_Peptide: IPDRYRAQRVQLLCPGGEAP-C (SEQ ID NO:68)
    Docking_Core_Cterm_Petide: QLLCPGGEAPRARKVRLVAS-C (SEQ ID NO:69)
    For Rat SOST:
    Amino acid sequence without Cys residues added or substituted:
    Docking_Core_N-terminal_Peptide: IPDRYRAQRVQLLSPGG (SEQ ID NO:70)
    Docking_Core_Cterm_Peptide: PGGAAPRSRKVRLVAS (SEQ ID NO:71)
    Peptide immunogens with Cys added and substituted:
    Docking_Core_N-terminal_Peptide: IPDRYRAQRVQLLSPGG-C (SEQ ID NO:72)
    Docking_Core_Cterm_Peptide: PGGAAPRSRKVRLVAS-C (SEQ ID NO:73)
  • Two regions within SOST that potentially interact to form SOST homodimers include the amino acids with the SOST core region that are not present in Noggin. Human SOST peptides designed to contain this sequence had a C-terminal or N-terminal Cys that was conjugated to KLH. For the rat SOST peptide, a cysteine was added to the carboxy terminus of the sequence (SEQ ID NO: 76). The KLH conjugated peptides were used for immunization. [0385]
    For Human SOST:
    CGPARLLPNAIGRGKWWRPS (SEQ ID NO:74)
    IGRGKWWRPSGPDFRC (SEQ ID NO:75)
    For Rat SOST:
    PNAIGRVKWWRPNGPDFR (SEQ ID NO:76)
    Rat SOST peptide with cysteine added
    PNAIGRVKWWRPNGPDFR-C (SEQ ID NO:77)
  • The second region within SOST that potentially interacts to form SOST homodimers includes the C-terminal region. Peptide immunogens were designed to include amino acid sequences within this region (see below). For conjugation to KLH, a cysteine residue was added to the C-terminal end, and the conjugated peptides were used for immunization. [0386]
    For Human SOST:
    (SEQ ID NO:78)
    KRLTRFHNQS ELKDFGTEAA
    (SEQ ID NO:79)
    ELKDFGTEAA RPQKGRKPRP
    (SEQ ID NO:80)
    RPQKGRKPRP RARSAKANQA
    (SEQ ID NO:81)
    RARSAKANQA ELENAY
    Peptide immunogens with Cys added at C-terminus:
    (SEQ ID NO:82)
    KRLTRFHNQS ELKDFGTEAA-C
    (SEQ ID NO:83)
    ELKDFGTEAA RPQKGRKPRP-C
    (SEQ ID NO:84)
    RPQKGRKPRP RARSAKANQA-C
    (SEQ ID NO:85)
    RARSAKANQA ELENAY-C
    For Rat SOST:
    (SEQ ID NO:86)
    KRLTRFHNQSELKDFGPETARPQ
    (SEQ ID NO:87)
    KGRKPRPRARGAKANQAELENAY
    (SEQ ID NO:88)
    SELKDFGPETARPQKGRKPRPRAR
    Peptide immunogens with Cys added at C-terminus:
    (SEQ ID NO:89)
    KRLTRFHNQSELKDFGPETARPQ-C
    (SEQ ID NO:90)
    KGRKPRPRARGAKANQAELENAY-C
    (SEQ ID NO:91)
    SELKDFGPETARPQKGRKPRPRAR-C
  • Example 14
  • Assay for Detecting Binding of Antibodies to a TGF-Beta Binding-Protein [0387]
  • This example describes an assay for detecting binding of a ligand, for example, an antibody or antibody fragment thereof, to sclerostin. [0388]
  • A FLAG®-sclerostin fusion protein was prepared according to protocols provided by the manufacturer (Sigma Aldrich, St. Louis, Mo.) and as described in U.S. Pat. No. 6,395,511. Each well of a 96 well microtiter plate is coated with anti-FLAG® monoclonal antibody (Sigma Aldrich) and then blocked with 10% BSA in PBS. The fusion protein (20 ng) is added to 100 μl PBS/0.2% BSA and adsorbed onto the 96-well plate for 60 minutes at room temperature. This protein solution is removed and the wells are washed to remove unbound fusion protein. A BMP, for example, BMP-4, BMP-5, BMP-6, or BMP-7, is diluted in PBS/0.2% BSA and added to each well at concentrations ranging from 10 pM to 500 nM. After an incubation for 2 hours at room temperature, the binding solution is removed and the plate is washed three times with 200 μl volumes of PBS/0.2% BSA. Binding of the BMP to sclerostin is detected using polyclonal antiserum or monoclonal antibody specific for the BMP and an appropriate enzyme-conjugated second step reagent according to standard ELISA techniques (see, e.g., Ausubel et al., [0389] Current Protocols in Mol Biol. Vol 2 11.2.1-11.2.22 (1998)). Specific binding is calculated by subtracting non-specific binding from total binding and analyzed using the LIGAND program (Munson and Podbard, Anal. Biochem. 107:220-39 (1980)).
  • Binding of sclerostin to a BMP is also detected by homogeneous time resolved fluorescence detection (Mellor et al., [0390] J Biomol. Screening, 3:91-99 (1998)). A polynucleotide sequence encoding sclerostin is operatively linked to a human immunoglobulin constant region in a recombinant nucleic acid construct and expressed as a human Fc-sclerostin fusion protein according to methods known in the art and described herein. Similarly, a BMP ligand is engineered and expressed as a BMP-mouse Fe fusion protein. These two fusion proteins are incubated together and the assay conducted as described by Mellor et al.
  • Example 15
  • Screening Assay for Antibodies that Inhibit Binding of TGF-Beta Family Members to TGF-Beta Binding Protein [0391]
  • This example describes a method for detecting an antibody that inhibits binding of a TGF-beta family member to sclerostin. An ELISA is performed essentially as described in Example 14 except that the BMP concentration is held fixed at its Kd (determined, for example, by BIAcore analysis). In addition, an antibody or a library or collection of antibodies is added to the wells to a concentration of 1 μM. Antibodies are incubated for 2 hours at room temperature with the BMP and sclerostin, the solution removed, and the bound BMP is quantified as described (see Example 14). Antibodies that inhibit 40% of the BMP binding observed in the absence of antibody are considered antagonists of this interaction. These antibodies are further evaluated as potential inhibitors by performing titration studies to determine their inhibition constants and their effect on TGF-beta binding-protein binding affinity. Comparable specificity control assays may also be conducted to establish the selectivity profile for the identified antagonist using assays dependent on the BMP ligand action (e.g., a BMP/BMP receptor competition study). [0392]
  • Example 16
  • Inhibition of TGF-Beta Binding-Protein Localization to Bone Matrix [0393]
  • Evaluation of inhibition of localization to bone matrix (hydroxyapatite) is conducted using modifications to the method of Nicolas ([0394] Calcif. Tissue Int. 57:206-12 (1995)). Briefly, 125I-labelled TGF-beta binding-protein is prepared as described by Nicolas (supra). Hydroxyapatite is added to each well of a 96-well microtiter plate equipped with a polypropylene filtration membrane (Polyfiltroninc, Weymouth Mass.). TGF-beta binding-protein diluted in 0.2% albumin in PBS buffer is then added to the wells. The wells containing matrix are washed 3 times with 0.2% albumin in PBS buffer. Adsorbed TGF-beta binding-protein is eluted using 0.3 M NaOH and then quantified.
  • An antibody that inhibits or impairs binding of the sclerostin TGF-beta binding protein to the hydroxyapatite is identified by incubating the TGF-beta binding protein with the antibody and applying the mixture to the matrix as described above. The matrix is washed 3 times with 0.2% albumin in PBS buffer. Adsorbed sclerostin is eluted with 0.3 M NaOH and then quantified. An antibody that inhibits the level of binding of sclerostin to the hydroxyapatite by at least 40% compared to the level of binding observed in the absence of antibody is considered a bone localization inhibitor. Such an antibody is further characterized in dose response studies to determine its inhibition constant and its effect on TGF-beta binding-protein binding affinity. [0395]
  • From the foregoing, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims. [0396]
  • 1 143 1 2301 DNA Homo sapien 1 agagcctgtg ctactggaag gtggcgtgcc ctcctctggc tggtaccatg cagctcccac 60 tggccctgtg tctcgtctgc ctgctggtac acacagcctt ccgtgtagtg gagggccagg 120 ggtggcaggc gttcaagaat gatgccacgg aaatcatccc cgagctcgga gagtaccccg 180 agcctccacc ggagctggag aacaacaaga ccatgaaccg ggcggagaac ggagggcggc 240 ctccccacca cccctttgag accaaagacg tgtccgagta cagctgccgc gagctgcact 300 tcacccgcta cgtgaccgat gggccgtgcc gcagcgccaa gccggtcacc gagctggtgt 360 gctccggcca gtgcggcccg gcgcgcctgc tgcccaacgc catcggccgc ggcaagtggt 420 ggcgacctag tgggcccgac ttccgctgca tccccgaccg ctaccgcgcg cagcgcgtgc 480 agctgctgtg tcccggtggt gaggcgccgc gcgcgcgcaa ggtgcgcctg gtggcctcgt 540 gcaagtgcaa gcgcctcacc cgcttccaca accagtcgga gctcaaggac ttcgggaccg 600 aggccgctcg gccgcagaag ggccggaagc cgcggccccg cgcccggagc gccaaagcca 660 accaggccga gctggagaac gcctactaga gcccgcccgc gcccctcccc accggcgggc 720 gccccggccc tgaacccgcg ccccacattt ctgtcctctg cgcgtggttt gattgtttat 780 atttcattgt aaatgcctgc aacccagggc agggggctga gaccttccag gccctgagga 840 atcccgggcg ccggcaaggc ccccctcagc ccgccagctg aggggtccca cggggcaggg 900 gagggaattg agagtcacag acactgagcc acgcagcccc gcctctgggg ccgcctacct 960 ttgctggtcc cacttcagag gaggcagaaa tggaagcatt ttcaccgccc tggggtttta 1020 agggagcggt gtgggagtgg gaaagtccag ggactggtta agaaagttgg ataagattcc 1080 cccttgcacc tcgctgccca tcagaaagcc tgaggcgtgc ccagagcaca agactggggg 1140 caactgtaga tgtggtttct agtcctggct ctgccactaa cttgctgtgt aaccttgaac 1200 tacacaattc tccttcggga cctcaatttc cactttgtaa aatgagggtg gaggtgggaa 1260 taggatctcg aggagactat tggcatatga ttccaaggac tccagtgcct tttgaatggg 1320 cagaggtgag agagagagag agaaagagag agaatgaatg cagttgcatt gattcagtgc 1380 caaggtcact tccagaattc agagttgtga tgctctcttc tgacagccaa agatgaaaaa 1440 caaacagaaa aaaaaaagta aagagtctat ttatggctga catatttacg gctgacaaac 1500 tcctggaaga agctatgctg cttcccagcc tggcttcccc ggatgtttgg ctacctccac 1560 ccctccatct caaagaaata acatcatcca ttggggtaga aaaggagagg gtccgagggt 1620 ggtgggaggg atagaaatca catccgcccc aacttcccaa agagcagcat ccctcccccg 1680 acccatagcc atgttttaaa gtcaccttcc gaagagaagt gaaaggttca aggacactgg 1740 ccttgcaggc ccgagggagc agccatcaca aactcacaga ccagcacatc ccttttgaga 1800 caccgccttc tgcccaccac tcacggacac atttctgcct agaaaacagc ttcttactgc 1860 tcttacatgt gatggcatat cttacactaa aagaatatta ttgggggaaa aactacaagt 1920 gctgtacata tgctgagaaa ctgcagagca taatagctgc cacccaaaaa tctttttgaa 1980 aatcatttcc agacaacctc ttactttctg tgtagttttt aattgttaaa aaaaaaaagt 2040 tttaaacaga agcacatgac atatgaaagc ctgcaggact ggtcgttttt ttggcaattc 2100 ttccacgtgg gacttgtcca caagaatgaa agtagtggtt tttaaagagt taagttacat 2160 atttattttc tcacttaagt tatttatgca aaagtttttc ttgtagagaa tgacaatgtt 2220 aatattgctt tatgaattaa cagtctgttc ttccagagtc cagagacatt gttaataaag 2280 acaatgaatc atgaccgaaa g 2301 2 213 PRT Homo sapien 2 Met Gln Leu Pro Leu Ala Leu Cys Leu Val Cys Leu Leu Val His Thr 1 5 10 15 Ala Phe Arg Val Val Glu Gly Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 Ala Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser 115 120 125 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 145 150 155 160 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 Ser Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly 180 185 190 Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu 195 200 205 Leu Glu Asn Ala Tyr 210 3 2301 DNA Homo sapien 3 agagcctgtg ctactggaag gtggcgtgcc ctcctctggc tggtaccatg cagctcccac 60 tggccctgtg tctcgtctgc ctgctggtac acacagcctt ccgtgtagtg gagggctagg 120 ggtggcaggc gttcaagaat gatgccacgg aaatcatccc cgagctcgga gagtaccccg 180 agcctccacc ggagctggag aacaacaaga ccatgaaccg ggcggagaac ggagggcggc 240 ctccccacca cccctttgag accaaagacg tgtccgagta cagctgccgc gagctgcact 300 tcacccgcta cgtgaccgat gggccgtgcc gcagcgccaa gccggtcacc gagctggtgt 360 gctccggcca gtgcggcccg gcgcgcctgc tgcccaacgc catcggccgc ggcaagtggt 420 ggcgacctag tgggcccgac ttccgctgca tccccgaccg ctaccgcgcg cagcgcgtgc 480 agctgctgtg tcccggtggt gaggcgccgc gcgcgcgcaa ggtgcgcctg gtggcctcgt 540 gcaagtgcaa gcgcctcacc cgcttccaca accagtcgga gctcaaggac ttcgggaccg 600 aggccgctcg gccgcagaag ggccggaagc cgcggccccg cgcccggagc gccaaagcca 660 accaggccga gctggagaac gcctactaga gcccgcccgc gcccctcccc accggcgggc 720 gccccggccc tgaacccgcg ccccacattt ctgtcctctg cgcgtggttt gattgtttat 780 atttcattgt aaatgcctgc aacccagggc agggggctga gaccttccag gccctgagga 840 atcccgggcg ccggcaaggc ccccctcagc ccgccagctg aggggtccca cggggcaggg 900 gagggaattg agagtcacag acactgagcc acgcagcccc gcctctgggg ccgcctacct 960 ttgctggtcc cacttcagag gaggcagaaa tggaagcatt ttcaccgccc tggggtttta 1020 agggagcggt gtgggagtgg gaaagtccag ggactggtta agaaagttgg ataagattcc 1080 cccttgcacc tcgctgccca tcagaaagcc tgaggcgtgc ccagagcaca agactggggg 1140 caactgtaga tgtggtttct agtcctggct ctgccactaa cttgctgtgt aaccttgaac 1200 tacacaattc tccttcggga cctcaatttc cactttgtaa aatgagggtg gaggtgggaa 1260 taggatctcg aggagactat tggcatatga ttccaaggac tccagtgcct tttgaatggg 1320 cagaggtgag agagagagag agaaagagag agaatgaatg cagttgcatt gattcagtgc 1380 caaggtcact tccagaattc agagttgtga tgctctcttc tgacagccaa agatgaaaaa 1440 caaacagaaa aaaaaaagta aagagtctat ttatggctga catatttacg gctgacaaac 1500 tcctggaaga agctatgctg cttcccagcc tggcttcccc ggatgtttgg ctacctccac 1560 ccctccatct caaagaaata acatcatcca ttggggtaga aaaggagagg gtccgagggt 1620 ggtgggaggg atagaaatca catccgcccc aacttcccaa agagcagcat ccctcccccg 1680 acccatagcc atgttttaaa gtcaccttcc gaagagaagt gaaaggttca aggacactgg 1740 ccttgcaggc ccgagggagc agccatcaca aactcacaga ccagcacatc ccttttgaga 1800 caccgccttc tgcccaccac tcacggacac atttctgcct agaaaacagc ttcttactgc 1860 tcttacatgt gatggcatat cttacactaa aagaatatta ttgggggaaa aactacaagt 1920 gctgtacata tgctgagaaa ctgcagagca taatagctgc cacccaaaaa tctttttgaa 1980 aatcatttcc agacaacctc ttactttctg tgtagttttt aattgttaaa aaaaaaaagt 2040 tttaaacaga agcacatgac atatgaaagc ctgcaggact ggtcgttttt ttggcaattc 2100 ttccacgtgg gacttgtcca caagaatgaa agtagtggtt tttaaagagt taagttacat 2160 atttattttc tcacttaagt tatttatgca aaagtttttc ttgtagagaa tgacaatgtt 2220 aatattgctt tatgaattaa cagtctgttc ttccagagtc cagagacatt gttaataaag 2280 acaatgaatc atgaccgaaa g 2301 4 23 PRT Homo sapien 4 Met Gln Leu Pro Leu Ala Leu Cys Leu Val Cys Leu Leu Val His Thr 1 5 10 15 Ala Phe Arg Val Val Glu Gly 20 5 2301 DNA Homo sapien 5 agagcctgtg ctactggaag gtggcgtgcc ctcctctggc tggtaccatg cagctcccac 60 tggccctgtg tctcatctgc ctgctggtac acacagcctt ccgtgtagtg gagggccagg 120 ggtggcaggc gttcaagaat gatgccacgg aaatcatccg cgagctcgga gagtaccccg 180 agcctccacc ggagctggag aacaacaaga ccatgaaccg ggcggagaac ggagggcggc 240 ctccccacca cccctttgag accaaagacg tgtccgagta cagctgccgc gagctgcact 300 tcacccgcta cgtgaccgat gggccgtgcc gcagcgccaa gccggtcacc gagctggtgt 360 gctccggcca gtgcggcccg gcgcgcctgc tgcccaacgc catcggccgc ggcaagtggt 420 ggcgacctag tgggcccgac ttccgctgca tccccgaccg ctaccgcgcg cagcgcgtgc 480 agctgctgtg tcccggtggt gaggcgccgc gcgcgcgcaa ggtgcgcctg gtggcctcgt 540 gcaagtgcaa gcgcctcacc cgcttccaca accagtcgga gctcaaggac ttcgggaccg 600 aggccgctcg gccgcagaag ggccggaagc cgcggccccg cgcccggagc gccaaagcca 660 accaggccga gctggagaac gcctactaga gcccgcccgc gcccctcccc accggcgggc 720 gccccggccc tgaacccgcg ccccacattt ctgtcctctg cgcgtggttt gattgtttat 780 atttcattgt aaatgcctgc aacccagggc agggggctga gaccttccag gccctgagga 840 atcccgggcg ccggcaaggc ccccctcagc ccgccagctg aggggtccca cggggcaggg 900 gagggaattg agagtcacag acactgagcc acgcagcccc gcctctgggg ccgcctacct 960 ttgctggtcc cacttcagag gaggcagaaa tggaagcatt ttcaccgccc tggggtttta 1020 agggagcggt gtgggagtgg gaaagtccag ggactggtta agaaagttgg ataagattcc 1080 cccttgcacc tcgctgccca tcagaaagcc tgaggcgtgc ccagagcaca agactggggg 1140 caactgtaga tgtggtttct agtcctggct ctgccactaa cttgctgtgt aaccttgaac 1200 tacacaattc tccttcggga cctcaatttc cactttgtaa aatgagggtg gaggtgggaa 1260 taggatctcg aggagactat tggcatatga ttccaaggac tccagtgcct tttgaatggg 1320 cagaggtgag agagagagag agaaagagag agaatgaatg cagttgcatt gattcagtgc 1380 caaggtcact tccagaattc agagttgtga tgctctcttc tgacagccaa agatgaaaaa 1440 caaacagaaa aaaaaaagta aagagtctat ttatggctga catatttacg gctgacaaac 1500 tcctggaaga agctatgctg cttcccagcc tggcttcccc ggatgtttgg ctacctccac 1560 ccctccatct caaagaaata acatcatcca ttggggtaga aaaggagagg gtccgagggt 1620 ggtgggaggg atagaaatca catccgcccc aacttcccaa agagcagcat ccctcccccg 1680 acccatagcc atgttttaaa gtcaccttcc gaagagaagt gaaaggttca aggacactgg 1740 ccttgcaggc ccgagggagc agccatcaca aactcacaga ccagcacatc ccttttgaga 1800 caccgccttc tgcccaccac tcacggacac atttctgcct agaaaacagc ttcttactgc 1860 tcttacatgt gatggcatat cttacactaa aagaatatta ttgggggaaa aactacaagt 1920 gctgtacata tgctgagaaa ctgcagagca taatagctgc cacccaaaaa tctttttgaa 1980 aatcatttcc agacaacctc ttactttctg tgtagttttt aattgttaaa aaaaaaaagt 2040 tttaaacaga agcacatgac atatgaaagc ctgcaggact ggtcgttttt ttggcaattc 2100 ttccacgtgg gacttgtcca caagaatgaa agtagtggtt tttaaagagt taagttacat 2160 atttattttc tcacttaagt tatttatgca aaagtttttc ttgtagagaa tgacaatgtt 2220 aatattgctt tatgaattaa cagtctgttc ttccagagtc cagagacatt gttaataaag 2280 acaatgaatc atgaccgaaa g 2301 6 213 PRT Homo sapien 6 Met Gln Leu Pro Leu Ala Leu Cys Leu Ile Cys Leu Leu Val His Thr 1 5 10 15 Ala Phe Arg Val Val Glu Gly Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 Ala Thr Glu Ile Ile Arg Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser 115 120 125 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 145 150 155 160 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 Ser Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly 180 185 190 Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu 195 200 205 Leu Glu Asn Ala Tyr 210 7 2301 DNA Homo sapien 7 agagcctgtg ctactggaag gtggcgtgcc ctcctctggc tggtaccatg cagctcccac 60 tggccctgtg tctcgtctgc ctgctggtac acacagcctt ccgtgtagtg gagggccagg 120 ggtggcaggc gttcaagaat gatgccacgg aaatcatccg cgagctcgga gagtaccccg 180 agcctccacc ggagctggag aacaacaaga ccatgaaccg ggcggagaac ggagggcggc 240 ctccccacca cccctttgag accaaagacg tgtccgagta cagctgccgc gagctgcact 300 tcacccgcta cgtgaccgat gggccgtgcc gcagcgccaa gccggtcacc gagctggtgt 360 gctccggcca gtgcggcccg gcgcgcctgc tgcccaacgc catcggccgc ggcaagtggt 420 ggcgacctag tgggcccgac ttccgctgca tccccgaccg ctaccgcgcg cagcgcgtgc 480 agctgctgtg tcccggtggt gaggcgccgc gcgcgcgcaa ggtgcgcctg gtggcctcgt 540 gcaagtgcaa gcgcctcacc cgcttccaca accagtcgga gctcaaggac ttcgggaccg 600 aggccgctcg gccgcagaag ggccggaagc cgcggccccg cgcccggagc gccaaagcca 660 accaggccga gctggagaac gcctactaga gcccgcccgc gcccctcccc accggcgggc 720 gccccggccc tgaacccgcg ccccacattt ctgtcctctg cgcgtggttt gattgtttat 780 atttcattgt aaatgcctgc aacccagggc agggggctga gaccttccag gccctgagga 840 atcccgggcg ccggcaaggc ccccctcagc ccgccagctg aggggtccca cggggcaggg 900 gagggaattg agagtcacag acactgagcc acgcagcccc gcctctgggg ccgcctacct 960 ttgctggtcc cacttcagag gaggcagaaa tggaagcatt ttcaccgccc tggggtttta 1020 agggagcggt gtgggagtgg gaaagtccag ggactggtta agaaagttgg ataagattcc 1080 cccttgcacc tcgctgccca tcagaaagcc tgaggcgtgc ccagagcaca agactggggg 1140 caactgtaga tgtggtttct agtcctggct ctgccactaa cttgctgtgt aaccttgaac 1200 tacacaattc tccttcggga cctcaatttc cactttgtaa aatgagggtg gaggtgggaa 1260 taggatctcg aggagactat tggcatatga ttccaaggac tccagtgcct tttgaatggg 1320 cagaggtgag agagagagag agaaagagag agaatgaatg cagttgcatt gattcagtgc 1380 caaggtcact tccagaattc agagttgtga tgctctcttc tgacagccaa agatgaaaaa 1440 caaacagaaa aaaaaaagta aagagtctat ttatggctga catatttacg gctgacaaac 1500 tcctggaaga agctatgctg cttcccagcc tggcttcccc ggatgtttgg ctacctccac 1560 ccctccatct caaagaaata acatcatcca ttggggtaga aaaggagagg gtccgagggt 1620 ggtgggaggg atagaaatca catccgcccc aacttcccaa agagcagcat ccctcccccg 1680 acccatagcc atgttttaaa gtcaccttcc gaagagaagt gaaaggttca aggacactgg 1740 ccttgcaggc ccgagggagc agccatcaca aactcacaga ccagcacatc ccttttgaga 1800 caccgccttc tgcccaccac tcacggacac atttctgcct agaaaacagc ttcttactgc 1860 tcttacatgt gatggcatat cttacactaa aagaatatta ttgggggaaa aactacaagt 1920 gctgtacata tgctgagaaa ctgcagagca taatagctgc cacccaaaaa tctttttgaa 1980 aatcatttcc agacaacctc ttactttctg tgtagttttt aattgttaaa aaaaaaaagt 2040 tttaaacaga agcacatgac atatgaaagc ctgcaggact ggtcgttttt ttggcaattc 2100 ttccacgtgg gacttgtcca caagaatgaa agtagtggtt tttaaagagt taagttacat 2160 atttattttc tcacttaagt tatttatgca aaagtttttc ttgtagagaa tgacaatgtt 2220 aatattgctt tatgaattaa cagtctgttc ttccagagtc cagagacatt gttaataaag 2280 acaatgaatc atgaccgaaa g 2301 8 213 PRT Homo sapien 8 Met Gln Leu Pro Leu Ala Leu Cys Leu Val Cys Leu Leu Val His Thr 1 5 10 15 Ala Phe Arg Val Val Glu Gly Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 Ala Thr Glu Ile Ile Arg Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser 115 120 125 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 145 150 155 160 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 Ser Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly 180 185 190 Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu 195 200 205 Leu Glu Asn Ala Tyr 210 9 642 DNA Cercopithecus pygerythrus 9 atgcagctcc cactggccct gtgtcttgtc tgcctgctgg tacacgcagc cttccgtgta 60 gtggagggcc aggggtggca ggccttcaag aatgatgcca cggaaatcat ccccgagctc 120 ggagagtacc ccgagcctcc accggagctg gagaacaaca agaccatgaa ccgggcggag 180 aatggagggc ggcctcccca ccaccccttt gagaccaaag acgtgtccga gtacagctgc 240 cgagagctgc acttcacccg ctacgtgacc gatgggccgt gccgcagcgc caagccagtc 300 accgagttgg tgtgctccgg ccagtgcggc ccggcacgcc tgctgcccaa cgccatcggc 360 cgcggcaagt ggtggcgccc gagtgggccc gacttccgct gcatccccga ccgctaccgc 420 gcgcagcgtg tgcagctgct gtgtcccggt ggtgccgcgc cgcgcgcgcg caaggtgcgc 480 ctggtggcct cgtgcaagtg caagcgcctc acccgcttcc acaaccagtc ggagctcaag 540 gacttcggtc ccgaggccgc tcggccgcag aagggccgga agccgcggcc ccgcgcccgg 600 ggggccaaag ccaatcaggc cgagctggag aacgcctact ag 642 10 213 PRT Cercopithecus pygerythrus 10 Met Gln Leu Pro Leu Ala Leu Cys Leu Val Cys Leu Leu Val His Ala 1 5 10 15 Ala Phe Arg Val Val Glu Gly Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 Ala Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser 115 120 125 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 Gln Leu Leu Cys Pro Gly Gly Ala Ala Pro Arg Ala Arg Lys Val Arg 145 150 155 160 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 Ser Glu Leu Lys Asp Phe Gly Pro Glu Ala Ala Arg Pro Gln Lys Gly 180 185 190 Arg Lys Pro Arg Pro Arg Ala Arg Gly Ala Lys Ala Asn Gln Ala Glu 195 200 205 Leu Glu Asn Ala Tyr 210 11 638 DNA Mus musculus 11 atgcagccct cactagcccc gtgcctcatc tgcctacttg tgcacgctgc cttctgtgct 60 gtggagggcc aggggtggca agccttcagg aatgatgcca cagaggtcat cccagggctt 120 ggagagtacc ccgagcctcc tcctgagaac aaccagacca tgaaccgggc ggagaatgga 180 ggcagacctc cccaccatcc ctatgacgcc aaaggtgtgt ccgagtacag ctgccgcgag 240 ctgcactaca cccgcttcct gacagacggc ccatgccgca gcgccaagcc ggtcaccgag 300 ttggtgtgct ccggccagtg cggccccgcg cggctgctgc ccaacgccat cgggcgcgtg 360 aagtggtggc gcccgaacgg accggatttc cgctgcatcc cggatcgcta ccgcgcgcag 420 cgggtgcagc tgctgtgccc cgggggcgcg gcgccgcgct cgcgcaaggt gcgtctggtg 480 gcctcgtgca agtgcaagcg cctcacccgc ttccacaacc agtcggagct caaggacttc 540 gggccggaga ccgcgcggcc gcagaagggt cgcaagccgc ggcccggcgc ccggggagcc 600 aaagccaacc aggcggagct ggagaacgcc tactagag 638 12 211 PRT Mus musculus 12 Met Gln Pro Ser Leu Ala Pro Cys Leu Ile Cys Leu Leu Val His Ala 1 5 10 15 Ala Phe Cys Ala Val Glu Gly Gln Gly Trp Gln Ala Phe Arg Asn Asp 20 25 30 Ala Thr Glu Val Ile Pro Gly Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 Glu Asn Asn Gln Thr Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro 50 55 60 His His Pro Tyr Asp Ala Lys Asp Val Ser Glu Tyr Ser Cys Arg Glu 65 70 75 80 Leu His Tyr Thr Arg Phe Leu Thr Asp Gly Pro Cys Arg Ser Ala Lys 85 90 95 Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala Arg Leu 100 105 110 Leu Pro Asn Ala Ile Gly Arg Val Lys Trp Trp Arg Pro Asn Gly Pro 115 120 125 Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu 130 135 140 Leu Cys Pro Gly Gly Ala Ala Pro Arg Ser Arg Lys Val Arg Leu Val 145 150 155 160 Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu 165 170 175 Leu Lys Asp Phe Gly Pro Glu Thr Ala Arg Pro Gln Lys Gly Arg Lys 180 185 190 Pro Arg Pro Gly Ala Arg Gly Ala Lys Ala Asn Gln Ala Glu Leu Glu 195 200 205 Asn Ala Tyr 210 13 674 DNA Rattus norvegicus 13 gaggaccgag tgcccttcct ccttctggca ccatgcagct ctcactagcc ccttgccttg 60 cctgcctgct tgtacatgca gccttcgttg ctgtggagag ccaggggtgg caagccttca 120 agaatgatgc cacagaaatc atcccgggac tcagagagta cccagagcct cctcaggaac 180 tagagaacaa ccagaccatg aaccgggccg agaacggagg cagacccccc caccatcctt 240 atgacaccaa agacgtgtcc gagtacagct gccgcgagct gcactacacc cgcttcgtga 300 ccgacggccc gtgccgcagt gccaagccgg tcaccgagtt ggtgtgctcg ggccagtgcg 360 gccccgcgcg gctgctgccc aacgccatcg ggcgcgtgaa gtggtggcgc ccgaacggac 420 ccgacttccg ctgcatcccg gatcgctacc gcgcgcagcg ggtgcagctg ctgtgccccg 480 gcggcgcggc gccgcgctcg cgcaaggtgc gtctggtggc ctcgtgcaag tgcaagcgcc 540 tcacccgctt ccacaaccag tcggagctca aggacttcgg acctgagacc gcgcggccgc 600 agaagggtcg caagccgcgg ccccgcgccc ggggagccaa agccaaccag gcggagctgg 660 agaacgccta ctag 674 14 213 PRT Rattus norvegicus 14 Met Gln Leu Ser Leu Ala Pro Cys Leu Ala Cys Leu Leu Val His Ala 1 5 10 15 Ala Phe Val Ala Val Glu Ser Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 Ala Thr Glu Ile Ile Pro Gly Leu Arg Glu Tyr Pro Glu Pro Pro Gln 35 40 45 Glu Leu Glu Asn Asn Gln Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 Pro Pro His His Pro Tyr Asp Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 Arg Glu Leu His Tyr Thr Arg Phe Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 Arg Leu Leu Pro Asn Ala Ile Gly Arg Val Lys Trp Trp Arg Pro Asn 115 120 125 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 Gln Leu Leu Cys Pro Gly Gly Ala Ala Pro Arg Ser Arg Lys Val Arg 145 150 155 160 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 Ser Glu Leu Lys Asp Phe Gly Pro Glu Thr Ala Arg Pro Gln Lys Gly 180 185 190 Arg Lys Pro Arg Pro Arg Ala Arg Gly Ala Lys Ala Asn Gln Ala Glu 195 200 205 Leu Glu Asn Ala Tyr 210 15 532 DNA Bos torus 15 agaatgatgc cacagaaatc atccccgagc tgggcgagta ccccgagcct ctgccagagc 60 tgaacaacaa gaccatgaac cgggcggaga acggagggag acctccccac cacccctttg 120 agaccaaaga cgcctccgag tacagctgcc gggagctgca cttcacccgc tacgtgaccg 180 atgggccgtg ccgcagcgcc aagccggtca ccgagctggt gtgctcgggc cagtgcggcc 240 cggcgcgcct gctgcccaac gccatcggcc gcggcaagtg gtggcgccca agcgggcccg 300 acttccgctg catccccgac cgctaccgcg cgcagcgggt gcagctgttg tgtcctggcg 360 gcgcggcgcc gcgcgcgcgc aaggtgcgcc tggtggcctc gtgcaagtgc aagcgcctca 420 ctcgcttcca caaccagtcc gagctcaagg acttcgggcc cgaggccgcg cggccgcaaa 480 cgggccggaa gctgcggccc cgcgcccggg gcaccaaagc cagccgggcc ga 532 16 176 PRT Bos torus 16 Asn Asp Ala Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro 1 5 10 15 Leu Pro Glu Leu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly 20 25 30 Arg Pro Pro His His Pro Phe Glu Thr Lys Asp Ala Ser Glu Tyr Ser 35 40 45 Cys Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg 50 55 60 Ser Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro 65 70 75 80 Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro 85 90 95 Ser Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg 100 105 110 Val Gln Leu Leu Cys Pro Gly Gly Ala Ala Pro Arg Ala Arg Lys Val 115 120 125 Arg Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn 130 135 140 Gln Ser Glu Leu Lys Asp Phe Gly Pro Glu Ala Ala Arg Pro Gln Thr 145 150 155 160 Gly Arg Lys Leu Arg Pro Arg Ala Arg Gly Thr Lys Ala Ser Arg Ala 165 170 175 17 35828 DNA Mus musculus misc_feature (1)..(35828) n = A,T,C or G 17 cgcgttttgg tgagcagcaa tattgcgctt cgatgagcct tggcgttgag attgatacct 60 ctgctgcaca aaaggcaatc gaccgagctg gaccagcgca ttcgtgacac cgtctccttc 120 gaacttattc gcaatggagt gtcattcatc aaggacngcc tgatcgcaaa tggtgctatc 180 cacgcagcgg caatcgaaaa ccctcagccg gtgaccaata tctacaacat cagccttggt 240 atcctgcgtg atgagccagc gcagaacaag gtaaccgtca gtgccgataa gttcaaagtt 300 aaacctggtg ttgataccaa cattgaaacg ttgatcgaaa acgcgctgaa aaacgctgct 360 gaatgtgcgg cgctggatgt cacaaagcaa atggcagcag acaagaaagc gatggatgaa 420 ctggcttcct atgtccgcac ggccatcatg atggaatgtt tccccggtgg tgttatctgg 480 cagcagtgcc gtcgatagta tgcaattgat aattattatc atttgcgggt cctttccggc 540 gatccgcctt gttacggggc ggcgacctcg cgggttttcg ctatttatga aaattttccg 600 gtttaaggcg tttccgttct tcttcgtcat aacttaatgt ttttatttaa aataccctct 660 gaaaagaaag gaaacgacag gtgctgaaag cgagcttttt ggcctctgtc gtttcctttc 720 tctgtttttg tccgtggaat gaacaatgga agtcaacaaa aagcagagct tatcgatgat 780 aagcggtcaa acatgagaat tcgcggccgc ataatacgac tcactatagg gatcgacgcc 840 tactccccgc gcatgaagcg gaggagctgg actccgcatg cccagagacg ccccccaacc 900 cccaaagtgc ctgacctcag cctctaccag ctctggcttg ggcttgggcg gggtcaaggc 960 taccacgttc tcttaacagg tggctgggct gtctcttggc cgcgcgtcat gtgacagctg 1020 cctagttctg cagtgaggtc accgtggaat gtctgccttc gttgccatgg caacgggatg 1080 acgttacaat ctgggtgtgg agcttttcct gtccgtgtca ggaaatccaa ataccctaaa 1140 ataccctaga agaggaagta gctgagccaa ggctttcctg gcttctccag ataaagtttg 1200 acttagatgg aaaaaaacaa aatgataaag acccgagcca tctgaaaatt cctcctaatt 1260 gcaccactag gaaatgtgta tattattgag ctcgtatgtg ttcttatttt aaaaagaaaa 1320 ctttagtcat gttattaata agaatttctc agcagtggga gagaaccaat attaacacca 1380 agataaaagt tggcatgatc cacattgcag gaagatccac gttgggtttt catgaatgtg 1440 aagaccccat ttattaaagt cctaagctct gtttttgcac actaggaagc gatggccggg 1500 atggctgagg ggctgtaagg atctttcaat gtcttacatg tgtgtttcct gtcctgcacc 1560 taggacctgc tgcctagcct gcagcagagc cagaggggtt tcacatgatt agtctcagac 1620 acttgggggc aggttgcatg tactgcatcg cttatttcca tacggagcac ctactatgtg 1680 tcaaacacca tatggtgttc actcttcaga acggtggtgg tcatcatggt gcatttgctg 1740 acggttggat tggtggtaga gagctgagat atatggacgc actcttcagc attctgtcaa 1800 cgtggctgtg cattcttgct cctgagcaag tggctaaaca gactcacagg gtcagcctcc 1860 agctcagtcg ctgcatagtc ttagggaacc tctcccagtc ctccctacct caactatcca 1920 agaagccagg gggcttggcg gtctcaggag cctgcttgct gggggacagg ttgttgagtt 1980 ttatctgcag taggttgcct aggcatagtg tcaggactga tggctgcctt ggagaacaca 2040 tcctttgccc tctatgcaaa tctgaccttg acatgggggc gctgctcagc tgggaggatc 2100 aactgcatac ctaaagccaa gcctaaagct tcttcgtcca cctgaaactc ctggaccaag 2160 gggcttccgg cacatcctct caggccagtg agggagtctg tgtgagctgc actttccaat 2220 ctcagggcgt gagaggcaga gggaggtggg ggcagagcct tgcagctctt tcctcccatc 2280 tggacagcgc tctggctcag cagcccatat gagcacaggc acatccccac cccaccccca 2340 cctttcctgt cctgcagaat ttaggctctg ttcacggggg gggggggggg ggggcagtcc 2400 tatcctctct taggtagaca ggactctgca ggagacactg ctttgtaaga tactgcagtt 2460 taaatttgga tgttgtgagg ggaaagcgaa gggcctcttt gaccattcag tcaaggtacc 2520 ttctaactcc catcgtattg gggggctact ctagtgctag acattgcaga gagcctcaga 2580 actgtagtta ccagtgtggt aggattgatc cttcagggag cctgacatgt gacagttcca 2640 ttcttcaccc agtcaccgaa catttattca gtacctaccc cgtaacaggc accgtagcag 2700 gtactgaggg acggaccact caaagaactg acagaccgaa gccttggaat ataaacacca 2760 aagcatcagg ctctgccaac agaacactct ttaacactca ggccctttaa cactcaggac 2820 ccccaccccc accccaagca gttggcactg ctatccacat tttacagaga ggaaaaacta 2880 ggcacaggac gatataagtg gcttgcttaa gcttgtctgc atggtaaatg gcagggctgg 2940 attgagaccc agacattcca actctagggt ctatttttct tttttctcgt tgttcgaatc 3000 tgggtcttac tgggtaaact caggctagcc tcacactcat atccttctcc catggcttac 3060 gagtgctagg attccaggtg tgtgctacca tgtctgactc cctgtagctt gtctatacca 3120 tcctcacaac ataggaattg tgatagcagc acacacaccg gaaggagctg gggaaatccc 3180 acagagggct ccgcaggatg acaggcgaat gcctacacag aaggtgggga agggaagcag 3240 agggaacagc atgggcgtgg gaccacaagt ctatttgggg aagctgccgg taaccgtata 3300 tggctggggt gaggggagag gtcatgagat gaggcaggaa gagccacagc aggcagcggg 3360 tacgggctcc ttattgccaa gaggctcgga tcttcctcct cttcctcctt ccggggctgc 3420 ctgttcattt tccaccactg cctcccatcc aggtctgtgg ctcaggacat cacccagctg 3480 cagaaactgg gcatcaccca cgtcctgaat gctgccgagg gcaggtcctt catgcacgtc 3540 aacaccagtg ctagcttcta cgaggattct ggcatcacct acttgggcat caaggccaat 3600 gatacgcagg agttcaacct cagtgcttac tttgaaaggg ccacagattt cattgaccag 3660 gcgctggccc ataaaaatgg taaggaacgt acattccggc acccatggag cgtaagccct 3720 ctgggacctg cttcctccaa agaggccccc acttgaaaaa ggttccagaa agatcccaaa 3780 atatgccacc aactagggat taagtgtcct acatgtgagc cgatgggggc cactgcatat 3840 agtctgtgcc atagacatga caatggataa taatatttca gacagagagc aggagttagg 3900 tagctgtgct cctttccctt taattgagtg tgcccatttt tttattcatg tatgtgtata 3960 catgtgtgtg cacacatgcc ataggttgat actgaacacc gtcttcaatc gttccccacc 4020 ccaccttatt ttttgaggca gggtctcttc cctgatcctg gggctcattg gtttatctag 4080 gctgctggcc agtgagctct ggagttctgc ttttctctac ctccctagcc ctgggactgc 4140 aggggcatgt gctgggccag gcttttatgt cgcgttgggg atctgaactt aggtccctag 4200 gcctgagcac cgtaaagact ctgccacatc cccagcctgt ttgagcaagt gaaccattcc 4260 ccagaattcc cccagtgggg ctttcctacc cttttattgg ctaggcattc atgagtggtc 4320 acctcgccag aggaatgagt ggccacgact ggctcagggt cagcagccta gagatactgg 4380 gttaagtctt cctgccgctc gctccctgca gccgcagaca gaaagtagga ctgaatgaga 4440 gctggctagt ggtcagacag gacagaaggc tgagagggtc acagggcaga tgtcagcaga 4500 gcagacaggt tctccctctg tgggggaggg gtggcccact gcaggtgtaa ttggccttct 4560 ttgtgctcca tagaggcttc ctgggtacac agcagcttcc ctgtcctggt gattcccaaa 4620 gagaactccc taccactgga cttacagaag ttctattgac tggtgtaacg gttcaacagc 4680 tttggctctt ggtggacggt gcatactgct gtatcagctc aagagctcat tcacgaatga 4740 acacacacac acacacacac acacacacac acacaagcta attttgatat gccttaacta 4800 gctcagtgac tgggcatttc tgaacatccc tgaagttagc acacatttcc ctctggtgtt 4860 cctggcttaa caccttctaa atctatattt tatctttgct gccctgttac cttctgagaa 4920 gcccctaggg ccacttccct tcgcacctac attgctggat ggtttctctc ctgcagctct 4980 taaatctgat ccctctgcct ctgagccatg ggaacagccc aataactgag ttagacataa 5040 aaacgtctct agccaaaact tcagctaaat ttagacaata aatcttactg gttgtggaat 5100 ccttaagatt cttcatgacc tccttcacat ggcacgagta tgaagcttta ttacaattgt 5160 ttattgatca aactaactca taaaaagcca gttgtctttc acctgctcaa ggaaggaaca 5220 aaattcatcc ttaactgatc tgtgcacctt gcacaatcca tacgaatatc ttaagagtac 5280 taagattttg gttgtgagag tcacatgtta cagaatgtac agctttgaca aggtgcatcc 5340 ttgggatgcc gaagtgacct gctgttccag ccccctacct tctgaggctg ttttggaagc 5400 aatgctctgg aagcaacttt aggaggtagg atgctggaac agcgggtcac ttcagcatcc 5460 cgatgacgaa tcccgtcaaa gctgtacatt ctgtaacaga ctgggaaagc tgcagacttt 5520 aaggccaggg ccctatggtc cctcttaatc cctgtcacac ccaacccgag cccttctcct 5580 ccagccgttc tgtgcttctc actctggata gatggagaac acggccttgc tagttaaagg 5640 agtgaggctt cacccttctc acatggcagt ggttggtcat cctcattcag ggaactctgg 5700 ggcattctgc ctttacttcc tctttttgga ctacagggaa tatatgctga cttgttttga 5760 ccttgtgtat ggggagactg gatctttggt ctggaatgtt tcctgctagt ttttccccat 5820 cctttggcaa accctatcta tatcttacca ctaggcatag tggccctcgt tctggagcct 5880 gccttcaggc tggttctcgg ggaccatgtc cctggtttct ccccagcata tggtgttcac 5940 agtgttcact gcgggtggtt gctgaacaaa gcggggattg catcccagag ctccggtgcc 6000 ttgtgggtac actgctaaga taaaatggat actggcctct ctctgaccac ttgcagagct 6060 ctggtgcctt gtgggtacac tgctaagata aaatggatac tggcctctct ctatccactt 6120 gcaggactct agggaacagg aatccattac tgagaaaacc aggggctagg agcagggagg 6180 tagctgggca gctgaagtgc ttggcgacta accaatgaat accagagttt ggatctctag 6240 aatactctta aaatctgggt gggcagagtg gcctgcctgt aatcccagaa ctcgggaggc 6300 ggagacaggg aatcatcaga gcaaactggc taaccagaat agcaaaacac tgagctctgg 6360 gctctgtgag agatcctgcc ttaacatata agagagagaa taaaacattg aagaagacag 6420 tagatgccaa ttttaagccc ccacatgcac atggacaagt gtgcgtttga acacacatat 6480 gcactcatgt gaaccaggca tgcacactcg ggcttatcac acacataatt tgaaagagag 6540 agtgagagag gagagtgcac attagagttc acaggaaagt gtgagtgagc acacccatgc 6600 acacagacat gtgtgccagg gagtaggaaa ggagcctggg tttgtgtata agagggagcc 6660 atcatgtgtt tctaaggagg gcgtgtgaag gaggcgttgt gtgggctggg actggagcat 6720 ggttgtaact gagcatgctc cctgtgggaa acaggagggt ggccaccctg cagagggtcc 6780 cactgtccag cgggatcagt aaaagcccct gctgagaact ttaggtaata gccagagaga 6840 gaaaggtagg aaagtggggg gactcccatc tctgatgtag gaggatctgg gcaagtagag 6900 gtgcgtttga ggtagaaaga ggggtgcaga ggagatgctc ttaattctgg gtcagcagtt 6960 tctttccaaa taatgcctgt gaggaggtgt aggtggtggc cattcactca ctcagcagag 7020 ggatgatgat gcccggtgga tgctggaaat ggccgagcat caaccctggc tctggaagaa 7080 ctccatcttt cagaaggaga gtggatctgt gtatggccag cggggtcaca ggtgcttggg 7140 gcccctgggg gactcctagc actgggtgat gtttatcgag tgctcttgtg tgccaggcac 7200 tggcctgggg ctttgtttct gtctctgttt tgtttcgttt tttgagacag actcttgcta 7260 tgtatccgtg tcaatcttgg aatctcactg catagcccag gctgcggaga gaggggaggg 7320 caataggcct tgtaagcaag ccacacttca gagactagac tccaccctgc gaatgatgac 7380 aggtcagagc tgagttccgg aagatttttt ttccagctgc caggtggagt gtggagtggc 7440 agctagcggc aagggtagag ggcgagctcc ctgtgcagga gaaatgcaag caagagatgg 7500 caagccagtg agttaagcat tctgtgtggg gagcaggtgg atgaagagag aggctgggct 7560 ttcgcctctg gggggggggt gaggggtggg gatgaggtga gaggagggca gctccctgca 7620 gtgtgatgag atttttcctg acagtgacct ttggcctctc cctcccccac ttcccttctt 7680 tcctttcttc ccaccattgc tttccttgtc cttgagaaat tctgagtttc cacttcactg 7740 gtgatgcaga cggaaacaga agccgtgtgt gtgtgtgtgt gtgtgtgtgt gtgtgtgtgt 7800 gtgtgtgtgt ttgtgtgtat gtgtgtgtgt gtgtttgtgt gtatgtgtgt cagtgggaat 7860 ggctcatagt ctgcaggaag gtgggcagga aggaataagc tgtaggctga ggcagtgtgg 7920 gatgcaggga gagaggagag gagggatacc agagaaggaa attaagggag ctacaagagg 7980 gcattgttgg ggtgtgtgtg tgtgtgtgtt gtttatattt gtattggaaa tacattcttt 8040 taaaaaatac ttatccattt atttattttt atgtgcacgt gtgtgtgcct gcatgagttc 8100 atgtgtgcca cgtgtgtgcg ggaacccttg gaggccacaa gggggcatct gatcccctgg 8160 aactggagtt ggaggaggtt gtgagtcccc tgacatgttt gctgggaact gaaccccggt 8220 cctatgcaag agcaggaagt gcagttatct gctgagccat ctctccagtc ctgaaatcca 8280 ttctcttaaa atacacgtgg cagagacatg atgggattta cgtatggatt taatgtggcg 8340 gtcattaagt tccggcacag gcaagcacct gtaaagccat caccacaacc gcaacagtga 8400 atgtgaccat cacccccatg ttcttcatgt cccctgtccc ctccatcctc cattctcaag 8460 cacctcttgc tctgcctctg tcgctggaga acagtgtgca tctgcacact cttatgtcag 8520 tgaagtcaca cagcctgcac cccttcctgg tctgagtatt tgggttctga ctctgctatc 8580 acacactact gtactgcatt ctctcgctct ctttttttaa acatattttt atttgtttgt 8640 gtgtatgcac atgtgccaca tgtgtacaga tactatggag gccagaagag gccatggccg 8700 tccctggagc tggagttaca ggcagcgtgt gagctgcctg gtgtgggtgc tgggaaccaa 8760 acttgaatct aaagcaagca cttttaactg ctgaggcagc tctcagtacc cttcttcatt 8820 tctccgcctg ggttccattg tatggacaca tgtagctaga atatcttgct tatctaatta 8880 tgtacattgt tttgtgctaa gagagagtaa tgctctatag cctgagctgg cctcaacctt 8940 gccatcctcc tgcctcagcc tcctcctcct gagtgctagg atgacaggcg agtggtaact 9000 tacatggttt catgttttgt tcaagactga aggataacat tcatacagag aaggtctggg 9060 tcacaaagtg tgcagttcac tgaatggcac aacccgtgat caagaaacaa aactcagggg 9120 ctggagagat ggcactgact gctcttccag aggtccggag ttcaattccc agcaaccaca 9180 tggtggctca cagccatcta taacgagatc tgacgccctc ttctggtgtg tctgaagaca 9240 gctacagtgt actcacataa aataaataaa tctttaaaac acacacacac acacaattac 9300 caccccagaa agcccactcc atgttccctc ccacgtctct gcctacagta ctcccaggtt 9360 accactgttc aggcttctaa caacctggtt tacttgggcc tcttttctgc tctgtggagc 9420 cacacatttg tgtgcctcat acacgttctt tctagtaagt tgcatattac tctgcgtttt 9480 tacatgtatt tatttattgt agttgtgtgt gcgtgtgggc ccatgcatgg cacagtgtgt 9540 ggggatgtca gagtattgtg aacaggggac agttcttttc ttcaatcatg tgggttccag 9600 aggttgaact caggtcatca tgtgtggcag caaatgcctt tacccactga gacatctcca 9660 tattcttttt ttttcccctg aggtgggggc ttgttccata gcccaaactg gctttgcact 9720 tgcagttcaa agtgactccc tgtctccacc tcttagagta ttggaattac gatgtgtact 9780 accacacctg actggatcat taattctttg atgggggcgg ggaagcgcac atgctgcagg 9840 tgaagggatg actggactgg acatgagcgt ggaagccaga gaacagcttc agtctaatgc 9900 tctcccaact gagctatttc ggtttgccag agaacaactt acagaaagtt ctcagtgcca 9960 tgtggattcg gggttggagt tcaactcatc agcttgacat tggctcctct acccactgag 10020 ccttctcact actctctacc tagatcatta attctttttt aaaaagactt attagggggc 10080 tggagagatg gctcagccgt taagagcacc gaatgccctt ccagaggtcc tgagttcaat 10140 tcccagcatg ccattgctgg gcagtagggg gcgcaggtgt tcaacgtgag tagctgttgc 10200 cagttttccg cggtggagaa cctcttgaca ccctgctgtc cctggtcatt ctgggtgggt 10260 gcatggtgat atgcttgttg tatggaagac tttgactgtt acagtgaagt tgggcttcca 10320 cagttaccac gtctcccctg tttcttgcag gccgggtgct tgtccattgc cgcgagggct 10380 acagccgctc cccaacgcta gttatcgcct acctcatgat gcggcagaag atggacgtca 10440 agtctgctct gagtactgtg aggcagaatc gtgagatcgg ccccaacgat ggcttcctgg 10500 cccaactctg ccagctcaat gacagactag ccaaggaggg caaggtgaaa ctctagggtg 10560 cccacagcct cttttgcaga ggtctgactg ggagggccct ggcagccatg tttaggaaac 10620 acagtatacc cactccctgc accaccagac acgtgcccac atctgtccca ctctggtcct 10680 cgggggccac tccaccctta gggagcacat gaagaagctc cctaagaagt tctgctcctt 10740 agccatcctt tcctgtaatt tatgtctctc cctgaggtga ggttcaggtt tatgtccctg 10800 tctgtggcat agatacatct cagtgaccca gggtgggagg gctatcaggg tgcatggccc 10860 gggacacggg cactcttcat gacccctccc ccacctgggt tcttcctgtg tggtccagaa 10920 ccacgagcct ggtaaaggaa ctatgcaaac acaggccctg acctccccat gtctgttcct 10980 ggtcctcaca gcccgacacg ccctgctgag gcagacgaat gacattaagt tctgaagcag 11040 agtggagata gattagtgac tagatttcca aaaagaagga aaaaaaaggc tgcattttaa 11100 aattatttcc ttagaattaa agatactaca taggggccct tgggtaagca aatccatttt 11160 tcccagaggc tatcttgatt ctttggaatg tttaaagtgt gccttgccag agagcttacg 11220 atctatatct gctgcttcag agccttccct gaggatggct ctgttccttt gcttgttaga 11280 agagcgatgc cttgggcagg gtttccccct tttcagaata cagggtgtaa agtccagcct 11340 attacaaaca aacaaacaaa caaacaaaca aaggacctcc atttggagaa ttgcaaggat 11400 tttatcctga attatagtgt tggtgagttc aagtcatcac gccaagtgct tgccatcctg 11460 gttgctattc taagaataat taggaggagg aacctagcca attgcagctc atgtccgtgg 11520 gtgtgtgcac gggtgcatat gttggaaggg gtgcctgtcc ccttggggac agaaggaaaa 11580 tgaaaggccc ctctgctcac cctggccatt tacgggaggc tctgctggtt ccacggtgtc 11640 tgtgcaggat cctgaaactg actcgctgga cagaaacgag acttggcggc accatgagaa 11700 tggagagaga gagagcaaag aaagaaacag cctttaaaag aactttctaa gggtggtttt 11760 tgaacctcgc tggaccttgt atgtgtgcac atttgccaga gattgaacat aatcctcttg 11820 ggacttcacg ttctcattat ttgtatgtct ccggggtcac gcagagccgt cagccaccac 11880 cccagcaccc ggcacatagg cgtctcataa aagcccattt tatgagaacc agagctgttt 11940 gagtaccccg tgtatagaga gagttgttgt cgtggggcac ccggatccca gcagcctggt 12000 tgcctgcctg taggatgtct tacaggagtt tgcagagaaa ccttccttgg agggaaagaa 12060 atatcaggga tttttgttga atatttcaaa ttcagcttta agtgtaagac tcagcagtgt 12120 tcatggttaa ggtaaggaac atgccttttc cagagctgct gcaagaggca ggagaagcag 12180 acctgtctta ggatgtcact cccagggtaa agacctctga tcacagcagg agcagagctg 12240 tgcagcctgg atggtcattg tcccctattc tgtgtgacca cagcaaccct ggtcacatag 12300 ggctggtcat cctttttttt tttttttttt tttttttttg gcccagaatg aagtgaccat 12360 agccaagttg tgtacctcag tctttagttt ccaagcggct ctcttgctca atacaatgtg 12420 catttcaaaa taacactgta gagttgacag aactggttca tgtgttatga gagaggaaaa 12480 gagaggaaag aacaaaacaa aacaaaacac cacaaaccaa aaacatctgg gctagccagg 12540 catgattgca atgtctacag gcccagttca tgagaggcag agacaggaag accgccgaaa 12600 ggtcaaggat agcatggtct acgtatcgag actccagcca gggctacggt cccaagatcc 12660 taggttttgg attttgggct ttggtttttg agacagggtt tctctgtgta gccctggctg 12720 tcctggaact cgctctgtag accaggctgg cctcaaactt agagatctgc ctgactctgc 12780 ctttgagggc tgggacgaat gccaccactg cccaactaag attccattaa aaaaaaaaaa 12840 agttcaagat aattaagagt tgccagctcg ttaaagctaa gtagaagcag tctcaggcct 12900 gctgcttgag gctgttcttg gcttggacct gaaatctgcc cccaacagtg tccaagtgca 12960 catgactttg agccatctcc agagaaggaa gtgaaaattg tggctcccca gtcgattggg 13020 acacagtctc tctttgtcta ggtaacacat ggtgacacat agcattgaac tctccactct 13080 gagggtgggt ttccctcccc ctgcctcttc tgggttggtc accccatagg acagccacag 13140 gacagtcact agcacctact ggaaacctct ttgtgggaac atgaagaaag agcctttggg 13200 agattcctgg ctttccatta gggctgaaag tacaacggtt cttggttggc tttgcctcgt 13260 gtttataaaa ctagctacta ttcttcaggt aaaataccga tgttgtggaa aagccaaccc 13320 cgtggctgcc cgtgagtagg gggtggggtt gggaatcctg gatagtgttc tatccatgga 13380 aagtggtgga ataggaatta agggtgttcc cccccccccc aacctcttcc tcagacccag 13440 ccactttcta tgacttataa acatccaggt aaaaattaca aacataaaaa tggtttctct 13500 tctcaatctt ctaaagtctg cctgcctttt ccaggggtag gtctgtttct ttgctgttct 13560 attgtcttga gagcacagac taacacttac caaatgaggg aactcttggc ccatactaag 13620 gctcttctgg gctccagcac tcttaagtta ttttaagaat tctcacttgg cctttagcac 13680 acccgccacc cccaagtggg tgtggataat gccatggcca gcagggggca ctgttgaggc 13740 gggtgccttt ccaccttaag ttgcttatag tatttaagat gctaaatgtt ttaatcaaga 13800 gaagcactga tcttataata cgaggataag agattttctc acaggaaatt gtctttttca 13860 taattctttt acaggctttg tcctgatcgt agcatagaga gaatagctgg atatttaact 13920 tgtattccat tttcctctgc cagcgttagg ttaactccgt aaaaagtgat tcagtggacc 13980 gaagaggctc agagggcagg ggatggtggg gtgaggcaga gcactgtcac ctgccaggca 14040 tgggaggtcc tgccatccgg gaggaaaagg aaagtttagc ctctagtcta ccaccagtgt 14100 taacgcactc taaagttgta accaaaataa atgtcttaca ttacaaagac gtctgttttg 14160 tgtttccttt tgtgtgtttg ggctttttat gtgtgcttta taactgctgt ggtggtgctg 14220 ttgttagttt tgaggtagga tctcaggctg gccttgaact tctgatcgcc tgcccctgcc 14280 cctgcccctg cccctgtccc tgcctccaag tgctaggact aaaagcacat gccaccacac 14340 cagtacagca tttttctaac atttaaaaat aatcacctag gggctggaga gagggttcca 14400 gctaagagtg cacactgctc ttgggtagga cctgagttta gttcccagaa cctatactgg 14460 gtggctccag gtccagagga tccaggacct ctggcctcca tgggcatctg ctcttagcac 14520 atacccacat acagatacac acataaaaat aaaatgaagc ctttaaaaac ctcctaaaac 14580 ctagcccttg gaggtacgac tctggaaagc tggcatactg tgtaagtcca tctcatggtg 14640 ttctggctaa cgtaagactt acagagacag aaaagaactc agggtgtgct gggggttggg 14700 atggaggaag agggatgagt agggggagca cggggaactt gggcagtgaa aattctttgc 14760 aggacactag aggaggataa ataccagtca ttgcacccac tactggacaa ctccagggaa 14820 ttatgctggg tgaaaagaga aggccccagg tattggctgc attggctgca tttgcgtaac 14880 atttttttaa attgaaaaga aaaagatgta aatcaaggtt agatgagtgg ttgctgtgag 14940 ctgagagctg gggtgagtga gacatgtgga caactccatc aaaaagcgac agaaagaacg 15000 ggctgtggtg acagctacct ctaatctcca cctccgggag gtgatcaagg ttagccctca 15060 gctagcctgt ggtgcatgag accctgtttc aaaaacttta ataaagaaat aatgaaaaaa 15120 gacatcaggg cagatccttg gggccaaagg cggacaggcg agtctcgtgg taaggtcgtg 15180 tagaagcgga tgcatgagca cgtgccgcag gcatcatgag agagccctag gtaagtaagg 15240 atggatgtga gtgtgtcggc gtcggcgcac tgcacgtcct ggctgtggtg ctggactggc 15300 atctttggtg agctgtggag gggaaatggg tagggagatc ataaaatccc tccgaattat 15360 ttcaagaact gtctattaca attatctcaa aatattaaaa aaaaagaaga attaaaaaac 15420 aaaaaaccta tccaggtgtg gtggtgtgca cctatagcca cgggcacttg gaaagctgga 15480 gcaagaggat ggcgagtttg aaggtatctg gggctgtaca gcaagaccgt cgtccccaaa 15540 ccaaaccaaa cagcaaaccc attatgtcac acaagagtgt ttatagtgag cggcctcgct 15600 gagagcatgg ggtgggggtg ggggtggggg acagaaatat ctaaactgca gtcaataggg 15660 atccactgag accctggggc ttgactgcag cttaaccttg ggaaatgata agggttttgt 15720 gttgagtaaa agcatcgatt actgacttaa cctcaaatga agaaaaagaa aaaaagaaaa 15780 caacaaaagc caaaccaagg ggctggtgag atggctcagt gggtaagagc acccgactgc 15840 tcttccgaag gtccagagtt caaatcccag caaccacatg gtggctcaca accatctgta 15900 acgagatatg atgccctctt ctggtgtgtc tgaagacagc tacagtgtac ttacatataa 15960 taaataaatc ttaaaaaaaa aaaaaaaaaa aaaagccaaa ccgagcaaac caggccccca 16020 aacagaaggc aggcacgacg gcaggcacca cgagccatcc tgtgaaaagg cagggctacc 16080 catgggccga ggagggtcca gagagatagg ctggtaagct cagtttctct gtataccctt 16140 tttcttgttg acactacttc aattacagat aaaataacaa ataaacaaaa tctagagcct 16200 ggccactctc tgctcgcttg atttttcctg ttacgtccag caggtggcgg aagtgttcca 16260 aggacagatc gcatcattaa ggtggccagc ataatctccc atcagcaggt ggtgctgtga 16320 gaaccattat ggtgctcaca gaatcccggg cccaggagct gccctctccc aagtctggag 16380 caataggaaa gctttctggc ccagacaggg ttaacagtcc acattccaga gcaggggaaa 16440 aggagactgg aggtcacaga caaaagggcc agcttctaac aacttcacag ctctggtagg 16500 agagatagat cacccccaac aatggccaca gctggttttg tctgccccga aggaaactga 16560 cttaggaagc aggtatcaga gtccccttcc tgaggggact tctgtctgcc ttgtaaagct 16620 gtcagagcag ctgcattgat gtgtgggtga cagaagatga aaaggaggac ccaggcagat 16680 cgccacagat ggaccggcca cttacaagtc gaggcaggtg gcagagcctt gcagaagctc 16740 tgcaggtgga cgacactgat tcattaccca gttagcatac cacagcgggc taggcggacc 16800 acagcctcct tcccagtctt cctccagggc tggggagtcc tccaaccttc tgtctcagtg 16860 cagcttccgc cagcccctcc tccttttgca cctcaggtgt gaaccctccc tcctctcctt 16920 ctccctgtgg catggccctc ctgctactgc aggctgagca ttggatttct ttgtgcttag 16980 atagacctga gatggctttc tgatttatat atatatatcc atcccttgga tcttacatct 17040 aggacccaga gctgtttgtg ataccataag aggctgggga gatgatatgg taagagtgct 17100 tgctgtacaa gcatgaagac atgagttcga atccccagca accatgtgga aaaataacct 17160 tctaacctca gagttgaggg gaaaggcagg tggattctgg gggcttactg gccagctagc 17220 cagcctaacc taaatgtctc agtcagagat cctgtctcag ggaataactt gggagaatga 17280 ctgagaaaga cacctcctca ggtctcccat gcacccacac agacacacgg ggggggggta 17340 atgtaataag ctaagaaata atgagggaaa tgattttttg ctaagaaatg aaattctgtg 17400 ttggccgcaa gaagcctggc cagggaagga actgcctttg gcacaccagc ctataagtca 17460 ccatgagttc cctggctaag aatcacatgt aatggagccc aggtccctct tgcctggtgg 17520 ttgcctctcc cactggtttt gaagagaaat tcaagagaga tctccttggt cagaattgta 17580 ggtgctgagc aatgtggagc tggggtcaat gggattcctt taaaggcatc cttcccaggg 17640 ctgggtcata cttcaatagt agggtgcttg cacagcaagc gtgagaccct aggttagagt 17700 ccccagaatc tgcccccaac cccccaaaaa ggcatccttc tgcctctggg tgggtggggg 17760 gagcaaacac ctttaactaa gaccattagc tggcaggggt aacaaatgac cttggctaga 17820 ggaatttggt caagctggat tccgccttct gtagaagccc cacttgtttc ctttgttaag 17880 ctggcccaca gtttgttttg agaatgcctg aggggcccag ggagccagac aattaaaagc 17940 caagctcatt ttgatatctg aaaaccacag cctgactgcc ctgcccgtgg gaggtactgg 18000 gagagctggc tgtgtccctg cctcaccaac gccccccccc ccaacacaca ctcctcgggt 18060 cacctgggag gtgccagcag caatttggaa gtttactgag cttgagaagt cttgggaggg 18120 ctgacgctaa gcacacccct tctccacccc cccccacccc acccccgtga ggaggagggt 18180 gaggaaacat gggaccagcc ctgctccagc ccgtccttat tggctggcat gaggcagagg 18240 gggctttaaa aaggcaaccg tatctaggct ggacactgga gcctgtgcta ccgagtgccc 18300 tcctccacct ggcagcatgc agccctcact agccccgtgc ctcatctgcc tacttgtgca 18360 cgctgccttc tgtgctgtgg agggccaggg gtggcaagcc ttcaggaatg atgccacaga 18420 ggtcatccca gggcttggag agtaccccga gcctcctcct gagaacaacc agaccatgaa 18480 ccgggcggag aatggaggca gacctcccca ccatccctat gacgccaaag gtacgggatg 18540 aagaagcaca ttagtggggg ggggggtcct gggaggtgac tggggtggtt ttagcatctt 18600 cttcagaggt ttgtgtgggt ggctagcctc tgctacatca gggcagggac acatttgcct 18660 ggaagaatac tagcacagca ttagaacctg gagggcagca ttggggggct ggtagagagc 18720 acccaaggca gggtggaggc tgaggtcagc cgaagctggc attaacacgg gcatgggctt 18780 gtatgatggt ccagagaatc tcctcctaag gatgaggaca caggtcagat ctagctgctg 18840 accagtgggg aagtgatatg gtgaggctgg atgccagatg ccatccatgg ctgtactata 18900 tcccacatga ccaccacatg aggtaaagaa ggccccagct tgaagatgga gaaaccgaga 18960 ggctcctgag ataaagtcac ctgggagtaa gaagagctga gactggaagc tggtttgatc 19020 cagatgcaag gcaaccctag attgggtttg ggtgggaacc tgaagccagg aggaatccct 19080 ttagttcccc cttgcccagg gtctgctcaa tgagcccaga gggttagcat taaaagaaca 19140 gggtttgtag gtggcatgtg acatgagggg cagctgagtg aaatgtcccc tgtatgagca 19200 caggtggcac cacttgccct gagcttgcac cctgacccca gctttgcctc attcctgagg 19260 acagcagaaa ctgtggaggc agagccagca cagagagatg cctggggtgg gggtgggggt 19320 atcacgcacg gaactagcag caatgaatgg ggtggggtgg cagctggagg gacactccag 19380 agaaatgacc ttgctggtca ccatttgtgt gggaggagag ctcattttcc agcttgccac 19440 cacatgctgt ccctcctgtc tcctagccag taagggatgt ggaggaaagg gccaccccaa 19500 aggagcatgc aatgcagtca cgtttttgca gaggaagtgc ttgacctaag ggcactattc 19560 ttggaaagcc ccaaaactag tccttccctg ggcaaacagg cctcccccac ataccacctc 19620 tgcaggggtg agtaaattaa gccagccaca gaagggtggc aaggcctaca cctcccccct 19680 gttgtgcccc cccccccccc gtgaaggtgc atcctggcct ctgcccctct ggctttggta 19740 ctgggatttt ttttttcctt ttatgtcata ttgatcctga caccatggaa cttttggagg 19800 tagacaggac ccacacatgg attagttaaa agcctcccat ccatctaagc tcatggtagg 19860 agatagagca tgtccaagag aggagggcag gcatcagacc tagaagatat ggctgggcat 19920 ccaacccaat ctccttcccc ggagaacaga ctctaagtca gatccagcca cccttgagta 19980 accagctcaa ggtacacaga acaagagagt ctggtataca gcaggtgcta aacaaatgct 20040 tgtggtagca aaagctatag gttttgggtc agaactccga cccaagtcgc gagtgaagag 20100 cgaaaggccc tctactcgcc accgccccgc ccccacctgg ggtcctataa cagatcactt 20160 tcacccttgc gggagccaga gagccctggc atcctaggta gccccccccg cccccccccc 20220 gcaagcagcc cagccctgcc tttggggcaa gttcttttct cagcctggac ctgtgataat 20280 gagggggttg gacgcgccgc ctttggtcgc tttcaagtct aatgaattct tatccctacc 20340 acctgccctt ctaccccgct cctccacagc agctgtcctg atttattacc ttcaattaac 20400 ctccactcct ttctccatct cctgggatac cgcccctgtc ccagtggctg gtaaaggagc 20460 ttaggaagga ccagagccag gtgtggctag aggctaccag gcagggctgg ggatgaggag 20520 ctaaactgga agagtgtttg gttagtaggc acaaagcctt gggtgggatc cctagtaccg 20580 gagaagtgga gatgggcgct gagaagttca agaccatcca tccttaacta cacagccagt 20640 ttgaggccag cctgggctac ataaaaaccc aatctcaaaa gctgccaatt ctgattctgt 20700 gccacgtagt gcccgatgta atagtggatg aagtcgttga atcctggggc aacctatttt 20760 acagatgtgg ggaaaagcaa ctttaagtac cctgcccaca gatcacaaag aaagtaagtg 20820 acagagctcc agtgtttcat ccctgggttc caaggacagg gagagagaag ccagggtggg 20880 atctcactgc tccccggtgc ctccttccta taatccatac agattcgaaa gcgcagggca 20940 ggtttggaaa aagagagaag ggtggaagga gcagaccagt ctggcctagg ctgcagcccc 21000 tcacgcatcc ctctctccgc agatgtgtcc gagtacagct gccgcgagct gcactacacc 21060 cgcttcctga cagacggccc atgccgcagc gccaagccgg tcaccgagtt ggtgtgctcc 21120 ggccagtgcg gccccgcgcg gctgctgccc aacgccatcg ggcgcgtgaa gtggtggcgc 21180 ccgaacggac cggatttccg ctgcatcccg gatcgctacc gcgcgcagcg ggtgcagctg 21240 ctgtgccccg ggggcgcggc gccgcgctcg cgcaaggtgc gtctggtggc ctcgtgcaag 21300 tgcaagcgcc tcacccgctt ccacaaccag tcggagctca aggacttcgg gccggagacc 21360 gcgcggccgc agaagggtcg caagccgcgg cccggcgccc ggggagccaa agccaaccag 21420 gcggagctgg agaacgccta ctagagcgag cccgcgccta tgcagccccc gcgcgatccg 21480 attcgttttc agtgtaaagc ctgcagccca ggccaggggt gccaaacttt ccagaccgtg 21540 tggagttccc agcccagtag agaccgcagg tccttctgcc cgctgcgggg gatggggagg 21600 gggtggggtt cccgcgggcc aggagaggaa gcttgagtcc cagactctgc ctagccccgg 21660 gtgggatggg ggtctttcta ccctcgccgg acctatacag gacaaggcag tgtttccacc 21720 ttaaagggaa gggagtgtgg aacgaaagac ctgggactgg ttatggacgt acagtaagat 21780 ctactccttc cacccaaatg taaagcctgc gtgggctaga tagggtttct gaccctgacc 21840 tggccactga gtgtgatgtt gggctacgtg gttctctttt ggtacggtct tctttgtaaa 21900 atagggaccg gaactctgct gagattccaa ggattggggt accccgtgta gactggtgag 21960 agagaggaga acaggggagg ggttagggga gagattgtgg tgggcaaccg cctagaagaa 22020 gctgtttgtt ggctcccagc ctcgccgcct cagaggtttg gcttccccca ctccttcctc 22080 tcaaatctgc cttcaaatcc atatctggga tagggaaggc cagggtccga gagatggtgg 22140 aagggccaga aatcacactc ctggcccccc gaagagcagt gtcccgcccc caactgcctt 22200 gtcatattgt aaagggattt tctacacaac agtttaaggt cgttggagga aactgggctt 22260 gccagtcacc tcccatcctt gtcccttgcc aggacaccac ctcctgcctg ccacccacgg 22320 acacatttct gtctagaaac agagcgtcgt cgtgctgtcc tctgagacag catatcttac 22380 attaaaaaga ataatacggg gggggggggc ggagggcgca agtgttatac atatgctgag 22440 aagctgtcag gcgccacagc accacccaca atctttttgt aaatcatttc cagacacctc 22500 ttactttctg tgtagatttt aattgttaaa aggggaggag agagagcgtt tgtaacagaa 22560 gcacatggag gggggggtag gggggttggg gctggtgagt ttggcgaact ttccatgtga 22620 gactcatcca caaagactga aagccgcgtt ttttttttta agagttcagt gacatattta 22680 ttttctcatt taagttattt atgccaacat ttttttcttg tagagaaagg cagtgttaat 22740 atcgctttgt gaagcacaag tgtgtgtggt tttttgtttt ttgttttttc cccgaccaga 22800 ggcattgtta ataaagacaa tgaatctcga gcaggaggct gtggtcttgt tttgtcaacc 22860 acacacaatg tctcgccact gtcatctcac tcccttccct tggtcacaag acccaaacct 22920 tgacaacacc tccgactgct ctctggtagc ccttgtggca atacgtgttt cctttgaaaa 22980 gtcacattca tcctttcctt tgcaaacctg gctctcattc cccagctggg tcatcgtcat 23040 accctcaccc cagcctccct ttagctgacc actctccaca ctgtcttcca aaagtgcacg 23100 tttcaccgag ccagttccct ggtccaggtc atcccattgc tcctccttgc tccagaccct 23160 tctcccacaa agatgttcat ctcccactcc atcaagcccc agtggccctg cggctatccc 23220 tgtctcttca gttagctgaa tctacttgct gacaccacat gaattccttc ccctgtctta 23280 aggttcatgg aactcttgcc tgcccctgaa ccttccagga ctgtcccagc gtctgatgtg 23340 tcctctctct tgtaaagccc caccccacta tttgattccc aattctagat cttcccttgt 23400 tcattccttc acgggatagt gtctcatctg gccaagtcct gcttgatatt gggataaatg 23460 caaagccaag tacaattgag gaccagttca tcattgggcc aagctttttc aaaatgtgaa 23520 ttttacacct atagaagtgt aaaagccttc caaagcagag gcaatgcctg gctcttcctt 23580 caacatcagg gctcctgctt tatgggtctg gtggggtagt acattcataa acccaacact 23640 aggggtgtga aagcaagatg attgggagtt cgaggccaat cttggctatg aggccctgtc 23700 tcaacctctc ctccctccct ccagggtttt gttttgtttt gtttttttga tttgaaactg 23760 caacacttta aatccagtca agtgcatctt tgcgtgaggg gaactctatc cctaatataa 23820 gcttccatct tgatttgtgt atgtgcacac tgggggttga acctgggcct ttgtacctgc 23880 cgggcaagct ctctactgct ctaaacccag ccctcactgg ctttctgttt caactcccaa 23940 tgaattcccc taaatgaatt atcaatatca tgtctttgaa aaataccatt gagtgctgct 24000 ggtgtccctg tggttccaga ttccaggaag gacttttcag ggaatccagg catcctgaag 24060 aatgtcttag agcaggaggc catggagacc ttggccagcc ccacaaggca gtgtggtgca 24120 gagggtgagg atggaggcag gcttgcaatt gaagctgaga cagggtactc aggattaaaa 24180 agcttccccc aaaacaattc caagatcagt tcctggtact tgcacctgtt cagctatgca 24240 gagcccagtg ggcataggtg aagacaccgg ttgtactgtc atgtactaac tgtgcttcag 24300 agccggcaga gacaaataat gttatggtga ccccagggga cagtgattcc agaaggaaca 24360 cagaagagag tgctgctaga ggctgcctga aggagaaggg gtcccagact ctctaagcaa 24420 agactccact cacataaaga cacaggctga gcagagctgg ccgtggatgc agggagccca 24480 tccaccatcc tttagcatgc ccttgtattc ccatcacatg ccagggatga ggggcatcag 24540 agagtccaag tgatgcccaa acccaaacac acctaggact tgctttctgg gacagacaga 24600 tgcaggagag actaggttgg gctgtgatcc cattaccaca aagagggaaa aaacaaaaaa 24660 caaacaaaca aacaaaaaaa aacaaaacaa aacaaaaaaa aacccaaggt ccaaattgta 24720 ggtcaggtta gagtttattt atggaaagtt atattctacc tccatggggt ctacaaggct 24780 ggcgcccatc agaaagaaca aacaacaggc tgatctggga ggggtggtac tctatggcag 24840 ggagcacgtg tgcttggggt acagccagac acggggcttg tattaatcac agggcttgta 24900 ttaataggct gagagtcaag cagacagaga gacagaagga aacacacaca cacacacaca 24960 cacacacaca cacacacaca catgcacaca ccactcactt ctcactcgaa gagcccctac 25020 ttacattcta agaacaaacc attcctcctc ataaaggaga caaagttgca gaaacccaaa 25080 agagccacag ggtccccact ctctttgaaa tgacttggac ttgttgcagg gaagacagag 25140 gggtctgcag aggcttcctg ggtgacccag agccacagac actgaaatct ggtgctgaga 25200 cctgtataaa ccctcttcca caggttccct gaaaggagcc cacattcccc aaccctgtct 25260 cctgaccact gaggatgaga gcacttgggc cttccccatt cttggagtgc accctggttt 25320 ccccatctga gggcacatga ggtctcaggt cttgggaaag ttccacaagt attgaaagtg 25380 ttcttgtttt gtttgtgatt taatttaggt gtatgagtgc ttttgcttga atatatgcct 25440 gtgtagcatt tacaagcctg gtgcctgagg agatcagaag atggcatcag ataccctgga 25500 actggacttg cagacagtta tgagccactg tgtgggtgct aggaacagaa cctggatcct 25560 ccggaagagc agacagccag cgctcttagc cactaagcca tcactgaggt tctttctgtg 25620 gctaaagaga caggagacaa aggagagttt cttttagtca ataggaccat gaatgttcct 25680 cgtaacgtga gactagggca gggtgatccc ccagtgacac cgatggccct gtgtagttat 25740 tagcagctct agtcttattc cttaataagt cccagtttgg ggcaggagat atgtattccc 25800 tgctttgaag tggctgaggt ccagttatct acttccaagt acttgtttct ctttctggag 25860 ttggggaagc tccctgcctg cctgtaaatg tgtccattct tcaaccttag acaagatcac 25920 tttccctgag cagtcaggcc agtccaaagc ccttcaattt agctttcata aggaacaccc 25980 cttttgttgg gtggaggtag cacttgcctt gaatcccagc attaagaagg cagagacagt 26040 cggatctctg tgagttcaca gccagcctgg tctacggagt gagttccaag acagccaggc 26100 ctacacagag aaaccctgtc tcgaaaaaaa caaaaacaaa agaaataaag aaaaagaaaa 26160 caaaaacgaa caaacagaaa aacaagccag agtgtttgtc cccgtatttt attaatcata 26220 tttttgtccc tttgccattt tagactaaaa gactcgggaa agcaggtctc tctctgtttc 26280 tcatccggac acacccagaa ccagatgtat ggaagatggc taatgtgctg cagttgcaca 26340 tctggggctg ggtggattgg ttagatggca tgggctgggt gtggttacga tgactgcagg 26400 agcaaggagt atgtggtgca tagcaaacga ggaagtttgc acagaacaac actgtgtgta 26460 ctgatgtgca ggtatgggca catgcaagca gaagccaagg gacagcctta gggtagtgtt 26520 tccacagacc cctcccccct tttaacatgg gcatctctca ttggcctgga gcttgccaac 26580 tgggctgggc tggctagctt gtaggtccca gggatctgca tatctctgcc tccctagtgc 26640 tgggattaca gtcatatatg agcacacctg gcttttttat gtgggttctg ggctttgaac 26700 ccagatctga gtgcttgcaa ggcaatcggt tgaatgactg cttcatctcc ccagaccctg 26760 ggattctact ttctattaaa gtatttctat taaatcaatg agcccctgcc cctgcactca 26820 gcagttctta ggcctgctga gagtcaagtg gggagtgaga gcaagcctcg agaccccatc 26880 agcgaagcag aggacaaaga aatgaaaact tgggattcga ggctcgggat atggagatac 26940 agaaagggtc agggaaggaa atgaaccaga tgaatagagg caggaagggt agggccctgc 27000 atacatggaa cctggtgtac atgttatctg catggggttt gcattgcaat ggctcttcag 27060 caggttcacc acactgggaa acagaagcca aaaagaagag taggtggtgt tggagtcaga 27120 tactgtcagt catgcctgaa gaaatggaag caattaacga tgcgccgcaa ttaggatatt 27180 agctccctga agaaaggcaa gaagctgggc tgtgggcact gaagggagct ttgaatgatg 27240 tcacattctc tgtatgccta gcagggcagt attggagact gagacttgac ttgtgtgtcc 27300 atatgattcc tccttttcct acagtcatct ggggctcctg agcttcgtcc ttgtccaaga 27360 acctggagct ggcagtgggc agctgcagtg atagatgtct gcaagaaaga tctgaaaaga 27420 gggaggaaga tgaaggaccc agaggaccac cgacctctgc tgcctgacaa agctgcagga 27480 ccagtctctc ctacagatgg gagacagagg cgagagatga atggtcaggg gaggagtcag 27540 agaaaggaga gggtgaggca gagaccaaag gagggaaaca cttgtgctct acagctactg 27600 actgagtacc agctgcgtgg cagacagcca atgccaaggc tcggctgatc atggcacctc 27660 gtgggactcc tagcccagtg ctggcagagg ggagtgctga atggtgcatg gtttggatat 27720 gatctgaatg tggtccagcc ctagtttcct tccagttgct gggataaagc accctgacca 27780 aagctacttt tttgtttgtt tgttttggtt tggttttgtt tggtttttcg aggcagggtt 27840 tctctgtatc accctagctg tcctggaact cactctgtag accaggctgg cctcgaactc 27900 agaaatcccc ctgcctctgc ctcctaagtg ctggaattaa aggcctgcgc caccactgcc 27960 ggcccaaagc tactttaaga gagagagagg aatgtataag tattataatt ccaggttata 28020 gttcattgct gtagaattgg agtcttcata ttccaggtaa tctcccacag acatgccaca 28080 aaacaacctg ttctacgaaa tctctcatgg actcccttcc ccagtaattc taaactgtgt 28140 caaatctaca agaaatagtg acagtcacag tctctaacgt tttgggcatg agtctgaagt 28200 ctcattgcta agtactggga agatgaaaac tttacctagt gtcagcattt ggagcagagc 28260 ctttgggatt tgagatggtc ttttgcagag ctcctaatgg ctacatggag agagggggcc 28320 tgggagagac ccatacacct tttgctgcct tatgtcacct gacctgctcc ttgggaagct 28380 ctagcaagaa ggccttccct ggatcaccca ccaccttgca cctccagaac tcagagccaa 28440 attaaacttt cttgttactg tcgtcaaagc acagtcggtc tgggttgtat cactgtcaat 28500 gggaaacaga cttgcctgga tggataactt gtacattgca taatgtctag aaatgaaaag 28560 tcctatagag aaaaagaaaa ttagctggca cacagataga ggccctggag gaggctggct 28620 ttgtcctccc cgaggaggtg gcgagtaagg tgtaaatgtt catggatgta aatgggccca 28680 tatatgaggg tctggggtaa caagaaggcc tgtgaatata aagcactgaa ggtatgtcta 28740 gtctggagaa ggtcactaca gagagttctc caactcagtg cccatacaca cacacacaca 28800 cacacacaca cacacacaca cacacacaca ccacaaagaa aaaaaggaag aaaaatctga 28860 gagcaagtac agtacttaaa attgtgtgat tgtgtgtgtg actctgatgt cacatgctca 28920 tcttgcccta tgagttgaaa accaaatggc ccctgagagg cataacaacc acactgttgg 28980 ctgtgtgctc acgtttttct taaagcgtct gtctggtttg ctgctagcat caggcagact 29040 tgcagcagac tacatatgct cagccctgaa gtccttctag ggtgcatgtc tcttcagaat 29100 ttcagaaagt catctgtggc tccaggaccg cctgcactct ccctctgccg cgaggctgca 29160 gactctaggc tggggtggaa gcaacgctta cctctgggac aagtataaca tgttggcttt 29220 tctttccctc tgtggctcca acctggacat aaaatagatg caagctgtgt aataaatatt 29280 tcctcccgtc cacttagttc tcaacaataa ctactctgag agcacttatt aataggtggc 29340 ttagacataa gctttggctc attcccccac tagctcttac ttctttaact ctttcaaacc 29400 attctgtgtc ttccacatgg ttagttacct ctccttccat cctggttcgc ttcttccttc 29460 gagtcgccct cagtgtctct aggtgatgct tgtaagatat tctttctaca aagctgagag 29520 tggtggcact ctgggagttc aaagccagcc tgatctacac agcaagctcc aggatatcca 29580 gggcaatgtt gggaaaacct ttctcaaaca aaaagagggg ttcagttgtc aggaggagac 29640 ccatgggtta agaagtctag acgagccatg gtgatgcata cctttcatcc aagcacttag 29700 gaggcaaaga aaggtgaaac tctttgactt tgaggccagc taggttacat agtgataccc 29760 tgcttagtgt gtgtgtgtgt gtgtgtgtgt gtgtgtgtgt gtgtgtaatt taaaagtcta 29820 aaaatgcatt cttttaaaaa tatgtataag tatttgcctg cacatatgta tgtatgtatg 29880 tataccatgt gtgtgtctgg tgctgaagga ctaggcatag actccctaga actagagtca 29940 tagacagttg tgacactccc caacccccca ccatgtgggt gcttgaagct aaactcctgt 30000 cctttgtaaa gcagcaggtg tctatgaacc ctgaaccatc tctccagtct ccagatgtgc 30060 attctcaaag aggagtcctt catatttccc taaactgaac atccttatca gtgagcatcc 30120 tcgagtcacc aaagctactg caaaccctct tagggaacat tcactattca cttctacttg 30180 gctcatgaaa cttaagtaca cacacacaaa cacacacaca cacacagagt catgcactca 30240 caaaagcatg catgtacacc attcttatta gactatgctt tgctaaaaga ctttcctaga 30300 tactttaaaa catcacttct gccttttggt gggcaggttc caagattggt actggcgtac 30360 tggaaactga acaaggtaga gatctagaaa tcacagcagg tcagaagggc cagcctgtac 30420 aagagagagt tccacacctt ccaggaacac tgagcagggg gctgggacct tgcctctcag 30480 cccaagaaac tagtgcgttt cctgtatgca tgcctctcag agattccata agatctgcct 30540 tctgccataa gatctcctgc atccagacaa gcctagggga agttgagagg ctgcctgagt 30600 ctctcccaca ggccccttct tgcctggcag tattttttta tctggaggag aggaatcagg 30660 gtgggaatga tcaaatacaa ttatcaagga aaaagtaaaa aacatatata tatatatatt 30720 aactgatcta gggagctggc tcagcagtta agagttctgg ctgcccttgc ttcagatctt 30780 gctttgattc ccagcaccca catgatggct ttcaactgta tctctgcttc caggggatcc 30840 aacagcctct tctgacctcc atagacaaga cctagtcctc tgcaagagca ccaaatgctc 30900 ttatctgttg atccatctct ctagcctcat gccagatcat ttaaaactac tggacactgt 30960 cccattttac gaagatgtca ctgcccagtc atttgccatg agtggatatt tcgattcttt 31020 ctatgttctc acccttgcaa tttataagaa agatatctgc atttgtctcc tgagagaaca 31080 aagggtggag ggctactgag atggctctag gggtaaaggt gcttgccaca aaatctgaca 31140 acttaagttt ggtcttggaa tccacatggt ggagagagag aagagattcc cgtaagttgt 31200 cctcaaactt cccacacatg tgctgtggct tatgtgtaac cccaataagt aaagatagtt 31260 ttaaacacta cataaggtag ggtttcttca tgaccccaag gaatgatgcc cctgatagag 31320 cttatgctga aaccccatct ccattgtgcc atctggaaag agacaattgc atcccggaaa 31380 cagaatcttc atgaatggat taatgagcta ttaagaaagt ggcttggtta ttgcacatgc 31440 tggcggcgta atgacctcca ccatgatgtt atccagcatg aaggtcctca ccagaagtca 31500 tacaaatctt cttaggcttc cagagtcgtg agcaaaaaaa gcacacctct aaataaatta 31560 actagcctca ggtagttaac caccgaaaat gaaccaaggc agttctaata caaaaccact 31620 tcccttccct gttcaaacca cagtgcccta ttatctaaaa gataaacttc aagccaagct 31680 tttaggttgc cagtatttat gtaacaacaa ggcccgttga cacacatctg taactcctag 31740 tactgggcct caggggcaga gacaggtgga gccctggagt ttgaattcca ggttctgtga 31800 gaaactctgt ctgaaaagac aatatggtga gtgacccggg aggatatctg atattgactt 31860 ctggccaaca cacagccatc tctgcacatc tgtagttgca agccttttgc actaagtttg 31920 gccagagtca gagtttgcaa gtgtttgtgg actgaatgca cgtgttgctg gtgatctaca 31980 aagtcaccct ccttctcaag ctagcagcac tggcttcggc cagctgctca ttcaagcctc 32040 tttgcagagt catcacgggg atgggggagc agggcccctc cctagaacac caagcctgtg 32100 gttgtttatt caggacatta ttgagggcca agatgacaga taactctatc acttggccaa 32160 cagtcgggtg ttgcggtgtt aggttatttc tgtgtctgca gaaaacagtg caacctggac 32220 aaaagaaata aatgatatca tttttcattc aggcaactag attccgtggt acaaaaggct 32280 ccctggggaa cgaggccggg acagcgcggc tcctgagtcg ctatttccgt ctgtcaactt 32340 ctctaatctc ttgatttcct ccctctgtct gtttccttcc tcttgctggg gcccagtgga 32400 gtctgtgtac tcacagggag gagggtggca aagccctggt cctctacggg ctgggggaag 32460 gggggaagct gtcggcccag tgactttttc ccctttctct ttttcttaga aaccagtctc 32520 aatttaagat aatgagtctc ctcattcacg tgtgctcact attcataggg acttatccac 32580 ccccgccctg tcaatctggc taagtaagac aagtcaaatt taaaagggaa cgtttttcta 32640 aaaatgtggc tggaccgtgt gccggcacga aaccagggat ggcggtctaa gttacatgct 32700 ctctgccagc cccggtgcct tttcctttcg gaaaggagac ccggaggtaa aacgaagttg 32760 ccaacttttg atgatggtgt gcgccgggtg actctttaaa atgtcatcca tacctgggat 32820 agggaaggct cttcagggag tcatctagcc ctcccttcag gaaaagattc cacttccggt 32880 ttagttagct tccacctggt cccttatccg ctgtctctgc ccactagtcc tcatccatcc 32940 ggtttccgcc ctcatccacc ttgccctttt agttcctaga aagcagcacc gtagtcttgg 33000 caggtgggcc attggtcact ccgctaccac tgttaccatg gccaccaagg tgtcatttaa 33060 atatgagctc actgagtcct gcgggatggc ttggttggta atatgcttgc tgcaaaatcg 33120 tgagaactgg agttcaattc ccagcacatg gatgtatttc cagcacctgg aaggcaggga 33180 gcagagatct taaagctcct ggccagacag cccagcctaa ttagtaatca gtgagagacc 33240 ctgtctcaag aaacaagatg gaacatcaaa ggtcaacctc ttgtctccac acacacaaat 33300 acacacatgc acatacatcc acacacaggc aaacacatgc acacacctga acaccctcca 33360 caaatacata cataaaaaaa taaatacata cacacataca tacatacacc aacattccct 33420 ctccttagtc tcctggctac gctcttgtca cccccactaa ggcttcaact tcttctattt 33480 cttcatcttg actcctctgt actttgcatg ccttttccag caaaggcttt tctttaaatc 33540 tccgtcattc ataaactccc tctaaatttc ttcccctgcc cttttctttc tctctaggga 33600 gataaagaca cacactacaa agtcaccgtg ggaccagttt attcacccac ccacccctgc 33660 ttctgttcat ccggccagct aagtagtcca acctctctgg tgctgtaccc tggaccctgg 33720 cttcaccaca gctcctccat gctacccagc cctgcaaacc ttcagcctag cctctggttc 33780 tccaaccagc acaggcccag tctggcttct atgtcctaga aatctccttc attctctcca 33840 tttccctcct gaatctacca ccttctttct cccttctcct gacctctaat gtcttggtca 33900 aacgattaca aggaagccaa tgaaattagc agtttggggt acctcagagt cagcagggga 33960 gctgggatga attcacattt ccaggccttt gctttgctcc ccggattctg acaggcagtt 34020 ccgaagctga gtccaggaag ctgaatttaa aatcacactc cagctgggtt ctgaggcagc 34080 cctaccacat cagctggccc tgactgagct gtgtctgggt ggcagtggtg ctggtggtgc 34140 tggtggtgct ggtggtggtg gtggtggtgg tggtggtggt ggtggtggtg tgtgtgtgtg 34200 ttttctgctt ttacaaaact tttctaattc ttatacaaag gacaaatctg cctcatatag 34260 gcagaaagat gacttatgcc tatataagat ataaagatga ctttatgcca cttattagca 34320 atagttactg tcaaaagtaa ttctatttat acacccttat acatggtatt gcttttgttg 34380 gagactctaa aatccagatt atgtatttaa aaaaaaattc cccagtcctt aaaaggtgaa 34440 gaatggaccc agatagaagg tcacggcaca agtatggagt cggagtgtgg agtcctgcca 34500 atggtctgga cagaagcatc cagagagggt ccaagacaaa tgcctcgcct cctaaggaac 34560 actggcagcc ctgatgaggt accagagatt gctaagtgga ggaatacagg atcagaccca 34620 tggaggggct taaagcgtga ctgtagcagc cctccgctga ggggctccag gtgggcgccc 34680 aaggtgctgc agtgggagcc acatgagagg tgatgtcttg gagtcacctc gggtaccatt 34740 gtttagggag gtggggattt gtggtgtgga gacaggcagc ctcaaggatg cttttcaaca 34800 atggttgatg agttggaact aaaacagggg ccatcacact ggctcccata gctctgggct 34860 tgccagcttc cacatctgcc ccccaccccc tgtctggcac cagctcaagc tctgtgattc 34920 tacacatcca aaagaggaag agtagcctac tgggcatgcc acctcttctg gaccatcagg 34980 tgagagtgtg gcaagcccta ggctcctgtc caggatgcag ggctgccaga taggatgctc 35040 agctatctcc tgagctggaa ctattttagg aataaggatt atgcccgccc ggggttggcc 35100 agcaccccag cagcctgtgc ttgcgtaaaa gcaagtgctg ttgatttatc taaaaacaga 35160 gccgtggacc cacccacagg acaagtatgt atgcatctgt ttcatgtatc tgaaaagcga 35220 cacaaccatt tttcacatca tggcatcttc ctaaccccca ttcttttttg ttttgttttt 35280 ttgagacagg gtttctctgt gtagtcctgg ctgtcctgga actcactttg tagaccaggc 35340 tggcctcgaa ctcagaaatc ctgggattaa aggtgtgtgc caccacgccc ggccctaacc 35400 cccattctta atggtgatcc agtggttgaa atttcgggcc acacacatgt ccattaggga 35460 ttagctgctg tcttctgagc tacctggtac aatctttatc ccctggggcc tgggctcctg 35520 atccctgact cgggcccgat caagtccagt tcctgggccc gatcaagtcc agttcctggg 35580 cccgaacaag tccagtccct agctcgatta gctcatcctg gctccctggc ctgttcttac 35640 ttacactctt ccccttgctc tggacttgtt gctttcttta ctcaagttgt ctgccacagt 35700 ccctaagcca cctctgtaag acaactaaga taatacttcc ctcaagcacg gaaagtcctg 35760 agtcaccaca ccctctggag gtgtgtggac acatgttcat gcgtgtggtt gcgcttacgt 35820 acgtgtgc 35828 18 9301 DNA Homo sapien 18 tagaggagaa gtctttgggg agggtttgct ctgagcacac ccctttccct ccctccgggg 60 ctgagggaaa catgggacca gccctgcccc agcctgtcct cattggctgg catgaagcag 120 agaggggctt taaaaaggcg accgtgtctc ggctggagac cagagcctgt gctactggaa 180 ggtggcgtgc cctcctctgg ctggtaccat gcagctccca ctggccctgt gtctcgtctg 240 cctgctggta cacacagcct tccgtgtagt ggagggccag gggtggcagg cgttcaagaa 300 tgatgccacg gaaatcatcc ccgagctcgg agagtacccc gagcctccac cggagctgga 360 gaacaacaag accatgaacc gggcggagaa cggagggcgg cctccccacc acccctttga 420 gaccaaaggt atggggtgga ggagagaatt cttagtaaaa gatcctgggg aggttttaga 480 aacttctctt tgggaggctt ggaagactgg ggtagaccca gtgaagattg ctggcctctg 540 ccagcactgg tcgaggaaca gtcttgcctg gaggtggggg aagaatggct cgctggtgca 600 gccttcaaat tcaggtgcag aggcatgagg caacagacgc tggtgagagc ccagggcagg 660 gaggacgctg gggtggtgag ggtatggcat cagggcatca gaacaggctc aggggctcag 720 aaaagaaaag gtttcaaaga atctcctcct gggaatatag gagccacgtc cagctgctgg 780 taccactggg aagggaacaa ggtaagggag cctcccatcc acagaacagc acctgtgggg 840 caccggacac tctatgctgg tggtggctgt ccccaccaca cagacccaca tcatggaatc 900 cccaggaggt gaacccccag ctcgaagggg aagaaacagg ttccaggcac tcagtaactt 960 ggtagtgaga agagctgagg tgtgaacctg gtttgatcca actgcaagat agccctggtg 1020 tgtggggggg tgtgggggac agatctccac aaagcagtgg ggaggaaggc cagagaggca 1080 cccctgcagt gtgcattgcc catggcctgc ccagggagct ggcacttgaa ggaatgggag 1140 ttttcggcac agttttagcc cctgacatgg gtgcagctga gtccaggccc tggaggggag 1200 agcagcatcc tctgtgcagg agtagggaca tctgtcctca gcagccaccc cagtcccaac 1260 cttgcctcat tccaggggag ggagaaggaa gaggaaccct gggttcctgg tcaggcctgc 1320 acagagaagc ccaggtgaca gtgtgcatct ggctctataa ttggcaggaa tcctgaggcc 1380 atgggggcgt ctgaaatgac acttcagact aagagcttcc ctgtcctctg gccattatcc 1440 aggtggcaga gaagtccact gcccaggctc ctggacccca gccctccccg cctcacaacc 1500 tgttgggact atggggtgct aaaaagggca actgcatggg aggccagcca ggaccctccg 1560 tcttcaaaat ggaggacaag ggcgcctccc cccacagctc cccttctagg caaggtcagc 1620 tgggctccag cgactgcctg aagggctgta aggaacccaa acacaaaatg tccaccttgc 1680 tggactccca cgagaggcca cagcccctga ggaagccaca tgctcaaaac aaagtcatga 1740 tctgcagagg aagtgcctgg cctaggggcg ctattctcga aaagccgcaa aatgccccct 1800 tccctgggca aatgcccccc tgaccacaca cacattccag ccctgcagag gtgaggatgc 1860 aaaccagccc acagaccaga aagcagcccc agacgatggc agtggccaca tctcccctgc 1920 tgtgcttgct cttcagagtg ggggtggggg gtggccttct ctgtcccctc tctggtttgg 1980 tcttaagact atttttcatt ctttcttgtc acattggaac tatccccatg aaacctttgg 2040 gggtggactg gtactcacac gacgaccagc tatttaaaaa gctcccaccc atctaagtcc 2100 accataggag acatggtcaa ggtgtgtgca ggggatcagg ccaggcctcg gagcccaatc 2160 tctgcctgcc cagggagtat caccatgagg cgcccattca gataacacag aacaagaaat 2220 gtgcccagca gagagccagg tcaatgtttg tggcagctga acctgtaggt tttgggtcag 2280 agctcagggc ccctatggta ggaaagtaac gacagtaaaa agcagccctc agctccatcc 2340 cccagcccag cctcccatgg atgctcgaac gcagagcctc cactcttgcc ggagccaaaa 2400 ggtgctggga ccccagggaa gtggagtccg gagatgcagc ccagcctttt gggcaagttc 2460 ttttctctgg ctgggcctca gtattctcat tgataatgag ggggttggac acactgcctt 2520 tgattccttt caagtctaat gaattcctgt cctgatcacc tccccttcag tccctcgcct 2580 ccacagcagc tgccctgatt tattaccttc aattaacctc tactcctttc tccatcccct 2640 gtccacccct cccaagtggc tggaaaagga atttgggaga agccagagcc aggcagaagg 2700 tgtgctgagt acttaccctg cccaggccag ggaccctgcg gcacaagtgt ggcttaaatc 2760 ataagaagac cccagaagag aaatgataat aataatacat aacagccgac gctttcagct 2820 atatgtgcca aatggtattt tctgcattgc gtgtgtaatg gattaactcg caatgcttgg 2880 ggcggcccat tttgcagaca ggaagaagag agaggttaag gaacttgccc aagatgacac 2940 ctgcagtgag cgatggagcc ctggtgtttg aaccccagca gtcatttggc tccgagggga 3000 cagggtgcgc aggagagctt tccaccagct ctagagcatc tgggaccttc ctgcaataga 3060 tgttcagggg caaaagcctc tggagacagg cttggcaaaa gcagggctgg ggtggagaga 3120 gacgggccgg tccagggcag gggtggccag gcgggcggcc accctcacgc gcgcctctct 3180 ccacagacgt gtccgagtac agctgccgcg agctgcactt cacccgctac gtgaccgatg 3240 ggccgtgccg cagcgccaag ccggtcaccg agctggtgtg ctccggccag tgcggcccgg 3300 cgcgcctgct gcccaacgcc atcggccgcg gcaagtggtg gcgacctagt gggcccgact 3360 tccgctgcat ccccgaccgc taccgcgcgc agcgcgtgca gctgctgtgt cccggtggtg 3420 aggcgccgcg cgcgcgcaag gtgcgcctgg tggcctcgtg caagtgcaag cgcctcaccc 3480 gcttccacaa ccagtcggag ctcaaggact tcgggaccga ggccgctcgg ccgcagaagg 3540 gccggaagcc gcggccccgc gcccggagcg ccaaagccaa ccaggccgag ctggagaacg 3600 cctactagag cccgcccgcg cccctcccca ccggcgggcg ccccggccct gaacccgcgc 3660 cccacatttc tgtcctctgc gcgtggtttg attgtttata tttcattgta aatgcctgca 3720 acccagggca gggggctgag accttccagg ccctgaggaa tcccgggcgc cggcaaggcc 3780 cccctcagcc cgccagctga ggggtcccac ggggcagggg agggaattga gagtcacaga 3840 cactgagcca cgcagccccg cctctggggc cgcctacctt tgctggtccc acttcagagg 3900 aggcagaaat ggaagcattt tcaccgccct ggggttttaa gggagcggtg tgggagtggg 3960 aaagtccagg gactggttaa gaaagttgga taagattccc ccttgcacct cgctgcccat 4020 cagaaagcct gaggcgtgcc cagagcacaa gactgggggc aactgtagat gtggtttcta 4080 gtcctggctc tgccactaac ttgctgtgta accttgaact acacaattct ccttcgggac 4140 ctcaatttcc actttgtaaa atgagggtgg aggtgggaat aggatctcga ggagactatt 4200 ggcatatgat tccaaggact ccagtgcctt ttgaatgggc agaggtgaga gagagagaga 4260 gaaagagaga gaatgaatgc agttgcattg attcagtgcc aaggtcactt ccagaattca 4320 gagttgtgat gctctcttct gacagccaaa gatgaaaaac aaacagaaaa aaaaaagtaa 4380 agagtctatt tatggctgac atatttacgg ctgacaaact cctggaagaa gctatgctgc 4440 ttcccagcct ggcttccccg gatgtttggc tacctccacc cctccatctc aaagaaataa 4500 catcatccat tggggtagaa aaggagaggg tccgagggtg gtgggaggga tagaaatcac 4560 atccgcccca acttcccaaa gagcagcatc cctcccccga cccatagcca tgttttaaag 4620 tcaccttccg aagagaagtg aaaggttcaa ggacactggc cttgcaggcc cgagggagca 4680 gccatcacaa actcacagac cagcacatcc cttttgagac accgccttct gcccaccact 4740 cacggacaca tttctgccta gaaaacagct tcttactgct cttacatgtg atggcatatc 4800 ttacactaaa agaatattat tgggggaaaa actacaagtg ctgtacatat gctgagaaac 4860 tgcagagcat aatagctgcc acccaaaaat ctttttgaaa atcatttcca gacaacctct 4920 tactttctgt gtagttttta attgttaaaa aaaaaaagtt ttaaacagaa gcacatgaca 4980 tatgaaagcc tgcaggactg gtcgtttttt tggcaattct tccacgtggg acttgtccac 5040 aagaatgaaa gtagtggttt ttaaagagtt aagttacata tttattttct cacttaagtt 5100 atttatgcaa aagtttttct tgtagagaat gacaatgtta atattgcttt atgaattaac 5160 agtctgttct tccagagtcc agagacattg ttaataaaga caatgaatca tgaccgaaag 5220 gatgtggtct cattttgtca accacacatg acgtcatttc tgtcaaagtt gacacccttc 5280 tcttggtcac tagagctcca accttggaca cacctttgac tgctctctgg tggcccttgt 5340 ggcaattatg tcttcctttg aaaagtcatg tttatccctt cctttccaaa cccagaccgc 5400 atttcttcac ccagggcatg gtaataacct cagccttgta tccttttagc agcctcccct 5460 ccatgctggc ttccaaaatg ctgttctcat tgtatcactc ccctgctcaa aagccttcca 5520 tagctccccc ttgcccagga tcaagtgcag tttccctatc tgacatggga ggccttctct 5580 gcttgactcc cacctcccac tccaccaagc ttcctactga ctccaaatgg tcatgcagat 5640 ccctgcttcc ttagtttgcc atccacactt agcaccccca ataactaatc ctctttcttt 5700 aggattcaca ttacttgtca tctcttcccc taaccttcca gagatgttcc aatctcccat 5760 gatccctctc tcctctgagg ttccagcccc ttttgtctac accactactt tggttcctaa 5820 ttctgttttc catttgacag tcattcatgg aggaccagcc tggccaagtc ctgcttagta 5880 ctggcataga caacacaaag ccaagtacaa ttcaggacca gctcacagga aacttcatct 5940 tcttcgaagt gtggatttga tgcctcctgg gtagaaatgt aggatcttca aaagtgggcc 6000 agcctcctgc acttctctca aagtctcgcc tccccaaggt gtcttaatag tgctggatgc 6060 tagctgagtt agcatcttca gatgaagagt aaccctaaag ttactcttca gttgccctaa 6120 ggtgggatgg tcaactggaa agctttaaat taagtccagc ctaccttggg ggaacccacc 6180 cccacaaaga aagctgaggt ccctcctgat gacttgtcag tttaactacc aataacccac 6240 ttgaattaat catcatcatc aagtctttga taggtgtgag tgggtatcag tggccggtcc 6300 cttcctgggg ctccagcccc cgaggaggcc tcagtgagcc cctgcagaaa atccatgcat 6360 catgagtgtc tcagggccca gaatatgaga gcaggtagga aacagagaca tcttccatcc 6420 ctgagaggca gtgcggtcca gtgggtgggg acacgggctc tgggtcaggt ttgtgttgtt 6480 tgtttgtttg ttttgagaca gagtctcgct ctattgccca ggctggagtg cagtgtcaca 6540 atctcggctt actgcaactt ctgccttccc ggattcaagt gattctcctg cctcagcctc 6600 cagagtagct gggattacag gtgcgtgcca ccacgcctgg ctaatttttg tatttttgat 6660 agagacgggg tttcaccatg ttggccaggc tagtctcgaa ctcttgacct caagtgatct 6720 gcctgcctcg gcctcccaaa gtgctgggat tacaggcgtg agccaccaca cccagcccca 6780 ggttggtgtt tgaatctgag gagactgaag caccaagggg ttaaatgttt tgcccacagc 6840 catacttggg ctcagttcct tgccctaccc ctcacttgag ctgcttagaa cctggtgggc 6900 acatgggcaa taaccaggtc acactgtttt gtaccaagtg ttatgggaat ccaagatagg 6960 agtaatttgc tctgtggagg ggatgaggga tagtggttag ggaaagcttc acaaagtggg 7020 tgttgcttag agattttcca ggtggagaag ggggcttcta ggcagaaggc atagcccaag 7080 caaagactgc aagtgcatgg ctgctcatgg gtagaagaga atccaccatt cctcaacatg 7140 taccgagtcc ttgccatgtg caaggcaaca tgggggtacc aggaattcca agcaatgtcc 7200 aaacctaggg tctgctttct gggacctgaa gatacaggat ggatcagccc aggctgcaat 7260 cccattacca cgagggggaa aaaaacctga aggctaaatt gtaggtcggg ttagaggtta 7320 tttatggaaa gttatattct acctacatgg ggtctataag cctggcgcca atcagaaaag 7380 gaacaaacaa cagacctagc tgggaggggc agcattttgt tgtagggggc ggggcacatg 7440 ttctgggggt acagccagac tcagggcttg tattaatagt ctgagagtaa gacagacaga 7500 gggatagaag gaaataggtc cctttctctc tctctctctc tctctctctc actctctctc 7560 tctctcacac acacacacag acacacacac acgctctgta ggggtctact tatgctccaa 7620 gtacaaatca ggccacattt acacaaggag gtaaaggaaa agaacgttgg aggagccaca 7680 ggaccccaaa attccctgtt ttccttgaat caggcaggac ttacgcagct gggagggtgg 7740 agagcctgca gaagccacct gcgagtaagc caagttcaga gtcacagaca ccaaaagctg 7800 gtgccatgtc ccacacccgc ccacctccca cctgctcctt gacacagccc tgtgctccac 7860 aacccggctc ccagatcatt gattatagct ctggggcctg caccgtcctt cctgccacat 7920 ccccacccca ttcttggaac ctgccctctg tcttctccct tgtccaaggg caggcaaggg 7980 ctcagctatt gggcagcttt gaccaacagc tgaggctcct tttgtggctg gagatgcagg 8040 aggcagggga atattcctct tagtcaatgc gaccatgtgc ctggtttgcc cagggtggtc 8100 tcgtttacac ctgtaggcca agcgtaatta ttaacagctc ccacttctac tctaaaaaat 8160 gacccaatct gggcagtaaa ttatatggtg cccatgctat taagagctgc aacttgctgg 8220 gcgtggtggc tcacacctgt aatcccagta ctttgggacg tcaaggcggg tggatcacct 8280 gaggtcacga gttagagact ggcctggcca gcatggcaaa accccatctt tactaaaaat 8340 acaaaaatta gcaaggcatg gtggcatgca cctgtaatcc caggtactcg ggaggctgag 8400 acaggagaat ggcttgaacc caggaggcag aggttgcagt gagccaagat tgtgccactg 8460 ccctccagcc ctggcaacag agcaagactt catctcaaaa gaaaaaggat actgtcaatc 8520 actgcaggaa gaacccaggt aatgaatgag gagaagagag gggctgagtc accatagtgg 8580 cagcaccgac tcctgcagga aaggcgagac actgggtcat gggtactgaa gggtgccctg 8640 aatgacgttc tgctttagag accgaacctg agccctgaaa gtgcatgcct gttcatgggt 8700 gagagactaa attcatcatt ccttggcagg tactgaatcc tttcttacgg ctgccctcca 8760 atgcccaatt tccctacaat tgtctggggt gcctaagctt ctgcccacca agagggccag 8820 agctggcagc gagcagctgc aggtaggaga gataggtacc cataagggag gtgggaaaga 8880 gagatggaag gagaggggtg cagagcacac acctcccctg cctgacaact tcctgagggc 8940 tggtcatgcc agcagattta aggcggaggc aggggagatg gggcgggaga ggaagtgaaa 9000 aaggagaggg tggggatgga gaggaagaga gggtgatcat tcattcattc cattgctact 9060 gactggatgc cagctgtgag ccaggcacca ccctagctct gggcatgtgg ttgtaatctt 9120 ggagcctcat ggagctcaca gggagtgctg gcaaggagat ggataatgga cggataacaa 9180 ataaacattt agtacaatgt ccgggaatgg aaagttctcg aaagaaaaat aaagctggtg 9240 agcatataga cagccctgaa ggcggccagg ccaggcattt ctgaggaggt ggcatttgag 9300 c 9301 19 21 DNA Artificial Sequence Primer for PCR 19 ccggagctgg agaacaacaa g 21 20 19 DNA Artificial Sequence PRimer for PCR 20 gcactggccg gagcacacc 19 21 23 DNA Artificial Sequence Primer for PCR 21 aggccaaccg cgagaagatg acc 23 22 21 DNA Artificial Sequence Primer for PCR 22 gaagtccagg gcgacgtagc a 21 23 25 DNA Artificial Sequence Primer for PCR 23 aagcttggta ccatgcagct cccac 25 24 50 DNA Artificial Sequence Primer for PCR 24 aagcttctac ttgtcatcgt cgtccttgta gtcgtaggcg ttctccagct 50 25 19 DNA Artificial Sequence Primer for PCR 25 gcactggccg gagcacacc 19 26 39 DNA Artificial Sequence Primer for PCR 26 gtcgtcggat ccatggggtg gcaggcgttc aagaatgat 39 27 57 DNA Artificial Sequence Primer for PCR 27 gtcgtcaagc ttctacttgt catcgtcctt gtagtcgtag gcgttctcca gctcggc 57 28 29 DNA Artificial Sequence Primer for PCR 28 gacttggatc ccaggggtgg caggcgttc 29 29 29 DNA Artificial Sequence Primer for PCR 29 agcataagct tctagtaggc gttctccag 29 30 29 DNA Artificial Sequence Primer for PCR 30 gacttggatc cgaagggaaa aagaaaggg 29 31 29 DNA Artificial Sequence Primer for PCR 31 agcataagct tttaatccaa atcgatgga 29 32 33 DNA Artificial Sequence Primer for PCR 32 actacgagct cggccccacc acccatcaac aag 33 33 34 DNA Artificial Sequence Primer for PCR 33 acttagaagc tttcagtcct cagccccctc ttcc 34 34 66 DNA Artificial Sequence Primer for PCR 34 aatctggatc cataacttcg tatagcatac attatacgaa gttatctgca ggattcgagg 60 gcccct 66 35 82 DNA Artificial Sequence Primer for PCR 35 aatctgaatt ccaccggtgt taattaaata acttcgtata atgtatgcta tacgaagtta 60 tagatctaga gtcagcttct ga 82 36 62 DNA Artificial Sequence Primer for PCR 36 atttaggtga cactatagaa ctcgagcagc tgaagcttaa ccacatggtg gctcacaacc 60 at 62 37 54 DNA Artificial Sequence Primer for PCR 37 aacgacggcc agtgaatccg taatcatggt catgctgcca ggtggaggag ggca 54 38 31 DNA Artificial Sequence Primer for PCR 38 attaccaccg gtgacacccg cttcctgaca g 31 39 61 DNA Artificial Sequence Primer for PCR 39 attacttaat taaacatggc gcgccatatg gccggcccct aattgcggcg catcgttaat 60 t 61 40 34 DNA Artificial Sequence Primer for PCR 40 attacggccg gccgcaaagg aattcaagat ctga 34 41 34 DNA Artificial Sequence Primer for PCR 41 attacggcgc gcccctcaca ggccgcaccc agct 34 42 184 PRT Homo sapiens 42 Met Ser Arg Thr Ala Tyr Thr Val Gly Ala Leu Leu Leu Leu Leu Gly 1 5 10 15 Thr Leu Leu Pro Ala Ala Glu Gly Lys Lys Lys Gly Ser Gln Gly Ala 20 25 30 Ile Pro Pro Pro Asp Lys Ala Gln His Asn Asp Ser Glu Gln Thr Gln 35 40 45 Ser Pro Gln Gln Pro Gly Ser Arg Asn Arg Gly Arg Gly Gln Gly Arg 50 55 60 Gly Thr Ala Met Pro Gly Glu Glu Val Leu Glu Ser Ser Gln Glu Ala 65 70 75 80 Leu His Val Thr Glu Arg Lys Tyr Leu Lys Arg Asp Trp Cys Lys Thr 85 90 95 Gln Pro Leu Lys Gln Thr Ile His Glu Glu Gly Cys Asn Ser Arg Thr 100 105 110 Ile Ile Asn Arg Phe Cys Tyr Gly Gln Cys Asn Ser Phe Tyr Ile Pro 115 120 125 Arg His Ile Arg Lys Glu Glu Gly Ser Phe Gln Ser Cys Ser Phe Cys 130 135 140 Lys Pro Lys Lys Phe Thr Thr Met Met Val Thr Leu Asn Cys Pro Glu 145 150 155 160 Leu Gln Pro Pro Thr Lys Lys Lys Arg Val Thr Arg Val Lys Gln Cys 165 170 175 Arg Cys Ile Ser Ile Asp Leu Asp 180 43 267 PRT Homo sapiens 43 Met His Leu Leu Leu Phe Gln Leu Leu Val Leu Leu Pro Leu Gly Lys 1 5 10 15 Thr Thr Arg His Gln Asp Gly Arg Gln Asn Gln Ser Ser Leu Ser Pro 20 25 30 Val Leu Leu Pro Arg Asn Gln Arg Glu Leu Pro Thr Gly Asn His Glu 35 40 45 Glu Ala Glu Glu Lys Pro Asp Leu Phe Val Ala Val Pro His Leu Val 50 55 60 Ala Thr Ser Pro Ala Gly Glu Gly Gln Arg Gln Arg Glu Lys Met Leu 65 70 75 80 Ser Arg Phe Gly Arg Phe Trp Lys Lys Pro Glu Arg Glu Met His Pro 85 90 95 Ser Arg Asp Ser Asp Ser Glu Pro Phe Pro Pro Gly Thr Gln Ser Leu 100 105 110 Ile Gln Pro Ile Asp Gly Met Lys Met Glu Lys Ser Pro Leu Arg Glu 115 120 125 Glu Ala Lys Lys Phe Trp His His Phe Met Phe Arg Lys Thr Pro Ala 130 135 140 Ser Gln Gly Val Ile Leu Pro Ile Lys Ser His Glu Val His Trp Glu 145 150 155 160 Thr Cys Arg Thr Val Pro Phe Ser Gln Thr Ile Thr His Glu Gly Cys 165 170 175 Glu Lys Val Val Val Gln Asn Asn Leu Cys Phe Gly Lys Cys Gly Ser 180 185 190 Val His Phe Pro Gly Ala Ala Gln His Ser His Thr Ser Cys Ser His 195 200 205 Cys Leu Pro Ala Lys Phe Thr Thr Met His Leu Pro Leu Asn Cys Thr 210 215 220 Glu Leu Ser Ser Val Ile Lys Val Val Met Leu Val Glu Glu Cys Gln 225 230 235 240 Cys Lys Val Lys Thr Glu His Glu Asp Gly His Ile Leu His Ala Gly 245 250 255 Ser Gln Asp Ser Phe Ile Pro Gly Val Ser Ala 260 265 44 180 PRT Homo sapiens 44 Met Leu Arg Val Leu Val Gly Ala Val Leu Pro Ala Met Leu Leu Ala 1 5 10 15 Ala Pro Pro Pro Ile Asn Lys Leu Ala Leu Phe Pro Asp Lys Ser Ala 20 25 30 Trp Cys Glu Ala Lys Asn Ile Thr Gln Ile Val Gly His Ser Gly Cys 35 40 45 Glu Ala Lys Ser Ile Gln Asn Arg Ala Cys Leu Gly Gln Cys Phe Ser 50 55 60 Tyr Ser Val Pro Asn Thr Phe Pro Gln Ser Thr Glu Ser Leu Val His 65 70 75 80 Cys Asp Ser Cys Met Pro Ala Gln Ser Met Trp Glu Ile Val Thr Leu 85 90 95 Glu Cys Pro Gly His Glu Glu Val Pro Arg Val Asp Lys Leu Val Glu 100 105 110 Lys Ile Leu His Cys Ser Cys Gln Ala Cys Gly Lys Glu Pro Ser His 115 120 125 Glu Gly Leu Ser Val Tyr Val Gln Gly Glu Asp Gly Pro Gly Ser Gln 130 135 140 Pro Gly Thr His Pro His Pro His Pro His Pro His Pro Gly Gly Gln 145 150 155 160 Thr Pro Glu Pro Glu Asp Pro Pro Gly Ala Pro His Thr Glu Glu Glu 165 170 175 Gly Ala Glu Asp 180 45 642 DNA Homo sapiens CDS (1)..(639) 45 atg cag ctc cca ctg gcc ctg tgt ctc gtc tgc ctg ctg gta cac aca 48 Met Gln Leu Pro Leu Ala Leu Cys Leu Val Cys Leu Leu Val His Thr 1 5 10 15 gcc ttc cgt gta gtg gag ggc cag ggg tgg cag gcg ttc aag aat gat 96 Ala Phe Arg Val Val Glu Gly Gln Gly Trp Gln Ala Phe Lys Asn Asp 20 25 30 gcc acg gaa atc atc ccc gag ctc gga gag tac ccc gag cct cca ccg 144 Ala Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro 35 40 45 gag ctg gag aac aac aag acc atg aac cgg gcg gag aac gga ggg cgg 192 Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg 50 55 60 cct ccc cac cac ccc ttt gag acc aaa gac gtg tcc gag tac agc tgc 240 Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser Cys 65 70 75 80 cgc gag ctg cac ttc acc cgc tac gtg acc gat ggg ccg tgc cgc agc 288 Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg Ser 85 90 95 gcc aag ccg gtc acc gag ctg gtg tgc tcc ggc cag tgc ggc ccg gcg 336 Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro Ala 100 105 110 cgc ctg ctg ccc aac gcc atc ggc cgc ggc aag tgg tgg cga cct agt 384 Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser 115 120 125 ggg ccc gac ttc cgc tgc atc ccc gac cgc tac cgc gcg cag cgc gtg 432 Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val 130 135 140 cag ctg ctg tgt ccc ggt ggt gag gcg ccg cgc gcg cgc aag gtg cgc 480 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 145 150 155 160 ctg gtg gcc tcg tgc aag tgc aag cgc ctc acc cgc ttc cac aac cag 528 Leu Val Ala Ser Cys Lys Cys Lys Arg Leu Thr Arg Phe His Asn Gln 165 170 175 tcg gag ctc aag gac ttc ggg acc gag gcc gct cgg ccg cag aag ggc 576 Ser Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly 180 185 190 cgg aag ccg cgg ccc cgc gcc cgg agc gcc aaa gcc aac cag gcc gag 624 Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu 195 200 205 ctg gag aac gcc tac tag 642 Leu Glu Asn Ala Tyr 210 46 190 PRT Homo sapiens 46 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Glu 1 5 10 15 Leu Gly Glu Tyr Pro Glu Pro Pro Pro Glu Leu Glu Asn Asn Lys Thr 20 25 30 Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro Phe Glu 35 40 45 Thr Lys Asp Val Ser Glu Tyr Ser Cys Arg Glu Leu His Phe Thr Arg 50 55 60 Tyr Val Thr Asp Gly Pro Cys Arg Ser Ala Lys Pro Val Thr Glu Leu 65 70 75 80 Val Cys Ser Gly Gln Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile 85 90 95 Gly Arg Gly Lys Trp Trp Arg Pro Ser Gly Pro Asp Phe Arg Cys Ile 100 105 110 Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly Gly 115 120 125 Glu Ala Pro Arg Ala Arg Lys Val Arg Leu Val Ala Ser Cys Lys Cys 130 135 140 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 145 150 155 160 Thr Glu Ala Ala Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala 165 170 175 Arg Ser Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 180 185 190 47 20 PRT Homo sapiens 47 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Glu 1 5 10 15 Leu Gly Glu Tyr 20 48 20 PRT Homo sapiens 48 Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro Glu 1 5 10 15 Leu Glu Asn Asn 20 49 20 PRT Homo sapiens 49 Pro Glu Pro Pro Pro Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala 1 5 10 15 Glu Asn Gly Gly 20 50 20 PRT Homo sapiens 50 Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro 1 5 10 15 Phe Glu Thr Lys 20 51 16 PRT Homo sapiens 51 Arg Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser 1 5 10 15 52 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 52 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Glu 1 5 10 15 Leu Gly Glu Tyr Cys 20 53 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 53 Thr Glu Ile Ile Pro Glu Leu Gly Glu Tyr Pro Glu Pro Pro Pro Glu 1 5 10 15 Leu Glu Asn Asn Cys 20 54 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 54 Pro Glu Pro Pro Pro Glu Leu Glu Asn Asn Lys Thr Met Asn Arg Ala 1 5 10 15 Glu Asn Gly Gly Cys 20 55 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 55 Lys Thr Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro 1 5 10 15 Phe Glu Thr Lys Cys 20 56 17 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 56 Arg Pro Pro His His Pro Phe Glu Thr Lys Asp Val Ser Glu Tyr Ser 1 5 10 15 Cys 57 24 PRT Rattus norvegicus 57 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Gly 1 5 10 15 Leu Arg Glu Tyr Pro Glu Pro Pro 20 58 20 PRT Rattus morvegicus 58 Pro Glu Pro Pro Gln Glu Leu Glu Asn Asn Gln Thr Met Asn Arg Ala 1 5 10 15 Glu Asn Gly Gly 20 59 20 PRT Rattus norvegicus 59 Glu Asn Gly Gly Arg Pro Pro His His Pro Tyr Asp Thr Lys Asp Val 1 5 10 15 Ser Glu Tyr Ser 20 60 20 PRT Rattus norvegicus 60 Thr Glu Ile Ile Pro Gly Leu Arg Glu Tyr Pro Glu Pro Pro Gln Glu 1 5 10 15 Leu Glu Asn Asn 20 61 25 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 61 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Gly 1 5 10 15 Leu Arg Glu Tyr Pro Glu Pro Pro Cys 20 25 62 21 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 62 Pro Glu Pro Pro Gln Glu Leu Glu Asn Asn Gln Thr Met Asn Arg Ala 1 5 10 15 Glu Asn Gly Gly Cys 20 63 21 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 63 Glu Asn Gly Gly Arg Pro Pro His His Pro Tyr Asp Thr Lys Asp Val 1 5 10 15 Ser Glu Tyr Ser Cys 20 64 21 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 64 Thr Glu Ile Ile Pro Gly Leu Arg Glu Tyr Pro Glu Pro Pro Gln Glu 1 5 10 15 Leu Glu Asn Asn Cys 20 65 190 PRT Rattus norvegicus 65 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Gly 1 5 10 15 Leu Arg Glu Tyr Pro Glu Pro Pro Gln Glu Leu Glu Asn Asn Gln Thr 20 25 30 Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro Tyr Asp 35 40 45 Thr Lys Asp Val Ser Glu Tyr Ser Cys Arg Glu Leu His Tyr Thr Arg 50 55 60 Phe Val Thr Asp Gly Pro Cys Arg Ser Ala Lys Pro Val Thr Glu Leu 65 70 75 80 Val Cys Ser Gly Gln Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile 85 90 95 Gly Arg Val Lys Trp Trp Arg Pro Asn Gly Pro Asp Phe Arg Cys Ile 100 105 110 Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly Gly 115 120 125 Ala Ala Pro Arg Ser Arg Lys Val Arg Leu Val Ala Ser Cys Lys Cys 130 135 140 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 145 150 155 160 Pro Glu Thr Ala Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala 165 170 175 Arg Gly Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 180 185 190 66 20 PRT Homo sapiens 66 Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly 1 5 10 15 Gly Glu Ala Pro 20 67 20 PRT Homo sapiens 67 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 1 5 10 15 Leu Val Ala Ser 20 68 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 68 Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly 1 5 10 15 Gly Glu Ala Pro Cys 20 69 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 69 Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg 1 5 10 15 Leu Val Ala Ser Cys 20 70 17 PRT Rattus Norvegicus 70 Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly 1 5 10 15 Gly 71 16 PRT Rattus norvegicus 71 Pro Gly Gly Ala Ala Pro Arg Ser Arg Lys Val Arg Leu Val Ala Ser 1 5 10 15 72 18 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 72 Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Ser Pro Gly 1 5 10 15 Gly Cys 73 17 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 73 Pro Gly Gly Ala Ala Pro Arg Ser Arg Lys Val Arg Leu Val Ala Ser 1 5 10 15 Cys 74 20 PRT Homo sapiens 74 Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp 1 5 10 15 Trp Arg Pro Ser 20 75 16 PRT Homo sapiens 75 Ile Gly Arg Gly Lys Trp Trp Arg Pro Ser Gly Pro Asp Phe Arg Cys 1 5 10 15 76 18 PRT Rattus norvegicus 76 Pro Asn Ala Ile Gly Arg Val Lys Trp Trp Arg Pro Asn Gly Pro Asp 1 5 10 15 Phe Arg 77 19 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 77 Pro Asn Ala Ile Gly Arg Val Lys Trp Trp Arg Pro Asn Gly Pro Asp 1 5 10 15 Phe Arg Cys 78 20 PRT Homo sapiens 78 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 1 5 10 15 Thr Glu Ala Ala 20 79 20 PRT Homo sapiens 79 Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly Arg 1 5 10 15 Lys Pro Arg Pro 20 80 20 PRT Homo sapiens 80 Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys 1 5 10 15 Ala Asn Gln Ala 20 81 16 PRT Homo sapiens 81 Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 1 5 10 15 82 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 82 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 1 5 10 15 Thr Glu Ala Ala Cys 20 83 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 83 Glu Leu Lys Asp Phe Gly Thr Glu Ala Ala Arg Pro Gln Lys Gly Arg 1 5 10 15 Lys Pro Arg Pro Cys 20 84 21 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 84 Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Ser Ala Lys 1 5 10 15 Ala Asn Gln Ala Cys 20 85 17 PRT Artificial Sequence Human SOST peptide fragment with additional cysteine added 85 Arg Ala Arg Ser Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 1 5 10 15 Cys 86 23 PRT Rattus norvegicus 86 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 1 5 10 15 Pro Glu Thr Ala Arg Pro Gln 20 87 23 PRT Rattus norvegicus 87 Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Gly Ala Lys Ala Asn Gln 1 5 10 15 Ala Glu Leu Glu Asn Ala Tyr 20 88 24 PRT Rattus norvegicus 88 Ser Glu Leu Lys Asp Phe Gly Pro Glu Thr Ala Arg Pro Gln Lys Gly 1 5 10 15 Arg Lys Pro Arg Pro Arg Ala Arg 20 89 24 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 89 Lys Arg Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly 1 5 10 15 Pro Glu Thr Ala Arg Pro Gln Cys 20 90 24 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 90 Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Gly Ala Lys Ala Asn Gln 1 5 10 15 Ala Glu Leu Glu Asn Ala Tyr Cys 20 91 25 PRT Artificial Sequence Rat SOST peptide fragment with additional cysteine added 91 Ser Glu Leu Lys Asp Phe Gly Pro Glu Thr Ala Arg Pro Gln Lys Gly 1 5 10 15 Arg Lys Pro Arg Pro Arg Ala Arg Cys 20 25 92 56 PRT Homo sapiens 92 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Glu 1 5 10 15 Leu Gly Glu Tyr Pro Glu Pro Pro Pro Glu Leu Glu Asn Asn Lys Thr 20 25 30 Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro Phe Glu 35 40 45 Thr Lys Asp Val Ser Glu Tyr Ser 50 55 93 56 PRT Rattus norvegicus 93 Gln Gly Trp Gln Ala Phe Lys Asn Asp Ala Thr Glu Ile Ile Pro Gly 1 5 10 15 Leu Arg Glu Tyr Pro Glu Pro Pro Gln Glu Leu Glu Asn Asn Gln Thr 20 25 30 Met Asn Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro Tyr Asp 35 40 45 Thr Lys Asp Val Ser Glu Tyr Ser 50 55 94 32 PRT Homo sapiens 94 Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro 1 5 10 15 Gly Gly Glu Ala Pro Arg Ala Arg Lys Val Arg Leu Val Ala Ser Cys 20 25 30 95 32 PRT Rattus norvegicus 95 Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro 1 5 10 15 Gly Gly Ala Ala Pro Arg Ser Arg Lys Val Arg Leu Val Ala Ser Cys 20 25 30 96 44 PRT Homo sapiens 96 Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly Thr Glu 1 5 10 15 Ala Ala Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Ser 20 25 30 Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 35 40 97 44 PRT Rattus norvegicus 97 Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly Pro Glu 1 5 10 15 Thr Ala Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Arg Ala Arg Gly 20 25 30 Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 35 40 98 26 PRT Homo sapiens 98 Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp 1 5 10 15 Trp Arg Pro Ser Gly Pro Asp Phe Arg Cys 20 25 99 26 PRT Rattus norvegicus 99 Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg Val Lys Trp 1 5 10 15 Trp Arg Pro Asn Gly Pro Asp Phe Arg Cys 20 25 100 570 DNA Homo sapiens 100 caggggtggc aggcgttcaa gaatgatgcc acggaaatca tccccgagct cggagagtac 60 cccgagcctc caccggagct ggagaacaac aagaccatga accgggcgga gaacggaggg 120 cggcctcccc accacccctt tgagaccaaa gacgtgtccg agtacagctg ccgcgagctg 180 cacttcaccc gctacgtgac cgatgggccg tgccgcagcg ccaagccggt caccgagctg 240 gtgtgctccg gccagtgcgg cccggcgcgc ctgctgccca acgccatcgg ccgcggcaag 300 tggtggcgac ctagtgggcc cgacttccgc tgcatccccg accgctaccg cgcgcagcgc 360 gtgcagctgc tgtgtcccgg tggtgaggcg ccgcgcgcgc gcaaggtgcg cctggtggcc 420 tcgtgcaagt gcaagcgcct cacccgcttc cacaaccagt cggagctcaa ggacttcggg 480 accgaggccg ctcggccgca gaagggccgg aagccgcggc cccgcgcccg gagcgccaaa 540 gccaaccagg ccgagctgga gaacgcctac 570 101 570 DNA Rattus norvegicus 101 caggggtggc aagccttcaa gaatgatgcc acagaaatca tcccgggact cagagagtac 60 ccagagcctc ctcaggaact agagaacaac cagaccatga accgggccga gaacggaggc 120 agaccccccc accatcctta tgacaccaaa gacgtgtccg agtacagctg ccgcgagctg 180 cactacaccc gcttcgtgac cgacggcccg tgccgcagtg ccaagccggt caccgagttg 240 gtgtgctcgg gccagtgcgg ccccgcgcgg ctgctgccca acgccatcgg gcgcgtgaag 300 tggtggcgcc cgaacggacc cgacttccgc tgcatcccgg atcgctaccg cgcgcagcgg 360 gtgcagctgc tgtgccccgg cggcgcggcg ccgcgctcgc gcaaggtgcg tctggtggcc 420 tcgtgcaagt gcaagcgcct cacccgcttc cacaaccagt cggagctcaa ggacttcgga 480 cctgagaccg cgcggccgca gaagggtcgc aagccgcggc cccgcgcccg gggagccaaa 540 gccaaccagg cggagctgga gaacgcctac 570 102 532 PRT Homo sapiens 102 Met Thr Gln Leu Tyr Ile Tyr Ile Arg Leu Leu Gly Ala Tyr Leu Phe 1 5 10 15 Ile Ile Ser Arg Val Gln Gly Gln Asn Leu Asp Ser Met Leu His Gly 20 25 30 Thr Gly Met Lys Ser Asp Ser Asp Gln Lys Lys Ser Glu Asn Gly Val 35 40 45 Thr Leu Ala Pro Glu Asp Thr Leu Pro Phe Leu Lys Cys Tyr Cys Ser 50 55 60 Gly His Cys Pro Asp Asp Ala Ile Asn Asn Thr Cys Ile Thr Asn Gly 65 70 75 80 His Cys Phe Ala Ile Ile Glu Glu Asp Asp Gln Gly Glu Thr Thr Leu 85 90 95 Ala Ser Gly Cys Met Lys Tyr Glu Gly Ser Asp Phe Gln Cys Lys Asp 100 105 110 Ser Pro Lys Ala Gln Leu Arg Arg Thr Ile Glu Cys Cys Arg Thr Asn 115 120 125 Leu Cys Asn Gln Tyr Leu Gln Pro Thr Leu Pro Pro Val Val Ile Gly 130 135 140 Pro Phe Phe Asp Gly Ser Ile Arg Trp Leu Val Leu Leu Ile Ser Met 145 150 155 160 Ala Val Cys Ile Ile Ala Met Ile Ile Phe Ser Ser Cys Phe Cys Tyr 165 170 175 Lys His Tyr Cys Lys Ser Ile Ser Ser Arg Arg Arg Tyr Asn Arg Asp 180 185 190 Leu Glu Gln Asp Glu Ala Phe Ile Pro Val Gly Glu Ser Leu Lys Asp 195 200 205 Leu Ile Asp Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu Pro Leu 210 215 220 Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Arg Gln Val 225 230 235 240 Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg Gly Glu 245 250 255 Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser Trp Phe 260 265 270 Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu Asn Ile 275 280 285 Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp Thr Gln 290 295 300 Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr Asp Phe 305 310 315 320 Leu Lys Cys Ala Thr Leu Asp Thr Arg Ala Leu Leu Lys Leu Ala Tyr 325 330 335 Ser Ala Ala Cys Gly Leu Cys His Leu His Thr Glu Ile Tyr Gly Thr 340 345 350 Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys Asn Ile 355 360 365 Leu Ile Lys Lys Asn Gly Ser Cys Cys Ile Ala Asp Leu Gly Leu Ala 370 375 380 Val Lys Phe Asn Ser Asp Thr Asn Glu Val Asp Val Pro Leu Asn Thr 385 390 395 400 Arg Val Gly Thr Lys Arg Tyr Met Ala Pro Glu Val Leu Asp Glu Ser 405 410 415 Leu Asn Lys Asn His Phe Gln Pro Tyr Ile Met Ala Asp Ile Tyr Ser 420 425 430 Phe Gly Leu Ile Ile Trp Glu Met Ala Arg Arg Cys Ile Thr Gly Gly 435 440 445 Ile Val Glu Glu Tyr Gln Leu Pro Tyr Tyr Asn Met Val Pro Ser Asp 450 455 460 Pro Ser Tyr Glu Asp Met Arg Glu Val Val Cys Val Lys Arg Leu Arg 465 470 475 480 Pro Ile Val Ser Asn Arg Trp Asn Ser Asp Glu Cys Leu Arg Ala Val 485 490 495 Leu Lys Leu Met Ser Glu Cys Trp Ala His Asn Pro Ala Ser Arg Leu 500 505 510 Thr Ala Leu Arg Ile Lys Lys Thr Leu Ala Lys Met Val Glu Ser Gln 515 520 525 Asp Val Lys Ile 530 103 502 PRT Homo sapiens 103 Met Leu Leu Arg Ser Ala Gly Lys Leu Asn Val Gly Thr Lys Lys Glu 1 5 10 15 Asp Gly Glu Ser Thr Ala Pro Thr Pro Arg Pro Lys Val Leu Arg Cys 20 25 30 Lys Cys His His His Cys Pro Glu Asp Ser Val Asn Asn Ile Cys Ser 35 40 45 Thr Asp Gly Tyr Cys Phe Thr Met Ile Glu Glu Asp Asp Ser Gly Leu 50 55 60 Pro Val Val Thr Ser Gly Cys Leu Gly Leu Glu Gly Ser Asp Phe Gln 65 70 75 80 Cys Arg Asp Thr Pro Ile Pro His Gln Arg Arg Ser Ile Glu Cys Cys 85 90 95 Thr Glu Arg Asn Glu Cys Asn Lys Asp Leu His Pro Thr Leu Pro Pro 100 105 110 Leu Lys Asn Arg Asp Phe Val Asp Gly Pro Ile His His Arg Ala Leu 115 120 125 Leu Ile Ser Val Thr Val Cys Ser Leu Leu Leu Val Leu Ile Ile Leu 130 135 140 Phe Cys Tyr Phe Arg Tyr Lys Arg Gln Glu Thr Arg Pro Arg Tyr Ser 145 150 155 160 Ile Gly Leu Glu Gln Asp Glu Thr Tyr Ile Pro Pro Gly Glu Ser Leu 165 170 175 Arg Asp Leu Ile Glu Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu 180 185 190 Pro Leu Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Lys 195 200 205 Gln Ile Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg 210 215 220 Gly Glu Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser 225 230 235 240 Trp Phe Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu 245 250 255 Asn Ile Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp 260 265 270 Thr Gln Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr 275 280 285 Asp Tyr Leu Lys Ser Thr Thr Leu Asp Ala Lys Ser Met Leu Lys Leu 290 295 300 Ala Tyr Ser Ser Val Ser Gly Leu Cys His Leu His Thr Glu Ile Phe 305 310 315 320 Ser Thr Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys 325 330 335 Asn Ile Leu Val Lys Lys Asn Gly Thr Cys Cys Ile Ala Asp Leu Gly 340 345 350 Leu Ala Val Lys Phe Ile Ser Asp Thr Asn Glu Val Asp Ile Pro Pro 355 360 365 Asn Thr Arg Val Gly Thr Lys Arg Tyr Met Pro Pro Glu Val Leu Asp 370 375 380 Glu Ser Leu Asn Arg Asn His Phe Gln Ser Tyr Ile Met Ala Asp Met 385 390 395 400 Tyr Ser Phe Gly Leu Ile Leu Trp Glu Val Ala Arg Arg Cys Val Ser 405 410 415 Gly Gly Ile Val Glu Glu Tyr Gln Leu Pro Tyr His Asp Leu Val Pro 420 425 430 Ser Asp Pro Ser Tyr Glu Asp Met Arg Glu Ile Val Cys Ile Lys Lys 435 440 445 Leu Arg Pro Ser Phe Pro Asn Arg Trp Ser Ser Asp Glu Cys Leu Arg 450 455 460 Gln Met Gly Lys Leu Met Thr Glu Cys Trp Ala His Asn Pro Ala Ser 465 470 475 480 Arg Leu Thr Ala Leu Arg Val Lys Lys Thr Leu Ala Lys Met Ser Glu 485 490 495 Ser Gln Asp Ile Lys Leu 500 104 502 PRT Homo sapiens 104 Met Leu Leu Arg Ser Ala Gly Lys Leu Asn Val Gly Thr Lys Lys Glu 1 5 10 15 Asp Gly Glu Ser Thr Ala Pro Thr Pro Arg Pro Lys Val Leu Arg Cys 20 25 30 Lys Cys His His His Cys Pro Glu Asp Ser Val Asn Asn Ile Cys Ser 35 40 45 Thr Asp Gly Tyr Cys Phe Thr Met Ile Glu Glu Asp Asp Ser Gly Leu 50 55 60 Pro Val Val Thr Ser Gly Cys Leu Gly Leu Glu Gly Ser Asp Phe Gln 65 70 75 80 Cys Arg Asp Thr Pro Ile Pro His Gln Arg Arg Ser Ile Glu Cys Cys 85 90 95 Thr Glu Arg Asn Glu Cys Asn Lys Asp Leu His Pro Thr Leu Pro Pro 100 105 110 Leu Lys Asn Arg Asp Phe Val Asp Gly Pro Ile His His Arg Ala Leu 115 120 125 Leu Ile Ser Val Thr Val Cys Ser Leu Leu Leu Val Leu Ile Ile Leu 130 135 140 Phe Cys Tyr Phe Arg Tyr Lys Arg Gln Glu Thr Arg Pro Arg Tyr Ser 145 150 155 160 Ile Gly Leu Glu Gln Asp Glu Thr Tyr Ile Pro Pro Gly Glu Ser Leu 165 170 175 Arg Asp Leu Ile Glu Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu 180 185 190 Pro Leu Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Lys 195 200 205 Gln Ile Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg 210 215 220 Gly Glu Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser 225 230 235 240 Trp Phe Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu 245 250 255 Asn Ile Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp 260 265 270 Thr Gln Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr 275 280 285 Asp Tyr Leu Lys Ser Thr Thr Leu Asp Ala Lys Ser Met Leu Lys Leu 290 295 300 Ala Tyr Ser Ser Val Ser Gly Leu Cys His Leu His Thr Glu Ile Phe 305 310 315 320 Ser Thr Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys 325 330 335 Asn Ile Leu Val Lys Lys Asn Gly Thr Cys Cys Ile Ala Asp Leu Gly 340 345 350 Leu Ala Val Lys Phe Ile Ser Asp Thr Asn Glu Val Asp Ile Pro Pro 355 360 365 Asn Thr Arg Val Gly Thr Lys Arg Tyr Met Pro Pro Glu Val Leu Asp 370 375 380 Glu Ser Leu Asn Arg Asn His Phe Gln Ser Tyr Ile Met Ala Asp Met 385 390 395 400 Tyr Ser Phe Gly Leu Ile Leu Trp Glu Val Ala Arg Arg Cys Val Ser 405 410 415 Gly Gly Ile Val Glu Glu Tyr Gln Leu Pro Tyr His Asp Leu Val Pro 420 425 430 Ser Asp Pro Ser Tyr Glu Asp Met Arg Glu Ile Val Cys Ile Lys Lys 435 440 445 Leu Arg Pro Ser Phe Pro Asn Arg Trp Ser Ser Asp Glu Cys Leu Arg 450 455 460 Gln Met Gly Lys Leu Met Thr Glu Cys Trp Ala His Asn Pro Ala Ser 465 470 475 480 Arg Leu Thr Ala Leu Arg Val Lys Lys Thr Leu Ala Lys Met Ser Glu 485 490 495 Ser Gln Asp Ile Lys Leu 500 105 532 PRT Rattus sp. 105 Met Thr Gln Leu Tyr Thr Tyr Ile Arg Leu Leu Gly Ala Cys Leu Phe 1 5 10 15 Ile Ile Ser His Val Gln Gly Gln Asn Leu Asp Ser Met Leu His Gly 20 25 30 Thr Gly Met Lys Ser Asp Val Asp Gln Lys Lys Pro Glu Asn Gly Val 35 40 45 Thr Leu Ala Pro Glu Asp Thr Leu Pro Phe Leu Lys Cys Tyr Cys Ser 50 55 60 Gly His Cys Pro Asp Asp Ala Ile Asn Asn Thr Cys Ile Thr Asn Gly 65 70 75 80 His Cys Phe Ala Ile Ile Glu Glu Asp Asp Gln Gly Glu Thr Thr Leu 85 90 95 Thr Ser Gly Cys Met Lys Tyr Glu Gly Ser Asp Phe Gln Cys Lys Asp 100 105 110 Ser Pro Lys Ala Gln Leu Arg Arg Thr Ile Glu Cys Cys Arg Thr Asn 115 120 125 Leu Cys Asn Gln Tyr Leu Gln Pro Thr Leu Pro Pro Val Val Ile Gly 130 135 140 Pro Phe Phe Asp Gly Ser Val Arg Trp Leu Ala Val Leu Ile Ser Met 145 150 155 160 Ala Val Cys Ile Val Ala Met Ile Val Phe Ser Ser Cys Phe Cys Tyr 165 170 175 Lys His Tyr Cys Lys Ser Ile Ser Ser Arg Gly Arg Tyr Asn Arg Asp 180 185 190 Leu Glu Gln Asp Glu Ala Phe Ile Pro Val Gly Glu Ser Leu Lys Asp 195 200 205 Leu Ile Asp Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu Pro Leu 210 215 220 Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Arg Gln Val 225 230 235 240 Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg Gly Glu 245 250 255 Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser Trp Phe 260 265 270 Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu Asn Ile 275 280 285 Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp Thr Gln 290 295 300 Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr Asp Phe 305 310 315 320 Leu Lys Cys Ala Thr Leu Asp Thr Arg Ala Leu Leu Lys Leu Ala Tyr 325 330 335 Ser Ala Ala Cys Gly Leu Cys His Leu His Thr Glu Ile Tyr Gly Thr 340 345 350 Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys Asn Ile 355 360 365 Leu Ile Lys Lys Asn Gly Ser Cys Cys Ile Ala Asp Leu Gly Leu Ala 370 375 380 Val Lys Phe Asn Ser Asp Thr Asn Glu Val Asp Ile Pro Leu Asn Thr 385 390 395 400 Arg Val Gly Thr Arg Arg Tyr Met Ala Pro Glu Val Leu Asp Glu Ser 405 410 415 Leu Ser Lys Asn His Phe Gln Pro Tyr Ile Met Ala Asp Ile Tyr Ser 420 425 430 Phe Gly Leu Ile Ile Trp Glu Met Ala Arg Arg Cys Ile Thr Gly Gly 435 440 445 Ile Val Glu Glu Tyr Gln Leu Pro Tyr Tyr Asn Met Val Pro Ser Asp 450 455 460 Pro Ser Tyr Glu Asp Met Arg Glu Val Val Cys Val Lys Arg Leu Arg 465 470 475 480 Pro Ile Val Ser Asn Arg Trp Asn Ser Asp Glu Cys Leu Arg Ala Val 485 490 495 Leu Lys Leu Met Ser Glu Cys Trp Ala His Asn Pro Ala Ser Arg Leu 500 505 510 Thr Ala Leu Arg Ile Lys Lys Thr Leu Ala Lys Met Val Glu Ser Gln 515 520 525 Asp Val Lys Ile 530 106 532 PRT Rattus norvegicus 106 Met Thr Gln Leu Tyr Thr Tyr Ile Arg Leu Leu Gly Ala Cys Leu Phe 1 5 10 15 Ile Ile Ser His Val Gln Gly Gln Asn Leu Asp Ser Met Leu His Gly 20 25 30 Thr Gly Met Lys Ser Asp Val Asp Gln Lys Lys Pro Glu Asn Gly Val 35 40 45 Thr Leu Ala Pro Glu Asp Thr Leu Pro Phe Leu Lys Cys Tyr Cys Ser 50 55 60 Gly His Cys Pro Asp Asp Ala Ile Asn Asn Thr Cys Ile Thr Asn Gly 65 70 75 80 His Cys Phe Ala Ile Ile Glu Glu Asp Asp Gln Gly Glu Thr Thr Leu 85 90 95 Thr Ser Gly Cys Met Lys Tyr Glu Gly Ser Asp Phe Gln Cys Lys Asp 100 105 110 Ser Pro Lys Ala Gln Leu Arg Arg Thr Ile Glu Cys Cys Arg Thr Asn 115 120 125 Leu Cys Asn Gln Tyr Leu Gln Pro Thr Leu Pro Pro Val Val Ile Gly 130 135 140 Pro Phe Phe Asp Gly Ser Val Arg Trp Leu Ala Val Leu Ile Ser Met 145 150 155 160 Ala Val Cys Ile Val Ala Met Ile Val Phe Ser Ser Cys Phe Cys Tyr 165 170 175 Lys His Tyr Cys Lys Ser Ile Ser Ser Arg Gly Arg Tyr Asn Arg Asp 180 185 190 Leu Glu Gln Asp Glu Ala Phe Ile Pro Val Gly Glu Ser Leu Lys Asp 195 200 205 Leu Ile Asp Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu Pro Leu 210 215 220 Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Arg Gln Val 225 230 235 240 Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg Gly Glu 245 250 255 Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser Trp Phe 260 265 270 Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu Asn Ile 275 280 285 Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp Thr Gln 290 295 300 Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr Asp Phe 305 310 315 320 Leu Lys Cys Ala Thr Leu Asp Thr Arg Ala Leu Leu Lys Leu Ala Tyr 325 330 335 Ser Ala Ala Cys Gly Leu Cys His Leu His Thr Glu Ile Tyr Gly Thr 340 345 350 Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys Asn Ile 355 360 365 Leu Ile Lys Lys Asn Gly Ser Cys Cys Ile Ala Asp Leu Gly Leu Ala 370 375 380 Val Lys Phe Asn Ser Asp Thr Asn Glu Val Asp Ile Pro Leu Asn Thr 385 390 395 400 Arg Val Gly Thr Arg Arg Tyr Met Ala Pro Glu Val Leu Asp Glu Ser 405 410 415 Leu Ser Lys Asn His Phe Gln Pro Tyr Ile Met Ala Asp Ile Tyr Ser 420 425 430 Phe Gly Leu Ile Ile Trp Glu Met Ala Arg Arg Cys Ile Thr Gly Gly 435 440 445 Ile Val Glu Glu Tyr Gln Leu Pro Tyr Tyr Asn Met Val Pro Ser Asp 450 455 460 Pro Ser Tyr Glu Asp Met Arg Glu Val Val Cys Val Lys Arg Leu Arg 465 470 475 480 Pro Ile Val Ser Asn Arg Trp Asn Ser Asp Glu Cys Leu Arg Ala Val 485 490 495 Leu Lys Leu Met Ser Glu Cys Trp Ala His Asn Pro Ala Ser Arg Leu 500 505 510 Thr Ala Leu Arg Ile Lys Lys Thr Leu Ala Lys Met Val Glu Ser Gln 515 520 525 Asp Val Lys Ile 530 107 532 PRT Rattus norvegicus 107 Met Thr Gln Leu Tyr Thr Tyr Ile Arg Leu Leu Gly Ala Cys Leu Phe 1 5 10 15 Ile Ile Ser His Val Gln Gly Gln Asn Leu Asp Ser Met Leu His Gly 20 25 30 Thr Gly Met Lys Ser Asp Val Asp Gln Lys Lys Pro Glu Asn Gly Val 35 40 45 Thr Leu Ala Pro Glu Asp Thr Leu Pro Phe Leu Lys Cys Tyr Cys Ser 50 55 60 Gly His Cys Pro Asp Asp Ala Ile Asn Asn Thr Cys Ile Thr Asn Gly 65 70 75 80 His Cys Phe Ala Ile Ile Glu Glu Asp Asp Gln Gly Glu Thr Thr Leu 85 90 95 Thr Ser Gly Cys Met Lys Tyr Glu Gly Ser Asp Phe Gln Cys Lys Asp 100 105 110 Ser Pro Lys Ala Gln Leu Arg Arg Thr Ile Glu Cys Cys Arg Thr Asn 115 120 125 Leu Cys Asn Gln Tyr Leu Gln Pro Thr Leu Pro Pro Val Val Ile Gly 130 135 140 Pro Phe Phe Asp Gly Ser Val Arg Trp Leu Ala Val Leu Ile Ser Met 145 150 155 160 Ala Val Cys Ile Val Ala Met Ile Val Phe Ser Ser Cys Phe Cys Tyr 165 170 175 Lys His Tyr Cys Lys Ser Ile Ser Ser Arg Gly Arg Tyr Asn Arg Asp 180 185 190 Leu Glu Gln Asp Glu Ala Phe Ile Pro Val Gly Glu Ser Leu Lys Asp 195 200 205 Leu Ile Asp Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu Pro Leu 210 215 220 Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Arg Gln Val 225 230 235 240 Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg Gly Glu 245 250 255 Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser Trp Phe 260 265 270 Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu Asn Ile 275 280 285 Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp Thr Gln 290 295 300 Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr Asp Phe 305 310 315 320 Leu Lys Cys Ala Thr Leu Asp Thr Arg Ala Leu Leu Lys Leu Ala Tyr 325 330 335 Ser Ala Ala Cys Gly Leu Cys His Leu His Thr Glu Ile Tyr Gly Thr 340 345 350 Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys Asn Ile 355 360 365 Leu Ile Lys Lys Asn Gly Ser Cys Cys Ile Ala Asp Leu Gly Leu Ala 370 375 380 Val Lys Phe Asn Ser Asp Thr Asn Glu Val Asp Ile Pro Leu Asn Thr 385 390 395 400 Arg Val Gly Thr Arg Arg Tyr Met Ala Pro Glu Val Leu Asp Glu Ser 405 410 415 Leu Ser Lys Asn His Phe Gln Pro Tyr Ile Met Ala Asp Ile Tyr Ser 420 425 430 Phe Gly Leu Ile Ile Trp Glu Met Ala Arg Arg Cys Ile Thr Gly Gly 435 440 445 Ile Val Glu Glu Tyr Gln Leu Pro Tyr Tyr Asn Met Val Pro Ser Asp 450 455 460 Pro Ser Tyr Glu Asp Met Arg Glu Val Val Cys Val Lys Arg Leu Arg 465 470 475 480 Pro Ile Val Ser Asn Arg Trp Asn Ser Asp Glu Cys Leu Arg Ala Val 485 490 495 Leu Lys Leu Met Ser Glu Cys Trp Ala His Asn Pro Ala Ser Arg Leu 500 505 510 Thr Ala Leu Arg Ile Lys Lys Thr Leu Ala Lys Met Val Glu Ser Gln 515 520 525 Asp Val Lys Ile 530 108 502 PRT Homo sapiens 108 Met Leu Leu Arg Ser Ala Gly Lys Leu Asn Val Gly Thr Lys Lys Glu 1 5 10 15 Asp Gly Glu Ser Thr Ala Pro Thr Pro Arg Pro Lys Val Leu Arg Cys 20 25 30 Lys Cys His His His Cys Pro Glu Asp Ser Val Asn Asn Ile Cys Ser 35 40 45 Thr Asp Gly Tyr Cys Phe Thr Met Ile Glu Glu Asp Asp Ser Gly Leu 50 55 60 Pro Val Val Thr Ser Gly Cys Leu Gly Leu Glu Gly Ser Asp Phe Gln 65 70 75 80 Cys Arg Asp Thr Pro Ile Pro His Gln Arg Arg Ser Ile Glu Cys Cys 85 90 95 Thr Glu Arg Asn Glu Cys Asn Lys Asp Leu His Pro Thr Leu Pro Pro 100 105 110 Leu Lys Asn Arg Asp Phe Val Asp Gly Pro Ile His His Arg Ala Leu 115 120 125 Leu Ile Ser Val Thr Val Cys Ser Leu Leu Leu Val Leu Ile Ile Leu 130 135 140 Phe Cys Tyr Phe Arg Tyr Lys Arg Gln Glu Thr Arg Pro Arg Tyr Ser 145 150 155 160 Ile Gly Leu Glu Gln Asp Glu Thr Tyr Ile Pro Pro Gly Glu Ser Leu 165 170 175 Arg Asp Leu Ile Glu Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu 180 185 190 Pro Leu Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Lys 195 200 205 Gln Ile Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg 210 215 220 Gly Glu Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser 225 230 235 240 Trp Phe Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu 245 250 255 Asn Ile Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp 260 265 270 Thr Gln Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr 275 280 285 Asp Tyr Leu Lys Ser Thr Thr Leu Asp Ala Lys Ser Met Leu Lys Leu 290 295 300 Ala Tyr Ser Ser Val Ser Gly Leu Cys His Leu His Thr Glu Ile Phe 305 310 315 320 Ser Thr Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys 325 330 335 Asn Ile Leu Val Lys Lys Asn Gly Thr Cys Cys Ile Ala Asp Leu Gly 340 345 350 Leu Ala Val Lys Phe Ile Ser Asp Thr Asn Glu Val Asp Ile Pro Pro 355 360 365 Asn Thr Arg Val Gly Thr Lys Arg Tyr Met Pro Pro Glu Val Leu Asp 370 375 380 Glu Ser Leu Asn Arg Asn His Phe Gln Ser Tyr Ile Met Ala Asp Met 385 390 395 400 Tyr Ser Phe Gly Leu Ile Leu Trp Glu Val Ala Arg Arg Cys Val Ser 405 410 415 Gly Gly Ile Val Glu Glu Tyr Gln Leu Pro Tyr His Asp Leu Val Pro 420 425 430 Ser Asp Pro Ser Tyr Glu Asp Met Arg Glu Ile Val Cys Ile Lys Lys 435 440 445 Leu Arg Pro Ser Phe Pro Asn Arg Trp Ser Ser Asp Glu Cys Leu Arg 450 455 460 Gln Met Gly Lys Leu Met Thr Glu Cys Trp Ala His Asn Pro Ala Ser 465 470 475 480 Arg Leu Thr Ala Leu Arg Val Lys Lys Thr Leu Ala Lys Met Ser Glu 485 490 495 Ser Gln Asp Ile Lys Leu 500 109 502 PRT Homo sapiens 109 Met Leu Leu Arg Ser Ala Gly Lys Leu Asn Val Gly Thr Lys Lys Glu 1 5 10 15 Asp Gly Glu Ser Thr Ala Pro Thr Pro Arg Pro Lys Val Leu Arg Cys 20 25 30 Lys Cys His His His Cys Pro Glu Asp Ser Val Asn Asn Ile Cys Ser 35 40 45 Thr Asp Gly Tyr Cys Phe Thr Met Ile Glu Glu Asp Asp Ser Gly Leu 50 55 60 Pro Val Val Thr Ser Gly Cys Leu Gly Leu Glu Gly Ser Asp Phe Gln 65 70 75 80 Cys Arg Asp Thr Pro Ile Pro His Gln Arg Arg Ser Ile Glu Cys Cys 85 90 95 Thr Glu Arg Asn Glu Cys Asn Lys Asp Leu His Pro Thr Leu Pro Pro 100 105 110 Leu Lys Asn Arg Asp Phe Val Asp Gly Pro Ile His His Arg Ala Leu 115 120 125 Leu Ile Ser Val Thr Val Cys Ser Leu Leu Leu Val Leu Ile Ile Leu 130 135 140 Phe Cys Tyr Phe Arg Tyr Lys Arg Gln Glu Thr Arg Pro Arg Tyr Ser 145 150 155 160 Ile Gly Leu Glu Gln Asp Glu Thr Tyr Ile Pro Pro Gly Glu Ser Leu 165 170 175 Arg Asp Leu Ile Glu Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu 180 185 190 Pro Leu Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Lys 195 200 205 Gln Ile Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg 210 215 220 Gly Glu Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser 225 230 235 240 Trp Phe Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu 245 250 255 Asn Ile Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp 260 265 270 Thr Gln Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr 275 280 285 Asp Tyr Leu Lys Ser Thr Thr Leu Asp Ala Lys Ser Met Leu Lys Leu 290 295 300 Ala Tyr Ser Ser Val Ser Gly Leu Cys His Leu His Thr Glu Ile Phe 305 310 315 320 Ser Thr Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys 325 330 335 Asn Ile Leu Val Lys Lys Asn Gly Thr Cys Cys Ile Ala Asp Leu Gly 340 345 350 Leu Ala Val Lys Phe Ile Ser Asp Thr Asn Glu Val Asp Ile Pro Pro 355 360 365 Asn Thr Arg Val Gly Thr Lys Arg Tyr Met Pro Pro Glu Val Leu Asp 370 375 380 Glu Ser Leu Asn Arg Asn His Phe Gln Ser Tyr Ile Met Ala Asp Met 385 390 395 400 Tyr Ser Phe Gly Leu Ile Leu Trp Glu Val Ala Arg Arg Cys Val Ser 405 410 415 Gly Gly Ile Val Glu Glu Tyr Gln Leu Pro Tyr His Asp Leu Val Pro 420 425 430 Ser Asp Pro Ser Tyr Glu Asp Met Arg Glu Ile Val Cys Ile Lys Lys 435 440 445 Leu Arg Pro Ser Phe Pro Asn Arg Trp Ser Ser Asp Glu Cys Leu Arg 450 455 460 Gln Met Gly Lys Leu Met Thr Glu Cys Trp Ala His Asn Pro Ala Ser 465 470 475 480 Arg Leu Thr Ala Leu Arg Val Lys Lys Thr Leu Ala Lys Met Ser Glu 485 490 495 Ser Gln Asp Ile Lys Leu 500 110 532 PRT Rattus sp. 110 Met Thr Gln Leu Tyr Thr Tyr Ile Arg Leu Leu Gly Ala Cys Leu Phe 1 5 10 15 Ile Ile Ser His Val Gln Gly Gln Asn Leu Asp Ser Met Leu His Gly 20 25 30 Thr Gly Met Lys Ser Asp Val Asp Gln Lys Lys Pro Glu Asn Gly Val 35 40 45 Thr Leu Ala Pro Glu Asp Thr Leu Pro Phe Leu Lys Cys Tyr Cys Ser 50 55 60 Gly His Cys Pro Asp Asp Ala Ile Asn Asn Thr Cys Ile Thr Asn Gly 65 70 75 80 His Cys Phe Ala Ile Ile Glu Glu Asp Asp Gln Gly Glu Thr Thr Leu 85 90 95 Thr Ser Gly Cys Met Lys Tyr Glu Gly Ser Asp Phe Gln Cys Lys Asp 100 105 110 Ser Pro Lys Ala Gln Leu Arg Arg Thr Ile Glu Cys Cys Arg Thr Asn 115 120 125 Leu Cys Asn Gln Tyr Leu Gln Pro Thr Leu Pro Pro Val Val Ile Gly 130 135 140 Pro Phe Phe Asp Gly Ser Val Arg Trp Leu Ala Val Leu Ile Ser Met 145 150 155 160 Ala Val Cys Ile Val Ala Met Ile Val Phe Ser Ser Cys Phe Cys Tyr 165 170 175 Lys His Tyr Cys Lys Ser Ile Ser Ser Arg Gly Arg Tyr Asn Arg Asp 180 185 190 Leu Glu Gln Asp Glu Ala Phe Ile Pro Val Gly Glu Ser Leu Lys Asp 195 200 205 Leu Ile Asp Gln Ser Gln Ser Ser Gly Ser Gly Ser Gly Leu Pro Leu 210 215 220 Leu Val Gln Arg Thr Ile Ala Lys Gln Ile Gln Met Val Arg Gln Val 225 230 235 240 Gly Lys Gly Arg Tyr Gly Glu Val Trp Met Gly Lys Trp Arg Gly Glu 245 250 255 Lys Val Ala Val Lys Val Phe Phe Thr Thr Glu Glu Ala Ser Trp Phe 260 265 270 Arg Glu Thr Glu Ile Tyr Gln Thr Val Leu Met Arg His Glu Asn Ile 275 280 285 Leu Gly Phe Ile Ala Ala Asp Ile Lys Gly Thr Gly Ser Trp Thr Gln 290 295 300 Leu Tyr Leu Ile Thr Asp Tyr His Glu Asn Gly Ser Leu Tyr Asp Phe 305 310 315 320 Leu Lys Cys Ala Thr Leu Asp Thr Arg Ala Leu Leu Lys Leu Ala Tyr 325 330 335 Ser Ala Ala Cys Gly Leu Cys His Leu His Thr Glu Ile Tyr Gly Thr 340 345 350 Gln Gly Lys Pro Ala Ile Ala His Arg Asp Leu Lys Ser Lys Asn Ile 355 360 365 Leu Ile Lys Lys Asn Gly Ser Cys Cys Ile Ala Asp Leu Gly Leu Ala 370 375 380 Val Lys Phe Asn Ser Asp Thr Asn Glu Val Asp Ile Pro Leu Asn Thr 385 390 395 400 Arg Val Gly Thr Arg Arg Tyr Met Ala Pro Glu Val Leu Asp Glu Ser 405 410 415 Leu Ser Lys Asn His Phe Gln Pro Tyr Ile Met Ala Asp Ile Tyr Ser 420 425 430 Phe Gly Leu Ile Ile Trp Glu Met Ala Arg Arg Cys Ile Thr Gly Gly 435 440 445 Ile Val Glu Glu Tyr Gln Leu Pro Tyr Tyr Asn Met Val Pro Ser Asp 450 455 460 Pro Ser Tyr Glu Asp Met Arg Glu Val Val Cys Val Lys Arg Leu Arg 465 470 475 480 Pro Ile Val Ser Asn Arg Trp Asn Ser Asp Glu Cys Leu Arg Ala Val 485 490 495 Leu Lys Leu Met Ser Glu Cys Trp Ala His Asn Pro Ala Ser Arg Leu 500 505 510 Thr Ala Leu Arg Ile Lys Lys Thr Leu Ala Lys Met Val Glu Ser Gln 515 520 525 Asp Val Lys Ile 530 111 530 PRT Homo sapiens 111 Met Thr Ser Ser Leu Gln Arg Pro Trp Arg Val Pro Trp Leu Pro Trp 1 5 10 15 Thr Ile Leu Leu Val Ser Thr Ala Ala Ala Ser Gln Asn Gln Glu Arg 20 25 30 Leu Cys Ala Phe Lys Asp Pro Tyr Gln Gln Asp Leu Gly Ile Gly Glu 35 40 45 Ser Arg Ile Ser His Glu Asn Gly Thr Ile Leu Cys Ser Lys Gly Ser 50 55 60 Thr Cys Tyr Gly Leu Trp Glu Lys Ser Lys Gly Asp Ile Asn Leu Val 65 70 75 80 Lys Gln Gly Cys Trp Ser His Ile Gly Asp Pro Gln Glu Cys His Tyr 85 90 95 Glu Glu Cys Val Val Thr Thr Thr Pro Pro Ser Ile Gln Asn Gly Thr 100 105 110 Tyr Arg Phe Cys Cys Cys Ser Thr Asp Leu Cys Asn Val Asn Phe Thr 115 120 125 Glu Asn Phe Pro Pro Pro Asp Thr Thr Pro Leu Ser Pro Pro His Ser 130 135 140 Phe Asn Arg Asp Glu Thr Ile Ile Ile Ala Leu Ala Ser Val Ser Val 145 150 155 160 Leu Ala Val Leu Ile Val Ala Leu Cys Phe Gly Tyr Arg Met Leu Thr 165 170 175 Gly Asp Arg Lys Gln Gly Leu His Ser Met Asn Met Met Glu Ala Ala 180 185 190 Ala Ser Glu Pro Ser Leu Asp Leu Asp Asn Leu Lys Leu Leu Glu Leu 195 200 205 Ile Gly Arg Gly Arg Tyr Gly Ala Val Tyr Lys Gly Ser Leu Asp Glu 210 215 220 Arg Pro Val Ala Val Lys Val Phe Ser Phe Ala Asn Arg Gln Asn Phe 225 230 235 240 Ile Asn Glu Lys Asn Ile Tyr Arg Val Pro Leu Met Glu His Asp Asn 245 250 255 Ile Ala Arg Phe Ile Val Gly Asp Glu Arg Val Thr Ala Asp Gly Arg 260 265 270 Met Glu Tyr Leu Leu Val Met Glu Tyr Tyr Pro Asn Gly Ser Leu Cys 275 280 285 Lys Tyr Leu Ser Leu His Thr Ser Asp Trp Val Ser Ser Cys Arg Leu 290 295 300 Ala His Ser Val Thr Arg Gly Leu Ala Tyr Leu His Thr Glu Leu Pro 305 310 315 320 Arg Gly Asp His Tyr Lys Pro Ala Ile Ser His Arg Asp Leu Asn Ser 325 330 335 Arg Asn Val Leu Val Lys Asn Asp Gly Thr Cys Val Ile Ser Asp Phe 340 345 350 Gly Leu Ser Met Arg Leu Thr Gly Asn Arg Leu Val Arg Pro Gly Glu 355 360 365 Glu Asp Asn Ala Ala Ile Ser Glu Val Gly Thr Ile Arg Tyr Met Ala 370 375 380 Pro Glu Val Leu Glu Gly Ala Val Asn Leu Arg Asp Cys Glu Ser Ala 385 390 395 400 Leu Lys Gln Val Asp Met Tyr Ala Leu Gly Leu Ile Tyr Trp Glu Ile 405 410 415 Phe Met Arg Cys Thr Asp Leu Phe Pro Gly Glu Ser Val Pro Glu Tyr 420 425 430 Gln Met Ala Phe Gln Thr Glu Val Gly Asn His Pro Thr Phe Glu Asp 435 440 445 Met Gln Val Leu Val Ser Arg Glu Lys Gln Arg Pro Lys Phe Pro Glu 450 455 460 Ala Trp Lys Glu Asn Ser Leu Ala Val Arg Ser Leu Lys Glu Thr Ile 465 470 475 480 Glu Asp Cys Trp Asp Gln Asp Ala Glu Ala Arg Leu Thr Ala Gln Cys 485 490 495 Ala Glu Glu Arg Met Ala Glu Leu Met Met Ile Trp Glu Arg Asn Lys 500 505 510 Ser Val Ser Pro Thr Val Asn Pro Met Ser Thr Ala Met Gln Asn Glu 515 520 525 Arg Arg 530 112 530 PRT Homo sapiens 112 Met Thr Ser Ser Leu Gln Arg Pro Trp Arg Val Pro Trp Leu Pro Trp 1 5 10 15 Thr Ile Leu Leu Val Ser Thr Ala Ala Ala Ser Gln Asn Gln Glu Arg 20 25 30 Leu Cys Ala Phe Lys Asp Pro Tyr Gln Gln Asp Leu Gly Ile Gly Glu 35 40 45 Ser Arg Ile Ser His Glu Asn Gly Thr Ile Leu Cys Ser Lys Gly Ser 50 55 60 Thr Cys Tyr Gly Leu Trp Glu Lys Ser Lys Gly Asp Ile Asn Leu Val 65 70 75 80 Lys Gln Gly Cys Trp Ser His Ile Gly Asp Pro Gln Glu Cys His Tyr 85 90 95 Glu Glu Cys Val Val Thr Thr Thr Pro Pro Ser Ile Gln Asn Gly Thr 100 105 110 Tyr Arg Phe Cys Cys Cys Ser Thr Asp Leu Cys Asn Val Asn Phe Thr 115 120 125 Glu Asn Phe Pro Pro Pro Asp Thr Thr Pro Leu Ser Pro Pro His Ser 130 135 140 Phe Asn Arg Asp Glu Thr Ile Ile Ile Ala Leu Ala Ser Val Ser Val 145 150 155 160 Leu Ala Val Leu Ile Val Ala Leu Cys Phe Gly Tyr Arg Met Leu Thr 165 170 175 Gly Asp Arg Lys Gln Gly Leu His Ser Met Asn Met Met Glu Ala Ala 180 185 190 Ala Ser Glu Pro Ser Leu Asp Leu Asp Asn Leu Lys Leu Leu Glu Leu 195 200 205 Ile Gly Arg Gly Arg Tyr Gly Ala Val Tyr Lys Gly Ser Leu Asp Glu 210 215 220 Arg Pro Val Ala Val Lys Val Phe Ser Phe Ala Asn Arg Gln Asn Phe 225 230 235 240 Ile Asn Glu Lys Asn Ile Tyr Arg Val Pro Leu Met Glu His Asp Asn 245 250 255 Ile Ala Arg Phe Ile Val Gly Asp Glu Arg Val Thr Ala Asp Gly Arg 260 265 270 Met Glu Tyr Leu Leu Val Met Glu Tyr Tyr Pro Asn Gly Ser Leu Cys 275 280 285 Lys Tyr Leu Ser Leu His Thr Ser Asp Trp Val Ser Ser Cys Arg Leu 290 295 300 Ala His Ser Val Thr Arg Gly Leu Ala Tyr Leu His Thr Glu Leu Pro 305 310 315 320 Arg Gly Asp His Tyr Lys Pro Ala Ile Ser His Arg Asp Leu Asn Ser 325 330 335 Arg Asn Val Leu Val Lys Asn Asp Gly Thr Cys Val Ile Ser Asp Phe 340 345 350 Gly Leu Ser Met Arg Leu Thr Gly Asn Arg Leu Val Arg Pro Gly Glu 355 360 365 Glu Asp Asn Ala Ala Ile Ser Glu Val Gly Thr Ile Arg Tyr Met Ala 370 375 380 Pro Glu Val Leu Glu Gly Ala Val Asn Leu Arg Asp Cys Glu Ser Ala 385 390 395 400 Leu Lys Gln Val Asp Met Tyr Ala Leu Gly Leu Ile Tyr Trp Glu Ile 405 410 415 Phe Met Arg Cys Thr Asp Leu Phe Pro Gly Glu Ser Val Pro Glu Tyr 420 425 430 Gln Met Ala Phe Gln Thr Glu Val Gly Asn His Pro Thr Phe Glu Asp 435 440 445 Met Gln Val Leu Val Ser Arg Glu Lys Gln Arg Pro Lys Phe Pro Glu 450 455 460 Ala Trp Lys Glu Asn Ser Leu Ala Val Arg Ser Leu Lys Glu Thr Ile 465 470 475 480 Glu Asp Cys Trp Asp Gln Asp Ala Glu Ala Arg Leu Thr Ala Gln Cys 485 490 495 Ala Glu Glu Arg Met Ala Glu Leu Met Met Ile Trp Glu Arg Asn Lys 500 505 510 Ser Val Ser Pro Thr Val Asn Pro Met Ser Thr Ala Met Gln Asn Glu 515 520 525 Arg Arg 530 113 1038 PRT Homo sapiens 113 Met Thr Ser Ser Leu Gln Arg Pro Trp Arg Val Pro Trp Leu Pro Trp 1 5 10 15 Thr Ile Leu Leu Val Ser Thr Ala Ala Ala Ser Gln Asn Gln Glu Arg 20 25 30 Leu Cys Ala Phe Lys Asp Pro Tyr Gln Gln Asp Leu Gly Ile Gly Glu 35 40 45 Ser Arg Ile Ser His Glu Asn Gly Thr Ile Leu Cys Ser Lys Gly Ser 50 55 60 Thr Cys Tyr Gly Leu Trp Glu Lys Ser Lys Gly Asp Ile Asn Leu Val 65 70 75 80 Lys Gln Gly Cys Trp Ser His Ile Gly Asp Pro Gln Glu Cys His Tyr 85 90 95 Glu Glu Cys Val Val Thr Thr Thr Pro Pro Ser Ile Gln Asn Gly Thr 100 105 110 Tyr Arg Phe Cys Cys Cys Ser Thr Asp Leu Cys Asn Val Asn Phe Thr 115 120 125 Glu Asn Phe Pro Pro Pro Asp Thr Thr Pro Leu Ser Pro Pro His Ser 130 135 140 Phe Asn Arg Asp Glu Thr Ile Ile Ile Ala Leu Ala Ser Val Ser Val 145 150 155 160 Leu Ala Val Leu Ile Val Ala Leu Cys Phe Gly Tyr Arg Met Leu Thr 165 170 175 Gly Asp Arg Lys Gln Gly Leu His Ser Met Asn Met Met Glu Ala Ala 180 185 190 Ala Ser Glu Pro Ser Leu Asp Leu Asp Asn Leu Lys Leu Leu Glu Leu 195 200 205 Ile Gly Arg Gly Arg Tyr Gly Ala Val Tyr Lys Gly Ser Leu Asp Glu 210 215 220 Arg Pro Val Ala Val Lys Val Phe Ser Phe Ala Asn Arg Gln Asn Phe 225 230 235 240 Ile Asn Glu Lys Asn Ile Tyr Arg Val Pro Leu Met Glu His Asp Asn 245 250 255 Ile Ala Arg Phe Ile Val Gly Asp Glu Arg Val Thr Ala Asp Gly Arg 260 265 270 Met Glu Tyr Leu Leu Val Met Glu Tyr Tyr Pro Asn Gly Ser Leu Cys 275 280 285 Lys Tyr Leu Ser Leu His Thr Ser Asp Trp Val Ser Ser Cys Arg Leu 290 295 300 Ala His Ser Val Thr Arg Gly Leu Ala Tyr Leu His Thr Glu Leu Pro 305 310 315 320 Arg Gly Asp His Tyr Lys Pro Ala Ile Ser His Arg Asp Leu Asn Ser 325 330 335 Arg Asn Val Leu Val Lys Asn Asp Gly Thr Cys Val Ile Ser Asp Phe 340 345 350 Gly Leu Ser Met Arg Leu Thr Gly Asn Arg Leu Val Arg Pro Gly Glu 355 360 365 Glu Asp Asn Ala Ala Ile Ser Glu Val Gly Thr Ile Arg Tyr Met Ala 370 375 380 Pro Glu Val Leu Glu Gly Ala Val Asn Leu Arg Asp Cys Glu Ser Ala 385 390 395 400 Leu Lys Gln Val Asp Met Tyr Ala Leu Gly Leu Ile Tyr Trp Glu Ile 405 410 415 Phe Met Arg Cys Thr Asp Leu Phe Pro Gly Glu Ser Val Pro Glu Tyr 420 425 430 Gln Met Ala Phe Gln Thr Glu Val Gly Asn His Pro Thr Phe Glu Asp 435 440 445 Met Gln Val Leu Val Ser Arg Glu Lys Gln Arg Pro Lys Phe Pro Glu 450 455 460 Ala Trp Lys Glu Asn Ser Leu Ala Val Arg Ser Leu Lys Glu Thr Ile 465 470 475 480 Glu Asp Cys Trp Asp Gln Asp Ala Glu Ala Arg Leu Thr Ala Gln Cys 485 490 495 Ala Glu Glu Arg Met Ala Glu Leu Met Met Ile Trp Glu Arg Asn Lys 500 505 510 Ser Val Ser Pro Thr Val Asn Pro Met Ser Thr Ala Met Gln Asn Glu 515 520 525 Arg Asn Leu Ser His Asn Arg Arg Val Pro Lys Ile Gly Pro Tyr Pro 530 535 540 Asp Tyr Ser Ser Ser Ser Tyr Ile Glu Asp Ser Ile His His Thr Asp 545 550 555 560 Ser Ile Val Lys Asn Ile Ser Ser Glu His Ser Met Ser Ser Thr Pro 565 570 575 Leu Thr Ile Gly Glu Lys Asn Arg Asn Ser Ile Asn Tyr Glu Arg Gln 580 585 590 Gln Ala Gln Ala Arg Ile Pro Ser Pro Glu Thr Ser Val Thr Ser Leu 595 600 605 Ser Thr Asn Thr Thr Thr Thr Asn Thr Thr Gly Leu Thr Pro Ser Thr 610 615 620 Gly Met Thr Thr Ile Ser Glu Met Pro Tyr Pro Asp Glu Thr Asn Leu 625 630 635 640 His Thr Thr Asn Val Ala Gln Ser Ile Gly Pro Thr Pro Val Cys Leu 645 650 655 Gln Leu Thr Glu Glu Asp Leu Glu Thr Asn Lys Leu Asp Pro Lys Glu 660 665 670 Val Asp Lys Asn Leu Lys Glu Ser Ser Asp Glu Asn Leu Met Glu His 675 680 685 Ser Leu Lys Gln Phe Ser Gly Pro Asp Pro Leu Ser Ser Thr Ser Ser 690 695 700 Ser Leu Leu Tyr Pro Leu Ile Lys Leu Ala Val Glu Ala Thr Gly Gln 705 710 715 720 Gln Asp Phe Thr Gln Thr Ala Asn Gly Gln Ala Cys Leu Ile Pro Asp 725 730 735 Val Leu Pro Thr Gln Ile Tyr Pro Leu Pro Lys Gln Gln Asn Leu Pro 740 745 750 Lys Arg Pro Thr Ser Leu Pro Leu Asn Thr Lys Asn Ser Thr Lys Glu 755 760 765 Pro Arg Leu Lys Phe Gly Ser Lys His Lys Ser Asn Leu Lys Gln Val 770 775 780 Glu Thr Gly Val Ala Lys Met Asn Thr Ile Asn Ala Ala Glu Pro His 785 790 795 800 Val Val Thr Val Thr Met Asn Gly Val Ala Gly Arg Asn His Ser Val 805 810 815 Asn Ser His Ala Ala Thr Thr Gln Tyr Ala Asn Gly Thr Val Leu Ser 820 825 830 Gly Gln Thr Thr Asn Ile Val Thr His Arg Ala Gln Glu Met Leu Gln 835 840 845 Asn Gln Phe Ile Gly Glu Asp Thr Arg Leu Asn Ile Asn Ser Ser Pro 850 855 860 Asp Glu His Glu Pro Leu Leu Arg Arg Glu Gln Gln Ala Gly His Asp 865 870 875 880 Glu Gly Val Leu Asp Arg Leu Val Asp Arg Arg Glu Arg Pro Leu Glu 885 890 895 Gly Gly Arg Thr Asn Ser Asn Asn Asn Asn Ser Asn Pro Cys Ser Glu 900 905 910 Gln Asp Val Leu Ala Gln Gly Val Pro Ser Thr Ala Ala Asp Pro Gly 915 920 925 Pro Ser Lys Pro Arg Arg Ala Gln Arg Pro Asn Ser Leu Asp Leu Ser 930 935 940 Ala Thr Asn Val Leu Asp Gly Ser Ser Ile Gln Ile Gly Glu Ser Thr 945 950 955 960 Gln Asp Gly Lys Ser Gly Ser Gly Glu Lys Ile Lys Lys Arg Val Lys 965 970 975 Thr Pro Tyr Ser Leu Lys Arg Trp Arg Pro Ser Thr Trp Val Ile Ser 980 985 990 Thr Glu Ser Leu Asp Cys Glu Val Asn Asn Asn Gly Ser Asn Arg Ala 995 1000 1005 Val His Ser Lys Ser Ser Thr Ala Val Tyr Leu Ala Glu Gly Gly Thr 1010 1015 1020 Ala Thr Thr Met Val Ser Lys Asp Ile Gly Met Asn Cys Leu 1025 1030 1035 114 1038 PRT Homo sapiens 114 Met Thr Ser Ser Leu Gln Arg Pro Trp Arg Val Pro Trp Leu Pro Trp 1 5 10 15 Thr Ile Leu Leu Val Ser Thr Ala Ala Ala Ser Gln Asn Gln Glu Arg 20 25 30 Leu Cys Ala Phe Lys Asp Pro Tyr Gln Gln Asp Leu Gly Ile Gly Glu 35 40 45 Ser Arg Ile Ser His Glu Asn Gly Thr Ile Leu Cys Ser Lys Gly Ser 50 55 60 Thr Cys Tyr Gly Leu Trp Glu Lys Ser Lys Gly Asp Ile Asn Leu Val 65 70 75 80 Lys Gln Gly Cys Trp Ser His Ile Gly Asp Pro Gln Glu Cys His Tyr 85 90 95 Glu Glu Cys Val Val Thr Thr Thr Pro Pro Ser Ile Gln Asn Gly Thr 100 105 110 Tyr Arg Phe Cys Cys Cys Ser Thr Asp Leu Cys Asn Val Asn Phe Thr 115 120 125 Glu Asn Phe Pro Pro Pro Asp Thr Thr Pro Leu Ser Pro Pro His Ser 130 135 140 Phe Asn Arg Asp Glu Thr Ile Ile Ile Ala Leu Ala Ser Val Ser Val 145 150 155 160 Leu Ala Val Leu Ile Val Ala Leu Cys Phe Gly Tyr Arg Met Leu Thr 165 170 175 Gly Asp Arg Lys Gln Gly Leu His Ser Met Asn Met Met Glu Ala Ala 180 185 190 Ala Ser Glu Pro Ser Leu Asp Leu Asp Asn Leu Lys Leu Leu Glu Leu 195 200 205 Ile Gly Arg Gly Arg Tyr Gly Ala Val Tyr Lys Gly Ser Leu Asp Glu 210 215 220 Arg Pro Val Ala Val Lys Val Phe Ser Phe Ala Asn Arg Gln Asn Phe 225 230 235 240 Ile Asn Glu Lys Asn Ile Tyr Arg Val Pro Leu Met Glu His Asp Asn 245 250 255 Ile Ala Arg Phe Ile Val Gly Asp Glu Arg Val Thr Ala Asp Gly Arg 260 265 270 Met Glu Tyr Leu Leu Val Met Glu Tyr Tyr Pro Asn Gly Ser Leu Cys 275 280 285 Lys Tyr Leu Ser Leu His Thr Ser Asp Trp Val Ser Ser Cys Arg Leu 290 295 300 Ala His Ser Val Thr Arg Gly Leu Ala Tyr Leu His Thr Glu Leu Pro 305 310 315 320 Arg Gly Asp His Tyr Lys Pro Ala Ile Ser His Arg Asp Leu Asn Ser 325 330 335 Arg Asn Val Leu Val Lys Asn Asp Gly Thr Cys Val Ile Ser Asp Phe 340 345 350 Gly Leu Ser Met Arg Leu Thr Gly Asn Arg Leu Val Arg Pro Gly Glu 355 360 365 Glu Asp Asn Ala Ala Ile Ser Glu Val Gly Thr Ile Arg Tyr Met Ala 370 375 380 Pro Glu Val Leu Glu Gly Ala Val Asn Leu Arg Asp Cys Glu Ser Ala 385 390 395 400 Leu Lys Gln Val Asp Met Tyr Ala Leu Gly Leu Ile Tyr Trp Glu Ile 405 410 415 Phe Met Arg Cys Thr Asp Leu Phe Pro Gly Glu Ser Val Pro Glu Tyr 420 425 430 Gln Met Ala Phe Gln Thr Glu Val Gly Asn His Pro Thr Phe Glu Asp 435 440 445 Met Gln Val Leu Val Ser Arg Glu Lys Gln Arg Pro Lys Phe Pro Glu 450 455 460 Ala Trp Lys Glu Asn Ser Leu Ala Val Arg Ser Leu Lys Glu Thr Ile 465 470 475 480 Glu Asp Cys Trp Asp Gln Asp Ala Glu Ala Arg Leu Thr Ala Gln Cys 485 490 495 Ala Glu Glu Arg Met Ala Glu Leu Met Met Ile Trp Glu Arg Asn Lys 500 505 510 Ser Val Ser Pro Thr Val Asn Pro Met Ser Thr Ala Met Gln Asn Glu 515 520 525 Arg Asn Leu Ser His Asn Arg Arg Val Pro Lys Ile Gly Pro Tyr Pro 530 535 540 Asp Tyr Ser Ser Ser Ser Tyr Ile Glu Asp Ser Ile His His Thr Asp 545 550 555 560 Ser Ile Val Lys Asn Ile Ser Ser Glu His Ser Met Ser Ser Thr Pro 565 570 575 Leu Thr Ile Gly Glu Lys Asn Arg Asn Ser Ile Asn Tyr Glu Arg Gln 580 585 590 Gln Ala Gln Ala Arg Ile Pro Ser Pro Glu Thr Ser Val Thr Ser Leu 595 600 605 Ser Thr Asn Thr Thr Thr Thr Asn Thr Thr Gly Leu Thr Pro Ser Thr 610 615 620 Gly Met Thr Thr Ile Ser Glu Met Pro Tyr Pro Asp Glu Thr Asn Leu 625 630 635 640 His Thr Thr Asn Val Ala Gln Ser Ile Gly Pro Thr Pro Val Cys Leu 645 650 655 Gln Leu Thr Glu Glu Asp Leu Glu Thr Asn Lys Leu Asp Pro Lys Glu 660 665 670 Val Asp Lys Asn Leu Lys Glu Ser Ser Asp Glu Asn Leu Met Glu His 675 680 685 Ser Leu Lys Gln Phe Ser Gly Pro Asp Pro Leu Ser Ser Thr Ser Ser 690 695 700 Ser Leu Leu Tyr Pro Leu Ile Lys Leu Ala Val Glu Ala Thr Gly Gln 705 710 715 720 Gln Asp Phe Thr Gln Thr Ala Asn Gly Gln Ala Cys Leu Ile Pro Asp 725 730 735 Val Leu Pro Thr Gln Ile Tyr Pro Leu Pro Lys Gln Gln Asn Leu Pro 740 745 750 Lys Arg Pro Thr Ser Leu Pro Leu Asn Thr Lys Asn Ser Thr Lys Glu 755 760 765 Pro Arg Leu Lys Phe Gly Ser Lys His Lys Ser Asn Leu Lys Gln Val 770 775 780 Glu Thr Gly Val Ala Lys Met Asn Thr Ile Asn Ala Ala Glu Pro His 785 790 795 800 Val Val Thr Val Thr Met Asn Gly Val Ala Gly Arg Asn His Ser Val 805 810 815 Asn Ser His Ala Ala Thr Thr Gln Tyr Ala Asn Arg Thr Val Leu Ser 820 825 830 Gly Gln Thr Thr Asn Ile Val Thr His Arg Ala Gln Glu Met Leu Gln 835 840 845 Asn Gln Phe Ile Gly Glu Asp Thr Arg Leu Asn Ile Asn Ser Ser Pro 850 855 860 Asp Glu His Glu Pro Leu Leu Arg Arg Glu Gln Gln Ala Gly His Asp 865 870 875 880 Glu Gly Val Leu Asp Arg Leu Val Asp Arg Arg Glu Arg Pro Leu Glu 885 890 895 Gly Gly Arg Thr Asn Ser Asn Asn Asn Asn Ser Asn Pro Cys Ser Glu 900 905 910 Gln Asp Val Leu Ala Gln Gly Val Pro Ser Thr Ala Ala Asp Pro Gly 915 920 925 Pro Ser Lys Pro Arg Arg Ala Gln Arg Pro Asn Ser Leu Asp Leu Ser 930 935 940 Ala Thr Asn Val Leu Asp Gly Ser Ser Ile Gln Ile Gly Glu Ser Thr 945 950 955 960 Gln Asp Gly Lys Ser Gly Ser Gly Glu Lys Ile Lys Lys Arg Val Lys 965 970 975 Thr Pro Tyr Ser Leu Lys Arg Trp Arg Pro Ser Thr Trp Val Ile Ser 980 985 990 Thr Glu Ser Leu Asp Cys Glu Val Asn Asn Asn Gly Ser Asn Arg Ala 995 1000 1005 Val His Ser Lys Ser Ser Thr Ala Val Tyr Leu Ala Glu Gly Gly Thr 1010 1015 1020 Ala Thr Thr Met Val Ser Lys Asp Ile Gly Met Asn Cys Leu 1025 1030 1035 115 1038 PRT Homo sapiens 115 Met Thr Ser Ser Leu Gln Arg Pro Trp Arg Val Pro Trp Leu Pro Trp 1 5 10 15 Thr Ile Leu Leu Val Ser Thr Ala Ala Ala Ser Gln Asn Gln Glu Arg 20 25 30 Leu Cys Ala Phe Lys Asp Pro Tyr Gln Gln Asp Leu Gly Ile Gly Glu 35 40 45 Ser Arg Ile Ser His Glu Asn Gly Thr Ile Leu Cys Ser Lys Gly Ser 50 55 60 Thr Cys Tyr Gly Leu Trp Glu Lys Ser Lys Gly Asp Ile Asn Leu Val 65 70 75 80 Lys Gln Gly Cys Trp Ser His Ile Gly Asp Pro Gln Glu Cys His Tyr 85 90 95 Glu Glu Cys Val Val Thr Thr Thr Pro Pro Ser Ile Gln Asn Gly Thr 100 105 110 Tyr Arg Phe Cys Cys Cys Ser Thr Asp Leu Cys Asn Val Asn Phe Thr 115 120 125 Glu Asn Phe Pro Pro Pro Asp Thr Thr Pro Leu Ser Pro Pro His Ser 130 135 140 Phe Asn Arg Asp Glu Thr Ile Ile Ile Ala Leu Ala Ser Val Ser Val 145 150 155 160 Leu Ala Val Leu Ile Val Ala Leu Cys Phe Gly Tyr Arg Met Leu Thr 165 170 175 Gly Asp Arg Lys Gln Gly Leu His Ser Met Asn Met Met Glu Ala Ala 180 185 190 Ala Ser Glu Pro Ser Leu Asp Leu Asp Asn Leu Lys Leu Leu Glu Leu 195 200 205 Ile Gly Arg Gly Arg Tyr Gly Ala Val Tyr Lys Gly Ser Leu Asp Glu 210 215 220 Arg Pro Val Ala Val Lys Val Phe Ser Phe Ala Asn Arg Gln Asn Phe 225 230 235 240 Ile Asn Glu Lys Asn Ile Tyr Arg Val Pro Leu Met Glu His Asp Asn 245 250 255 Ile Ala Arg Phe Ile Val Gly Asp Glu Arg Val Thr Ala Asp Gly Arg 260 265 270 Met Glu Tyr Leu Leu Val Met Glu Tyr Tyr Pro Asn Gly Ser Leu Cys 275 280 285 Lys Tyr Leu Ser Leu His Thr Ser Asp Trp Val Ser Ser Cys Arg Leu 290 295 300 Ala His Ser Val Thr Arg Gly Leu Ala Tyr Leu His Thr Glu Leu Pro 305 310 315 320 Arg Gly Asp His Tyr Lys Pro Ala Ile Ser His Arg Asp Leu Asn Ser 325 330 335 Arg Asn Val Leu Val Lys Asn Asp Gly Thr Cys Val Ile Ser Asp Phe 340 345 350 Gly Leu Ser Met Arg Leu Thr Gly Asn Arg Leu Val Arg Pro Gly Glu 355 360 365 Glu Asp Asn Ala Ala Ile Ser Glu Val Gly Thr Ile Arg Tyr Met Ala 370 375 380 Pro Glu Val Leu Glu Gly Ala Val Asn Leu Arg Asp Cys Glu Ser Ala 385 390 395 400 Leu Lys Gln Val Asp Met Tyr Ala Leu Gly Leu Ile Tyr Trp Glu Ile 405 410 415 Phe Met Arg Cys Thr Asp Leu Phe Pro Gly Glu Ser Val Pro Glu Tyr 420 425 430 Gln Met Ala Phe Gln Thr Glu Val Gly Asn His Pro Thr Phe Glu Asp 435 440 445 Met Gln Val Leu Val Ser Arg Glu Lys Gln Arg Pro Lys Phe Pro Glu 450 455 460 Ala Trp Lys Glu Asn Ser Leu Ala Val Arg Ser Leu Lys Glu Thr Ile 465 470 475 480 Glu Asp Cys Trp Asp Gln Asp Ala Glu Ala Arg Leu Thr Ala Gln Cys 485 490 495 Ala Glu Glu Arg Met Ala Glu Leu Met Met Ile Trp Glu Arg Asn Lys 500 505 510 Ser Val Ser Pro Thr Val Asn Pro Met Ser Thr Ala Met Gln Asn Glu 515 520 525 Arg Asn Leu Ser His Asn Arg Arg Val Pro Lys Ile Gly Pro Tyr Pro 530 535 540 Asp Tyr Ser Ser Ser Ser Tyr Ile Glu Asp Ser Ile His His Thr Asp 545 550 555 560 Ser Ile Val Lys Asn Ile Ser Ser Glu His Ser Met Ser Ser Thr Pro 565 570 575 Leu Thr Ile Gly Glu Lys Asn Arg Asn Ser Ile Asn Tyr Glu Arg Gln 580 585 590 Gln Ala Gln Ala Arg Ile Pro Ser Pro Glu Thr Ser Val Thr Ser Leu 595 600 605 Ser Thr Asn Thr Thr Thr Thr Asn Thr Thr Gly Leu Thr Pro Ser Thr 610 615 620 Gly Met Thr Thr Ile Ser Glu Met Pro Tyr Pro Asp Glu Thr Asn Leu 625 630 635 640 His Thr Thr Asn Val Ala Gln Ser Ile Gly Pro Thr Pro Val Cys Leu 645 650 655 Gln Leu Thr Glu Glu Asp Leu Glu Thr Asn Lys Leu Asp Pro Lys Glu 660 665 670 Val Asp Lys Asn Leu Lys Glu Ser Ser Asp Glu Asn Leu Met Glu His 675 680 685 Ser Leu Lys Gln Phe Ser Gly Pro Asp Pro Leu Ser Ser Thr Ser Ser 690 695 700 Ser Leu Leu Tyr Pro Leu Ile Lys Leu Ala Val Glu Ala Thr Gly Gln 705 710 715 720 Gln Asp Phe Thr Gln Thr Ala Asn Gly Gln Ala Cys Leu Ile Pro Asp 725 730 735 Val Leu Pro Thr Gln Ile Tyr Pro Leu Pro Lys Gln Gln Asn Leu Pro 740 745 750 Lys Arg Pro Thr Ser Leu Pro Leu Asn Thr Lys Asn Ser Thr Lys Glu 755 760 765 Pro Arg Leu Lys Phe Gly Ser Lys His Lys Ser Asn Leu Lys Gln Val 770 775 780 Glu Thr Gly Val Ala Lys Met Asn Thr Ile Asn Ala Ala Glu Pro His 785 790 795 800 Val Val Thr Val Thr Met Asn Gly Val Ala Gly Arg Asn His Ser Val 805 810 815 Asn Ser His Ala Ala Thr Thr Gln Tyr Ala Asn Arg Thr Val Leu Ser 820 825 830 Gly Gln Thr Thr Asn Ile Val Thr His Arg Ala Gln Glu Met Leu Gln 835 840 845 Asn Gln Phe Ile Gly Glu Asp Thr Arg Leu Asn Ile Asn Ser Ser Pro 850 855 860 Asp Glu His Glu Pro Leu Leu Arg Arg Glu Gln Gln Ala Gly His Asp 865 870 875 880 Glu Gly Val Leu Asp Arg Leu Val Asp Arg Arg Glu Arg Pro Leu Glu 885 890 895 Gly Gly Arg Thr Asn Ser Asn Asn Asn Asn Ser Asn Pro Cys Ser Glu 900 905 910 Gln Asp Val Leu Ala Gln Gly Val Pro Ser Thr Ala Ala Asp Pro Gly 915 920 925 Pro Ser Lys Pro Arg Arg Ala Gln Arg Pro Asn Ser Leu Asp Leu Ser 930 935 940 Ala Thr Asn Val Leu Asp Gly Ser Ser Ile Gln Ile Gly Glu Ser Thr 945 950 955 960 Gln Asp Gly Lys Ser Gly Ser Gly Glu Lys Ile Lys Lys Arg Val Lys 965 970 975 Thr Pro Tyr Ser Leu Lys Arg Trp Arg Pro Ser Thr Trp Val Ile Ser 980 985 990 Thr Glu Ser Leu Asp Cys Glu Val Asn Asn Asn Gly Ser Asn Arg Ala 995 1000 1005 Val His Ser Lys Ser Ser Thr Ala Val Tyr Leu Ala Glu Gly Gly Thr 1010 1015 1020 Ala Thr Thr Met Val Ser Lys Asp Ile Gly Met Asn Cys Leu 1025 1030 1035 116 2932 DNA Homo sapiens 116 gctccgcgcc gagggctgga ggatgcgttc cctggggtcc ggacttatga aaatatgcat 60 cagtttaata ctgtcttgga attcatgaga tggaagcata ggtcaaagct gtttggagaa 120 aatcagaagt acagttttat ctagccacat cttggaggag tcgtaagaaa gcagtgggag 180 ttgaagtcat tgtcaagtgc ttgcgatctt ttacaagaaa atctcactga atgatagtca 240 tttaaattgg tgaagtagca agaccaatta ttaaaggtga cagtacacag gaaacattac 300 aattgaacaa tgactcagct atacatttac atcagattat tgggagccta tttgttcatc 360 atttctcgtg ttcaaggaca gaatctggat agtatgcttc atggcactgg gatgaaatca 420 gactccgacc agaaaaagtc agaaaatgga gtaaccttag caccagagga taccttgcct 480 tttttaaagt gctattgctc agggcactgt ccagatgatg ctattaataa cacatgcata 540 actaatggac attgctttgc catcatagaa gaagatgacc agggagaaac cacattagct 600 tcagggtgta tgaaatatga aggatctgat tttcagtgca aagattctcc aaaagcccag 660 ctacgccgga caatagaatg ttgtcggacc aatttatgta accagtattt gcaacccaca 720 ctgccccctg ttgtcatagg tccgtttttt gatggcagca ttcgatggct ggttttgctc 780 atttctatgg ctgtctgcat aattgctatg atcatcttct ccagctgctt ttgttacaaa 840 cattattgca agagcatctc aagcagacgt cgttacaatc gtgatttgga acaggatgaa 900 gcatttattc cagttggaga atcactaaaa gaccttattg accagtcaca aagttctggt 960 agtgggtctg gactaccttt attggttcag cgaactattg ccaaacagat tcagatggtc 1020 cggcaagttg gtaaaggccg atatggagaa gtatggatgg gcaaatggcg tggcgaaaaa 1080 gtggcggtga aagtattctt taccactgaa gaagccagct ggtttcgaga aacagaaatc 1140 taccaaactg tgctaatgcg ccatgaaaac atacttggtt tcatagcggc agacattaaa 1200 ggtacaggtt cctggactca gctctatttg attactgatt accatgaaaa tggatctctc 1260 tatgacttcc tgaaatgtgc tacactggac accagagccc tgcttaaatt ggcttattca 1320 gctgcctgtg gtctgtgcca cctgcacaca gaaatttatg gcacccaagg aaagcccgca 1380 attgctcatc gagacctaaa gagcaaaaac atcctcatca agaaaaatgg gagttgctgc 1440 attgctgacc tgggccttgc tgttaaattc aacagtgaca caaatgaagt tgatgtgccc 1500 ttgaatacca gggtgggcac caaacgctac atggctcccg aagtgctgga cgaaagcctg 1560 aacaaaaacc acttccagcc ctacatcatg gctgacatct acagcttcgg cctaatcatt 1620 tgggagatgg ctcgtcgttg tatcacagga gggatcgtgg aagaatacca attgccatat 1680 tacaacatgg taccgagtga tccgtcatac gaagatatgc gtgaggttgt gtgtgtcaaa 1740 cgtttgcggc caattgtgtc taatcggtgg aacagtgatg aatgtctacg agcagttttg 1800 aagctaatgt cagaatgctg ggcccacaat ccagcctcca gactcacagc attgagaatt 1860 aagaagacgc ttgccaagat ggttgaatcc caagatgtaa aaatctgatg gttaaaccat 1920 cggaggagaa actctagact gcaagaactg tttttaccca tggcatgggt ggaattagag 1980 tggaataagg atgttaactt ggttctcaga ctctttcttc actacgtgtt cacaggctgc 2040 taatattaaa cctttcagta ctcttattag gatacaagct gggaacttct aaacacttca 2100 ttctttatat atggacagct ttattttaaa tgtggttttt gatgcctttt tttaagtggg 2160 tttttatgaa ctgcatcaag acttcaatcc tgattagtgt ctccagtcaa gctctgggta 2220 ctgaattgcc tgttcataaa acggtgcttt ctgtgaaagc cttaagaaga taaatgagcg 2280 cagcagagat ggagaaatag actttgcctt ttacctgaga cattcagttc gtttgtattc 2340 tacctttgta aaacagccta tagatgatga tgtgtttggg atactgctta ttttatgata 2400 gtttgtcctg tgtccttagt gatgtgtgtg tgtctccatg cacatgcacg ccgggattcc 2460 tctgctgcca tttgaattag aagaaaataa tttatatgca tgcacaggaa gatattggtg 2520 gccggtggtt ttgtgcttta aaaatgcaat atctgaccaa gattcgccaa tctcatacaa 2580 gccatttact ttgcaagtga gatagcttcc ccaccagctt tattttttaa catgaaagct 2640 gatgccaagg ccaaaagaag tttaaagcat ctgtaaattt ggactgtttt ccttcaacca 2700 ccattttttt tgtggttatt atttttgtca cggaaagcat cctctccaaa gttggagctt 2760 ctattgccat gaaccatgct tacaaagaaa gcacttctta ttgaagtgaa ttcctgcatt 2820 tgatagcaat gtaagtgcct ataaccatgt tctatattct ttattctcag taacttttaa 2880 aagggaagtt atttatattt tgtgtataat gtgctttatt tgcaaatcac cc 2932 117 1575 DNA Homo sapiens 117 gcaaacttcc ttgataacat gcttttgcga agtgcaggaa aattaaatgt gggcaccaag 60 aaagaggatg gtgagagtac agcccccacc ccccgtccaa aggtcttgcg ttgtaaatgc 120 caccaccatt gtccagaaga ctcagtcaac aatatttgca gcacagacgg atattgtttc 180 acgatgatag aagaggatga ctctgggttg cctgtggtca cttctggttg cctaggacta 240 gaaggctcag attttcagtg tcgggacact cccattcctc atcaaagaag atcaattgaa 300 tgctgcacag aaaggaacga atgtaataaa gacctacacc ctacactgcc tccattgaaa 360 aacagagatt ttgttgatgg acctatacac cacagggctt tacttatatc tgtgactgtc 420 tgtagtttgc tcttggtcct tatcatatta ttttgttact tccggtataa aagacaagaa 480 accagacctc gatacagcat tgggttagaa caggatgaaa cttacattcc tcctggagaa 540 tccctgagag acttaattga gcagtctcag agctcaggaa gtggatcagg cctccctctg 600 ctggtccaaa ggactatagc taagcagatt cagatggtga aacagattgg aaaaggtcgc 660 tatggggaag tttggatggg aaagtggcgt ggcgaaaagg tagctgtgaa agtgttcttc 720 accacagagg aagccagctg gttcagagag acagaaatat atcagacagt gttgatgagg 780 catgaaaaca ttttgggttt cattgctgca gatatcaaag ggacagggtc ctggacccag 840 ttgtacctaa tcacagacta tcatgaaaat ggttcccttt atgattatct gaagtccacc 900 accctagacg ctaaatcaat gctgaagtta gcctactctt ctgtcagtgg cttatgtcat 960 ttacacacag aaatctttag tactcaaggc aaaccagcaa ttgcccatcg agatctgaaa 1020 agtaaaaaca ttctggtgaa gaaaaatgga acttgctgta ttgctgacct gggcctggct 1080 gttaaattta ttagtgatac aaatgaagtt gacataccac ctaacactcg agttggcacc 1140 aaacgctata tgcctccaga agtgttggac gagagcttga acagaaatca cttccagtct 1200 tacatcatgg ctgacatgta tagttttggc ctcatccttt gggaggttgc taggagatgt 1260 gtatcaggag gtatagtgga agaataccag cttccttatc atgacctagt gcccagtgac 1320 ccctcttatg aggacatgag ggagattgtg tgcatcaaga agttacgccc ctcattccca 1380 aaccggtgga gcagtgatga gtgtctaagg cagatgggaa aactcatgac agaatgctgg 1440 gctcacaatc ctgcatcaag gctgacagcc ctgcgggtta agaaaacact tgccaaaatg 1500 tcagagtccc aggacattaa actctgatag gagaggaaaa gtaagcatct ctgcagaaag 1560 ccaacaggta ccctt 1575 118 2032 DNA Homo sapiens 118 cgcggggcgc ggagtcggcg gggcctcgcg ggacgcgggc agtgcggaga ccgcggcgct 60 gaggacgcgg gagccgggag cgcacgcgcg gggtggagtt cagcctactc tttcttagat 120 gtgaaaggaa aggaagatca tttcatgcct tgttgataaa ggttcagact tctgctgatt 180 cataaccatt tggctctgag ctatgacaag agaggaaaca aaaagttaaa cttacaagcc 240 tgccataagt gagaagcaaa cttccttgat aacatgcttt tgcgaagtgc aggaaaatta 300 aatgtgggca ccaagaaaga ggatggtgag agtacagccc ccaccccccg tccaaaggtc 360 ttgcgttgta aatgccacca ccattgtcca gaagactcag tcaacaatat ttgcagcaca 420 gacggatatt gtttcacgat gatagaagag gatgactctg ggttgcctgt ggtcacttct 480 ggttgcctag gactagaagg ctcagatttt cagtgtcggg acactcccat tcctcatcaa 540 agaagatcaa ttgaatgctg cacagaaagg aacgaatgta ataaagacct acaccctaca 600 ctgcctccat tgaaaaacag agattttgtt gatggaccta tacaccacag ggctttactt 660 atatctgtga ctgtctgtag tttgctcttg gtccttatca tattattttg ttacttccgg 720 tataaaagac aagaaaccag acctcgatac agcattgggt tagaacagga tgaaacttac 780 attcctcctg gagaatccct gagagactta attgagcagt ctcagagctc aggaagtgga 840 tcaggcctcc ctctgctggt ccaaaggact atagctaagc agattcagat ggtgaaacag 900 attggaaaag gtcgctatgg ggaagtttgg atgggaaagt ggcgtggcga aaaggtagct 960 gtgaaagtgt tcttcaccac agaggaagcc agctggttca gagagacaga aatatatcag 1020 acagtgttga tgaggcatga aaacattttg ggtttcattg ctgcagatat caaagggaca 1080 gggtcctgga cccagttgta cctaatcaca gactatcatg aaaatggttc cctttatgat 1140 tatctgaagt ccaccaccct agacgctaaa tcaatgctga agttagccta ctcttctgtc 1200 agtggcttat gtcatttaca cacagaaatc tttagtactc aaggcaaacc agcaattgcc 1260 catcgagatc tgaaaagtaa aaacattctg gtgaagaaaa atggaacttg ctgtattgct 1320 gacctgggcc tggctgttaa atttattagt gatacaaatg aagttgacat accacctaac 1380 actcgagttg gcaccaaacg ctatatgcct ccagaagtgt tggacgagag cttgaacaga 1440 aatcacttcc agtcttacat catggctgac atgtatagtt ttggcctcat cctttgggag 1500 gttgctagga gatgtgtatc aggaggtata gtggaagaat accagcttcc ttatcatgac 1560 ctagtgccca gtgacccctc ttatgaggac atgagggaga ttgtgtgcat caagaagtta 1620 cgcccctcat tcccaaaccg gtggagcagt gatgagtgtc taaggcagat gggaaaactc 1680 atgacagaat gctgggctca caatcctgca tcaaggctga cagccctgcg ggttaagaaa 1740 acacttgcca aaatgtcaga gtcccaggac attaaactct gataggagag gaaaagtaag 1800 catctctgca gaaagccaac aggtactctt ctgtttgtgg gcagagcaaa agacatcaaa 1860 taagcatcca cagtacaagc cttgaacatc gtcctgcttc ccagtgggtt cagacctcac 1920 ctttcaggga gcgacctggg caaagacaga gaagctccca gaaggagaga ttgatccgtg 1980 tctgtttgta ggcggagaaa ccgttgggta acttgttcaa gatatgatgc at 2032 119 3167 DNA Rattus sp. 119 gaattcatga gatggaaaca taggtcaaag ctgtttggag aaattggaac tacagtttta 60 tctagccaca tctctgagaa gtctgaagaa agcagcaggt gaaagtcatt gtcaagtgat 120 tttgttcttc tgtaaggaaa cctcgttcag taaggccgtt tacttcagtg aaacagcagg 180 accagtaatc aaggtggccc ggacaggaca cgtgcgaatt ggacaatgac tcagctatac 240 acttacatca gattactggg agcctgtctg ttcatcattt ctcatgttca agggcagaat 300 ctagatagta tgctccatgg tactggtatg aaatcagacg tggaccagaa gaagccggaa 360 aatggagtga cgttagcacc agaggacacc ttacctttct taaaatgcta ttgctcagga 420 cactgcccag atgacgctat taataacaca tgcataacta atggccattg ctttgccatt 480 atagaagaag atgatcaggg agaaaccacg ttaacttctg ggtgtatgaa gtatgaaggc 540 tctgattttc aatgcaagga ttcaccaaaa gcccagctac gcaggacaat agaatgttgt 600 cggaccaatt tgtgcaacca atatttgcag cctacactgc cccctgtcgt tataggccca 660 ttctttgatg gcagcgtccg atggctggct gtgctcatct ctatggctgt ctgtattgtc 720 gccatgatcg tcttctccag ctgcttctgt tacaaacatt actgtaagag tatctcaagc 780 agaggtcgtt acaaccgtga cttggaacag gatgaagcat ttattccagt aggagaatca 840 ctgaaagacc tgattgacca gtcacaaagc tctggtagtg gatctggatt acctttattg 900 gttcagcgaa ctattgccaa acagattcag atggttcggc aggttggtaa aggccggtat 960 ggagaagtat ggatgggtaa atggcgtggt gaaaaagtgg ctgtcaaagt attttttacc 1020 actgaagaag ctagctggtt tagagaaaca gaaatctacc agacggtgtt aatgcgtcat 1080 gaaaatatac ttggttttat agctgcagac attaaaggca ccggttcctg gactcagctg 1140 tatttgatta ctgattacca tgagaatggg tctctctatg acttcctgaa atgtgccacc 1200 ctggacacca gagccctact caagttagct tattctgctg cctgtggtct gtgccacctc 1260 cacacagaaa tttatggcac gcaaggcaag cctgcaattg ctcatcgaga cctgaagagc 1320 aaaaacatcc ttattaagaa aaatggtagt tgctgtattg ctgacctggg cctagctgtt 1380 aaattcaaca gtgacacaaa tgaagttgac atacccttga acaccagggt gggcaccagg 1440 cggtacatgg ctccagaagt gctggacgag agcctgagta aaaaccattt ccagccctac 1500 atcatggctg acatctacag ctttggtttg atcatttggg agatggcccg tcgctgtatt 1560 acaggaggaa tcgtggagga atatcaatta ccatattaca acatggtgcc tagtgaccca 1620 tcttatgaag acatgcgtga ggtcgtgtgt gtgaaacgct tgcggccaat cgtctctaac 1680 cgctggaaca gtgatgaatg tcttcgagcc gttttgaagc tgatgtcaga atgctgggcc 1740 cataatccag catccagact cacagctttg agaatcaaga agacgctcgc aaagatggtt 1800 gaatcccagg atgtaaagat ttgacaaaca gttttgagaa agaatttaga ctgcaagaaa 1860 ttcacccgag gaagggtgga gttagcatgg actaggatgt cggcttggtt tccagactct 1920 ctcctctaca tcttcacagg ctgctaacag taaactttca ggactctgca gaatgcaggg 1980 ttggagcttc agacatagga cttcagacat gctgttcttt gcgtatggac agctttgttt 2040 taaatgtggg cttttgatgc ctttttggtt tttatgaatt gcatcaagac tccaatcctg 2100 ataagaagtc tctggtcaaa ctctggttac tcactatcct gtccataaag tggtgctttc 2160 tgtgaaagcc ttaaggaaat tagtgagctc agcagagatg gagaaaggca tatttgccct 2220 ctacagagaa aatatctgtc tgtgttctgt ctctgtaaac agcctggact atgatctctt 2280 tgggatgctg cctggttgat gatggtgcat catgcctctg atatgcatac cagacttcct 2340 ctgctgccat gggcttacaa gacaagaatg tgaaggttgc acaggacggt atttgtggcc 2400 agtggtttaa atatgcaata tctaatcgac attcgccaat ctcataaaag ccatctacct 2460 tgtaactgaa gtaacttctc taccaacttt atttttagca taatagttgt aaaggccaaa 2520 ctatgtataa agtgtccata gactcgaact gttttcctcc agtcaccatt ttgttttcct 2580 tttggtaatt atttttgtta tataattcct cctatccaga attggcgctc actgtcttga 2640 accatacttt gaaagaaatg cctcttcctg gagtctgcct tactgcatct gatcaccatg 2700 tgcatacctc tgatcaaatt ctggagtctt tgttctcggt acctcttaaa aagggaaatt 2760 gtgtatcatg tgtagtgtgc ttttattttc aaaatcttca tagcctttat tctagccatt 2820 tttacctaca tactcattct gtacaaaaca gctcactcgg tctcacggct gatcctcagt 2880 ggaaatgatt taaagtagag ctgtgtacga atttcagaat tcatgtattt aaaaacttca 2940 cactaacact ttactaagat attgtctcat atcttttatg aggatgtcag ctgattttca 3000 atgactataa atgtatctta gctatctaaa tcttttgaaa tttggtttta taatttctgg 3060 tccctaactt gtgaagacaa agaggcagaa gtacccagtc taccacattt acactgtaca 3120 ttattaaata aaaaaatgta tattttaaaa aaaaaaaaaa aaaaaaa 3167 120 3167 DNA Rattus norvegicus 120 gaattcatga gatggaaaca taggtcaaag ctgtttggag aaattggaac tacagtttta 60 tctagccaca tctctgagaa gtctgaagaa agcagcaggt gaaagtcatt gtcaagtgat 120 tttgttcttc tgtaaggaaa cctcgttcag taaggccgtt tacttcagtg aaacagcagg 180 accagtaatc aaggtggccc ggacaggaca cgtgcgaatt ggacaatgac tcagctatac 240 acttacatca gattactggg agcctgtctg ttcatcattt ctcatgttca agggcagaat 300 ctagatagta tgctccatgg tactggtatg aaatcagacg tggaccagaa gaagccggaa 360 aatggagtga cgttagcacc agaggacacc ttacctttct taaaatgcta ttgctcagga 420 cactgcccag atgacgctat taataacaca tgcataacta atggccattg ctttgccatt 480 atagaagaag atgatcaggg agaaaccacg ttaacttctg ggtgtatgaa gtatgaaggc 540 tctgattttc aatgcaagga ttcaccaaaa gcccagctac gcaggacaat agaatgttgt 600 cggaccaatt tgtgcaacca atatttgcag cctacactgc cccctgtcgt tataggccca 660 ttctttgatg gcagcgtccg atggctggct gtgctcatct ctatggctgt ctgtattgtc 720 gccatgatcg tcttctccag ctgcttctgt tacaaacatt actgtaagag tatctcaagc 780 agaggtcgtt acaaccgtga cttggaacag gatgaagcat ttattccagt aggagaatca 840 ctgaaagacc tgattgacca gtcacaaagc tctggtagtg gatctggatt acctttattg 900 gttcagcgaa ctattgccaa acagattcag atggttcggc aggttggtaa aggccggtat 960 ggagaagtat ggatgggtaa atggcgtggt gaaaaagtgg ctgtcaaagt attttttacc 1020 actgaagaag ctagctggtt tagagaaaca gaaatctacc agacggtgtt aatgcgtcat 1080 gaaaatatac ttggttttat agctgcagac attaaaggca ccggttcctg gactcagctg 1140 tatttgatta ctgattacca tgagaatggg tctctctatg acttcctgaa atgtgccacc 1200 ctggacacca gagccctact caagttagct tattctgctg cctgtggtct gtgccacctc 1260 cacacagaaa tttatggcac gcaaggcaag cctgcaattg ctcatcgaga cctgaagagc 1320 aaaaacatcc ttattaagaa aaatggtagt tgctgtattg ctgacctggg cctagctgtt 1380 aaattcaaca gtgacacaaa tgaagttgac atacccttga acaccagggt gggcaccagg 1440 cggtacatgg ctccagaagt gctggacgag agcctgagta aaaaccattt ccagccctac 1500 atcatggctg acatctacag ctttggtttg atcatttggg agatggcccg tcgctgtatt 1560 acaggaggaa tcgtggagga atatcaatta ccatattaca acatggtgcc tagtgaccca 1620 tcttatgaag acatgcgtga ggtcgtgtgt gtgaaacgct tgcggccaat cgtctctaac 1680 cgctggaaca gtgatgaatg tcttcgagcc gttttgaagc tgatgtcaga atgctgggcc 1740 cataatccag catccagact cacagctttg agaatcaaga agacgctcgc aaagatggtt 1800 gaatcccagg atgtaaagat ttgacaaaca gttttgagaa agaatttaga ctgcaagaaa 1860 ttcacccgag gaagggtgga gttagcatgg actaggatgt cggcttggtt tccagactct 1920 ctcctctaca tcttcacagg ctgctaacag taaactttca ggactctgca gaatgcaggg 1980 ttggagcttc agacatagga cttcagacat gctgttcttt gcgtatggac agctttgttt 2040 taaatgtggg cttttgatgc ctttttggtt tttatgaatt gcatcaagac tccaatcctg 2100 ataagaagtc tctggtcaaa ctctggttac tcactatcct gtccataaag tggtgctttc 2160 tgtgaaagcc ttaaggaaat tagtgagctc agcagagatg gagaaaggca tatttgccct 2220 ctacagagaa aatatctgtc tgtgttctgt ctctgtaaac agcctggact atgatctctt 2280 tgggatgctg cctggttgat gatggtgcat catgcctctg atatgcatac cagacttcct 2340 ctgctgccat gggcttacaa gacaagaatg tgaaggttgc acaggacggt atttgtggcc 2400 agtggtttaa atatgcaata tctaatcgac attcgccaat ctcataaaag ccatctacct 2460 tgtaactgaa gtaacttctc taccaacttt atttttagca taatagttgt aaaggccaaa 2520 ctatgtataa agtgtccata gactcgaact gttttcctcc agtcaccatt ttgttttcct 2580 tttggtaatt atttttgtta tataattcct cctatccaga attggcgctc actgtcttga 2640 accatacttt gaaagaaatg cctcttcctg gagtctgcct tactgcatct gatcaccatg 2700 tgcatacctc tgatcaaatt ctggagtctt tgttctcggt acctcttaaa aagggaaatt 2760 gtgtatcatg tgtagtgtgc ttttattttc aaaatcttca tagcctttat tctagccatt 2820 tttacctaca tactcattct gtacaaaaca gctcactcgg tctcacggct gatcctcagt 2880 ggaaatgatt taaagtagag ctgtgtacga atttcagaat tcatgtattt aaaaacttca 2940 cactaacact ttactaagat attgtctcat atcttttatg aggatgtcag ctgattttca 3000 atgactataa atgtatctta gctatctaaa tcttttgaaa tttggtttta taatttctgg 3060 tccctaactt gtgaagacaa agaggcagaa gtacccagtc taccacattt acactgtaca 3120 ttattaaata aaaaaatgta tattttaaaa aaaaaaaaaa aaaaaaa 3167 121 3003 DNA Rattus norvegicus 121 cgttcagtaa ggccgtttac ttcagtgaaa cagcaggacc agtaatcaag gtggcccgga 60 caggacacgt gcgaattgga caatgactca gctatacact tacatcagat tactgggagc 120 ctgtctgttc atcatttctc atgttcaagg gcagaatcta gatagtatgc tccatggtac 180 tggtatgaaa tcagacgtgg accagaagaa gccggaaaat ggagtgacgt tagcaccaga 240 ggacacctta cctttcttaa aatgctattg ctcaggacac tgcccagatg acgctattaa 300 taacacatgc ataactaatg gccattgctt tgccattata gaagaagatg atcagggaga 360 aaccacgtta acttctgggt gtatgaagta tgaaggctct gattttcaat gcaaggattc 420 accaaaagcc cagctacgca ggacaataga atgttgtcgg accaatttgt gcaaccaata 480 tttgcagcct acactgcccc ctgtcgttat aggcccattc tttgatggca gcgtccgatg 540 gctggctgtg ctcatctcta tggctgtctg tattgtcgcc atgatcgtct tctccagctg 600 cttctgttac aaacattact gtaagagtat ctcaagcaga ggtcgttaca accgtgactt 660 ggaacaggat gaagcattta ttccagtagg agaatcactg aaagacctga ttgaccagtc 720 acaaagctct ggtagtggat ctggattacc tttattggtt cagcgaacta ttgccaaaca 780 gattcagatg gttcggcagg ttggtaaagg ccggtatgga gaagtatgga tgggtaaatg 840 gcgtggtgaa aaagtggctg tcaaagtatt ttttaccact gaagaagcta gctggtttag 900 agaaacagaa atctaccaga cggtgttaat gcgtcatgaa aatatacttg gttttatagc 960 tgcagacatt aaaggcaccg gttcctggac tcagctgtat ttgattactg attaccatga 1020 gaatgggtct ctctatgact tcctgaaatg tgccaccctg gacaccagag ccctactcaa 1080 gttagcttat tctgctgcct gtggtctgtg ccacctccac acagaaattt atggcacgca 1140 aggcaagcct gcaattgctc atcgagacct gaagagcaaa aacatcctta ttaagaaaaa 1200 tggtagttgc tgtattgctg acctgggcct agctgttaaa ttcaacagtg acacaaatga 1260 agttgacata cccttgaaca ccagggtggg caccaggcgg tacatggctc cagaagtgct 1320 ggacgagagc ctgagtaaaa accatttcca gccctacatc atggctgaca tctacagctt 1380 tggtttgatc atttgggaga tggcccgtcg ctgtattaca ggaggaatcg tggaggaata 1440 tcaattacca tattacaaca tggtgcctag tgacccatct tatgaagaca tgcgtgaggt 1500 cgtgtgtgtg aaacgcttgc ggccaatcgt ctctaaccgc tggaacagtg atgaatgtct 1560 tcgagccgtt ttgaagctga tgtcagaatg ctgggcccat aatccagcat ccagactcac 1620 agctttgaga atcaagaaga cgctcgcaaa gatggttgaa tcccaggatg taaagatttg 1680 acaaacagtt ttgagaaaga atttagactg caagaaattc acccgaggaa gggtggagtt 1740 agcatggact aggatgtcgg cttggtttcc agactctctc ctctacatct tcacaggctg 1800 ctaacagtaa actttcagga ctctgcagaa tgcagggttg gagcttcaga cataggactt 1860 cagacatgct gttctttgcg tatggacagc tttgttttaa atgtgggctt ttgatgcctt 1920 tttggttttt atgaattgca tcaagactcc aatcctgata agaagtctct ggtcaaactc 1980 tggttactca ctatcctgtc cataaagtgg tgctttctgt gaaagcctta aggaaattag 2040 tgagctcagc agagatggag aaaggcatat ttgccctcta cagagaaaat atctgtctgt 2100 gttctgtctc tgtaaacagc ctggactatg atctctttgg gatgctgcct ggttgatgat 2160 ggtgcatcat gcctctgata tgcataccag acttcctctg ctgccatggg cttacaagac 2220 aagaatgtga aggttgcaca ggacggtatt tgtggccagt ggtttaaata tgcaatatct 2280 aatcgacatt cgccaatctc ataaaagcca tctaccttgt aactgaagta acttctctac 2340 caactttatt tttagcataa tagttgtaaa ggccaaacta tgtataaagt gtccatagac 2400 tcgaactgtt ttcctccagt caccattttg ttttcctttt ggtaattatt tttgttatat 2460 aattcctcct atccagaatt ggcgctcact gtcttgaacc atactttgaa agaaatgcct 2520 cttcctggag tctgccttac tgcatctgat caccatgtgc atacctctga tcaaattctg 2580 gagtctttgt tctcggtacc tcttaaaaag ggaaattgtg tatcatgtgt agtgtgcttt 2640 tattttcaaa atcttcatag cctttattct agccattttt acctacatac tcattctgta 2700 caaaacagct cactcggtct cacggctgat cctcagtgga aatgatttaa agtagagctg 2760 tgtacgaatt tcagaattca tgtatttaaa aacttcacac taacacttta ctaagatatt 2820 gtctcatatc ttttatgagg atgtcagctg attttcaatg actataaatg tatcttagct 2880 atctaaatct tttgaaattt ggttttataa tttctggtcc ctaacttgtg aagacaaaga 2940 ggcagaagta cccagtctac cacatttaca ctgtacatta ttaaataaaa aaatgtatat 3000 ttt 3003 122 2063 DNA Homo sapiens 122 gaattccggt gatgatgatg gtgatggtga tgatggtgat gaggatgatg gtgatgatga 60 tgatggtgtt ggtgatggtt tttgcatctt ccattcatga actaagtact cttattagtg 120 aatttctttt ctttgccctc ctgattcttg gctggcccag ggatgacttc ctcgctgcag 180 cggccctggc gggtgccctg gctaccatgg accatcctgc tggtcagcac tgcggctgct 240 tcgcagaatc aagaacggct atgtgcgttt aaagatccgt atcagcaaga ccttgggata 300 ggtgagagta gaatctctca tgaaaatggg acaatattat gctcgaaagg tagcacctgc 360 tatggccttt gggagaaatc aaaaggggac ataaatcttg taaaacaagg atgttggtct 420 cacattggag atccccaaga gtgtcactat gaagaatgtg tagtaactac cactcctccc 480 tcaattcaga atggaacata ccgtttctgc tgttgtagca cagatttatg taatgtcaac 540 tttactgaga attttccacc tcctgacaca acaccactca gtccacctca ttcatttaac 600 cgagatgaga caataatcat tgctttggca tcagtctctg tattagctgt tttgatagtt 660 gccttatgct ttggatacag aatgttgaca ggagaccgta aacaaggtct tcacagtatg 720 aacatgatgg aggcagcagc atccgaaccc tctcttgatc tagataatct gaaactgttg 780 gagctgattg gccgaggtcg atatggagca gtatataaag gctccttgga tgagcgtcca 840 gttgctgtaa aagtgttttc ctttgcaaac cgtcagaatt ttatcaacga aaagaacatt 900 tacagagtgc ctttgatgga acatgacaac attgcccgct ttatagttgg agatgagaga 960 gtcactgcag atggacgcat ggaatatttg cttgtgatgg agtactatcc caatggatct 1020 ttatgcaagt atttaagtct ccacacaagt gactgggtaa gctcttgccg tcttgctcat 1080 tctgttacta gaggactggc ttatcttcac acagaattac cacgaggaga tcattataaa 1140 cctgcaattt cccatcgaga tttaaacagc agaaatgtcc tagtgaaaaa tgatggaacc 1200 tgtgttatta gtgactttgg actgtccatg aggctgactg gaaatagact ggtgcgccca 1260 ggggaggaag ataatgcagc cataagcgag gttggcacta tcagatatat ggcaccagaa 1320 gtgctagaag gagctgtgaa cttgagggac tgtgaatcag ctttgaaaca agtagacatg 1380 tatgctcttg gactaatcta ttgggagata tttatgagat gtacagacct cttcccaggg 1440 gaatccgtac cagagtacca gatggctttt cagacagagg ttggaaacca tcccactttt 1500 gaggatatgc aggttctcgt gtctagggaa aaacagagac ccaagttccc agaagcctgg 1560 aaagaaaata gcctggcagt gaggtcactc aaggagacaa tcgaagactg ttgggaccag 1620 gatgcagagg ctcggcttac tgcacagtgt gctgaggaaa ggatggctga acttatgatg 1680 atttgggaaa gaaacaaatc tgtgagccca acagtcaatc caatgtctac tgctatgcag 1740 aatgaacgta ggtgagtcaa cacaagatgg caaatcagga tcaggtgaaa agatcaagaa 1800 acgtgtgaaa actccctatt ctcttaagcg gtggcgcccc tccacctggg tcatctccac 1860 tgaatcgctg gactgtgaag tcaacaataa tggcagtaac agggcagttc attccaaatc 1920 cagcactgct gtttaccttg cagaaggagg cactgctaca accatggtgt ctaaagatat 1980 aggaatgaac tgtctgtgaa atgttttcaa gcctatggag tgaaattatt ttttgcatca 2040 tttaaacatg cagaagatgt tta 2063 123 1964 DNA Homo sapiens 123 atttcttttc tttgccctcc tgattcttgg ctggcccagg gatgacttcc tcgctgcagc 60 ggccctggcg ggtgccctgg ctaccatgga ccatcctgct ggtcagcact gcggctgctt 120 cgcagaatca agaacggcta tgtgcgttta aagatccgta tcagcaagac cttgggatag 180 gtgagagtag aatctctcat gaaaatggga caatattatg ctcgaaaggt agcacctgct 240 atggcctttg ggagaaatca aaaggggaca taaatcttgt aaaacaagga tgttggtctc 300 acattggaga tccccaagag tgtcactatg aagaatgtgt agtaactacc actcctccct 360 caattcagaa tggaacatac cgtttctgct gttgtagcac agatttatgt aatgtcaact 420 ttactgagaa ttttccacct cctgacacaa caccactcag tccacctcat tcatttaacc 480 gagatgagac aataatcatt gctttggcat cagtctctgt attagctgtt ttgatagttg 540 ccttatgctt tggatacaga atgttgacag gagaccgtaa acaaggtctt cacagtatga 600 acatgatgga ggcagcagca tccgaaccct ctcttgatct agataatctg aaactgttgg 660 agctgattgg ccgaggtcga tatggagcag tatataaagg ctccttggat gagcgtccag 720 ttgctgtaaa agtgttttcc tttgcaaacc gtcagaattt tatcaacgaa aagaacattt 780 acagagtgcc tttgatggaa catgacaaca ttgcccgctt tatagttgga gatgagagag 840 tcactgcaga tggacgcatg gaatatttgc ttgtgatgga gtactatccc aatggatctt 900 tatgcaagta tttaagtctc cacacaagtg actgggtaag ctcttgccgt cttgctcatt 960 ctgttactag aggactggct tatcttcaca cagaattacc acgaggagat cattataaac 1020 ctgcaatttc ccatcgagat ttaaacagca gaaatgtcct agtgaaaaat gatggaacct 1080 gtgttattag tgactttgga ctgtccatga ggctgactgg aaatagactg gtgcgcccag 1140 gggaggaaga taatgcagcc ataagcgagg ttggcactat cagatatatg gcaccagaag 1200 tgctagaagg agctgtgaac ttgagggact gtgaatcagc tttgaaacaa gtagacatgt 1260 atgctcttgg actaatctat tgggagatat ttatgagatg tacagacctc ttcccagggg 1320 aatccgtacc agagtaccag atggcttttc agacagaggt tggaaaccat cccacttttg 1380 aggatatgca ggttctcgtg tctagggaaa aacagagacc caagttccca gaagcctgga 1440 aagaaaatag cctggcagtg aggtcactca aggagacaat cgaagactgt tgggaccagg 1500 atgcagaggc tcggcttact gcacagtgtg ctgaggaaag gatggctgaa cttatgatga 1560 tttgggaaag aaacaaatct gtgagcccaa cagtcaatcc aatgtctact gctatgcaga 1620 atgaacgtag gtgagtcaac acaagatggc aaatcaggat caggtgaaaa gatcaagaaa 1680 cgtgtgaaaa ctccctattc tcttaagcgg tggcgcccct ccacctgggt catctccact 1740 gaatcgctgg actgtgaagt caacaataat ggcagtaaca gggcagttca ttccaaatcc 1800 agcactgctg tttaccttgc agaaggaggc actgctacaa ccatggtgtc taaagatata 1860 ggaatgaact gtctgtgaaa tgttttcaag cctatggagt gaaattattt tttgcatcat 1920 ttaaacatgc agaagatgtt taaaaataaa aaaaaaactg cttt 1964 124 3611 DNA Homo sapiens 124 cgccccccga ccccggatcg aatccccgcc ctccgcaccc tggatatgtt ttctcccaga 60 cctggatatt tttttgatat cgtgaaacta cgagggaaat aatttggggg atttcttctt 120 ggctccctgc tttccccaca gacatgcctt ccgtttggag ggccgcggca ccccgtccga 180 ggcgaaggaa cccccccagc cgcgagggag agaaatgaag ggaatttctg cagcggcatg 240 aaagctctgc agctaggtcc tctcatcagc catttgtcct ttcaaactgt attgtgatac 300 gggcaggatc agtccacggg agagaagacg agcctcccgg ctgtttctcc gccggtctac 360 ttcccatatt tcttttcttt gccctcctga ttcttggctg gcccagggat gacttcctcg 420 ctgcagcggc cctggcgggt gccctggcta ccatggacca tcctgctggt cagcactgcg 480 gctgcttcgc agaatcaaga acggctatgt gcgtttaaag atccgtatca gcaagacctt 540 gggataggtg agagtagaat ctctcatgaa aatgggacaa tattatgctc gaaaggtagc 600 acctgctatg gcctttggga gaaatcaaaa ggggacataa atcttgtaaa acaaggatgt 660 tggtctcaca ttggagatcc ccaagagtgt cactatgaag aatgtgtagt aactaccact 720 cctccctcaa ttcagaatgg aacataccgt ttctgctgtt gtagcacaga tttatgtaat 780 gtcaacttta ctgagaattt tccacctcct gacacaacac cactcagtcc acctcattca 840 tttaaccgag atgagacaat aatcattgct ttggcatcag tctctgtatt agctgttttg 900 atagttgcct tatgctttgg atacagaatg ttgacaggag accgtaaaca aggtcttcac 960 agtatgaaca tgatggaggc agcagcatcc gaaccctctc ttgatctaga taatctgaaa 1020 ctgttggagc tgattggccg aggtcgatat ggagcagtat ataaaggctc cttggatgag 1080 cgtccagttg ctgtaaaagt gttttccttt gcaaaccgtc agaattttat caacgaaaag 1140 aacatttaca gagtgccttt gatggaacat gacaacattg cccgctttat agttggagat 1200 gagagagtca ctgcagatgg acgcatggaa tatttgcttg tgatggagta ctatcccaat 1260 ggatctttat gcaagtattt aagtctccac acaagtgact gggtaagctc ttgccgtctt 1320 gctcattctg ttactagagg actggcttat cttcacacag aattaccacg aggagatcat 1380 tataaacctg caatttccca tcgagattta aacagcagaa atgtcctagt gaaaaatgat 1440 ggaacctgtg ttattagtga ctttggactg tccatgaggc tgactggaaa tagactggtg 1500 cgcccagggg aggaagataa tgcagccata agcgaggttg gcactatcag atatatggca 1560 ccagaagtgc tagaaggagc tgtgaacttg agggactgtg aatcagcttt gaaacaagta 1620 gacatgtatg ctcttggact aatctattgg gagatattta tgagatgtac agacctcttc 1680 ccaggggaat ccgtaccaga gtaccagatg gcttttcaga cagaggttgg aaaccatccc 1740 acttttgagg atatgcaggt tctcgtgtct agggaaaaac agagacccaa gttcccagaa 1800 gcctggaaag aaaatagcct ggcagtgagg tcactcaagg agacaatcga agactgttgg 1860 gaccaggatg cagaggctcg gcttactgca cagtgtgctg aggaaaggat ggctgaactt 1920 atgatgattt gggaaagaaa caaatctgtg agcccaacag tcaatccaat gtctactgct 1980 atgcagaatg aacgcaacct gtcacataat aggcgtgtgc caaaaattgg tccttatcca 2040 gattattctt cctcctcata cattgaagac tctatccatc atactgacag catcgtgaag 2100 aatatttcct ctgagcattc tatgtccagc acacctttga ctatagggga aaaaaaccga 2160 aattcaatta actatgaacg acagcaagca caagctcgaa tccccagccc tgaaacaagt 2220 gtcaccagcc tctccaccaa cacaacaacc acaaacacca caggactcac gccaagtact 2280 ggcatgacta ctatatctga gatgccatac ccagatgaaa caaatctgca taccacaaat 2340 gttgcacagt caattgggcc aacccctgtc tgcttacagc tgacagaaga agacttggaa 2400 accaacaagc tagacccaaa agaagttgat aagaacctca aggaaagctc tgatgagaat 2460 ctcatggagc actctcttaa acagttcagt ggcccagacc cactgagcag tactagttct 2520 agcttgcttt acccactcat aaaacttgca gtagaagcaa ctggacagca ggacttcaca 2580 cagactgcaa atggccaagc atgtttgatt cctgatgttc tgcctactca gatctatcct 2640 ctccccaagc agcagaacct tcccaagaga cctactagtt tgcctttgaa caccaaaaat 2700 tcaacaaaag agccccggct aaaatttggc agcaagcaca aatcaaactt gaaacaagtc 2760 gaaactggag ttgccaagat gaatacaatc aatgcagcag aacctcatgt ggtgacagtc 2820 accatgaatg gtgtggcagg tagaaaccac agtgttaact cccatgctgc cacaacccaa 2880 tatgccaatg ggacagtact atctggccaa acaaccaaca tagtgacaca tagggcccaa 2940 gaaatgttgc agaatcagtt tattggtgag gacacccggc tgaatattaa ttccagtcct 3000 gatgagcatg agcctttact gagacgagag caacaagctg gccatgatga aggtgttctg 3060 gatcgtcttg tggacaggag ggaacggcca ctagaaggtg gccgaactaa ttccaataac 3120 aacaacagca atccatgttc agaacaagat gttcttgcac agggtgttcc aagcacagca 3180 gcagatcctg ggccatcaaa gcccagaaga gcacagaggc ctaattctct ggatctttca 3240 gccacaaatg tcctggatgg cagcagtata cagataggtg agtcaacaca agatggcaaa 3300 tcaggatcag gtgaaaagat caagaaacgt gtgaaaactc cctattctct taagcggtgg 3360 cgcccctcca cctgggtcat ctccactgaa tcgctggact gtgaagtcaa caataatggc 3420 agtaacaggg cagttcattc caaatccagc actgctgttt accttgcaga aggaggcact 3480 gctacaacca tggtgtctaa agatatagga atgaactgtc tgtgaaatgt tttcaagcct 3540 atggagtgaa attatttttt gcatcattta aacatgcaga agatgtttaa aaataaaaaa 3600 aaaactgctt t 3611 125 3871 DNA Homo sapiens 125 ggcctccgca ccctggatat gttttctccc agacctggat atttttttga tatcgtgaaa 60 ctacgaggga aataatttgg gggatttctt cttggctccc tgctttcccc acagacatac 120 cttccgtttg gagggccgcg gcaccccgtc cgaggcgaag gaaccccccc atccgcgagg 180 gagagaaatg aagggaattt ctgcagcggc atgaaagctc tgcagctagg tcctctcatc 240 agccatttgt cctttcaaac tgtattgtga tacgggcagg atcagtccac gggagagaag 300 acgagcctcc cggctgtttc tccgccggtc tacttcccat atttcttttc tttgccctcc 360 tgattcttgg ctggcccagg gatgacttcc tcgctgcagc ggccctggcg ggtgccctgg 420 ctaccatgga ccatcctgct ggtcagcact gcggctgctt cgcagaatca agaacggcta 480 tgtgcgttta aagatccgta tcagcaagac cttgggatag gtgagagtag aatctctcat 540 gaaaatggga caatattatg ctcgaaaggt agcacctgct atggcctttg ggagaaatca 600 aaaggggaca taaatcttgt aaaacaagga tgttggtctc acattggaga tccccaagag 660 tgtcactatg aagaatgtgt agtaactacc actcctccct caattcagaa tggaacatac 720 cgtttctgct gttgtagcac agatttatgt aatgtcaact ttactgagaa ttttccacct 780 cctgacacaa caccactcag tccacctcat tcatttaacc gagatgagac aataatcatt 840 gctttggcat cagtctctgt attagctgtt ttgatagttg ccttatgctt tggatacaga 900 atgttgacag gagaccgtaa acaaggtctt cacagtatga acatgatgga ggcagcagca 960 tccgaaccct ctcttgatct agataatctg aaactgttgg agctgattgg ccgaggtcga 1020 tatggagcag tatataaagg ctccttggat gagcgtccag ttgctgtaaa agtgttttcc 1080 tttgcaaacc gtcagaattt tatcaacgaa aagaacattt acagagtgcc tttgatggaa 1140 catgacaaca ttgcccgctt tatagttgga gatgagagag tcactgcaga tggacgcatg 1200 gaatatttgc ttgtgatgga gtactatccc aatggatctt tatgcaagta tttaagtctc 1260 cacacaagtg actgggtaag ctcttgccgt cttgctcatt ctgttactag aggactggct 1320 tatcttcaca cagaattacc acgaggagat cattataaac ctgcaatttc ccatcgagat 1380 ttaaacagca gaaatgtcct agtgaaaaat gatggaacct gtgttattag tgactttgga 1440 ctgtccatga ggctgactgg aaatagactg gtgcgcccag gggaggaaga taatgcagcc 1500 ataagcgagg ttggcactat cagatatatg gcaccagaag tgctagaagg agctgtgaac 1560 ttgagggact gtgaatcagc tttgaaacaa gtagacatgt atgctcttgg actaatctat 1620 tgggagatat ttatgagatg tacagacctc ttcccagggg aatccgtacc agagtaccag 1680 atggcttttc agacagaggt tggaaaccat cccacttttg aggatatgca ggttctcgtg 1740 tctagggaaa aacagagacc caagttccca gaagcctgga aagaaaatag cctggcagtg 1800 aggtcactca aggagacaat cgaagactgt tgggaccagg atgcagaggc tcggcttact 1860 gcacagtgtg ctgaggaaag gatggctgaa cttatgatga tttgggaaag aaacaaatct 1920 gtgagcccaa cagtcaatcc aatgtctact gctatgcaga atgaacgcaa cctgtcacat 1980 aataggcgtg tgccaaaaat tggtccttat ccagattatt cttcctcctc atacattgaa 2040 gactctatcc atcatactga cagcatcgtg aagaatattt cctctgagca ttctatgtcc 2100 agcacacctt tgactatagg ggaaaaaaac cgaaattcaa ttaactatga acgacagcaa 2160 gcacaagctc gaatccccag ccctgaaaca agtgtcacca gcctctccac caacacaaca 2220 accacaaaca ccacaggact cacgccaagt actggcatga ctactatatc tgagatgcca 2280 tacccagatg aaacaaatct gcataccaca aatgttgcac agtcaattgg gccaacccct 2340 gtctgcttac agctgacaga agaagacttg gaaaccaaca agctagaccc aaaagaagtt 2400 gataagaacc tcaaggaaag ctctgatgag aatctcatgg agcactctct taaacagttc 2460 agtggcccag acccactgag cagtactagt tctagcttgc tttacccact cataaaactt 2520 gcagtagaag caactggaca gcaggacttc acacagactg caaatggcca agcatgtttg 2580 attcctgatg ttctgcctac tcagatctat cctctcccca agcagcagaa ccttcccaag 2640 agacctacta gtttgccttt gaacaccaaa aattcaacaa aagagccccg gctaaaattt 2700 ggcagcaagc acaaatcaaa cttgaaacaa gtcgaaactg gagttgccaa gatgaataca 2760 atcaatgcag cagaacctca tgtggtgaca gtcaccatga atggtgtggc aggtagaaac 2820 cacagtgtta actcccatgc tgccacaacc caatatgcca ataggacagt actatctggc 2880 caaacaacca acatagtgac acatagggcc caagaaatgt tgcagaatca gtttattggt 2940 gaggacaccc ggctgaatat taattccagt cctgatgagc atgagccttt actgagacga 3000 gagcaacaag ctggccatga tgaaggtgtt ctggatcgtc ttgtggacag gagggaacgg 3060 ccactagaag gtggccgaac taattccaat aacaacaaca gcaatccatg ttcagaacaa 3120 gatgttcttg cacagggtgt tccaagcaca gcagcagatc ctgggccatc aaagcccaga 3180 agagcacaga ggcctaattc tctggatctt tcagccacaa atgtcctgga tggcagcagt 3240 atacagatag gtgagtcaac acaagatggc aaatcaggat caggtgaaaa gatcaagaaa 3300 cgtgtgaaaa ctccctattc tcttaagcgg tggcgcccct ccacctgggt catctccact 3360 gaatcgctgg actgtgaagt caacaataat ggcagtaaca gggcagttca ttccaaatcc 3420 agcactgctg tttaccttgc agaaggaggc actgctacaa ccatggtgtc taaagatata 3480 ggaatgaact gtctgtgaaa tgttttcaag cctatggagt gaaattattt tttgcatcat 3540 ttaaacatgc agaagatgtt taccgggcgg ggtgacagga gagagcgtca gcggcaagct 3600 gtggaggatg gggctcagaa tgcagacctg ggctggccgc atggcctctc cctgagccct 3660 gatttgtggt agggaagcag tatgggtgca gtcccctcct aggcctccct ctggggtccc 3720 ccgatcctat cccacctctt cagggtgagc cagcctcacc tcttcctagt cctgagggtg 3780 agggcaggct gaggcaacga gtgggaggtt caaacaagag tgggctggag ccaagggaaa 3840 atagagatga tgtaatttct ttccggaatt c 3871 126 88 PRT Homo sapiens 126 Cys Arg Glu Leu His Phe Thr Arg Tyr Val Thr Asp Gly Pro Cys Arg 1 5 10 15 Ser Ala Lys Pro Val Thr Glu Leu Val Cys Ser Gly Gln Cys Gly Pro 20 25 30 Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg Gly Lys Trp Trp Arg Pro 35 40 45 Ser Gly Pro Asp Phe Arg Cys Ile Pro Asp Arg Tyr Arg Ala Gln Arg 50 55 60 Val Gln Leu Leu Cys Pro Gly Gly Glu Ala Pro Arg Ala Arg Lys Val 65 70 75 80 Arg Leu Val Ala Ser Cys Lys Cys 85 127 82 PRT Homo sapiens 127 Cys Arg Pro Ile Asn Ala Thr Leu Ala Val Glu Lys Glu Gly Cys Pro 1 5 10 15 Val Cys Ile Thr Val Asn Thr Thr Ile Cys Ala Gly Tyr Cys Pro Thr 20 25 30 Met Thr Arg Val Leu Gln Gly Val Leu Pro Ala Leu Pro Gln Val Val 35 40 45 Cys Asn Tyr Arg Asp Val Arg Phe Glu Ser Ile Arg Leu Pro Gly Cys 50 55 60 Pro Arg Gly Val Asn Pro Val Val Ser Tyr Ala Val Ala Leu Ser Cys 65 70 75 80 Gln Cys 128 82 PRT Homo sapiens 128 Cys Glu Leu Thr Asn Ile Thr Ile Ala Ile Glu Lys Glu Glu Cys Arg 1 5 10 15 Phe Cys Ile Ser Ile Asn Thr Thr Trp Cys Ala Gly Tyr Cys Tyr Thr 20 25 30 Arg Asp Leu Val Tyr Lys Asp Pro Ala Arg Pro Lys Ile Gln Lys Thr 35 40 45 Cys Thr Phe Lys Glu Leu Val Tyr Glu Thr Val Arg Val Pro Gly Cys 50 55 60 Ala His His Ala Asp Ser Leu Tyr Thr Tyr Pro Val Ala Thr Gln Cys 65 70 75 80 His Cys 129 84 PRT Homo sapiens 129 Cys Ile Pro Thr Glu Tyr Thr Met His Ile Glu Arg Arg Glu Cys Ala 1 5 10 15 Tyr Cys Leu Thr Ile Asn Thr Thr Ile Cys Ala Gly Tyr Cys Met Thr 20 25 30 Arg Asp Ile Asn Gly Lys Leu Phe Leu Pro Lys Tyr Ala Leu Ser Gln 35 40 45 Asp Val Cys Thr Tyr Arg Asp Phe Ile Tyr Arg Thr Val Glu Ile Pro 50 55 60 Gly Cys Pro Leu His Val Ala Pro Tyr Phe Ser Tyr Pro Val Ala Leu 65 70 75 80 Ser Cys Lys Cys 130 83 PRT Homo sapiens 130 Cys Asn Asp Ile Thr Ala Arg Leu Gln Tyr Val Lys Val Gly Ser Cys 1 5 10 15 Lys Ser Glu Val Glu Val Asp Ile His Tyr Cys Gln Gly Lys Cys Ala 20 25 30 Ser Lys Ala Met Tyr Ser Ile Asp Ile Asn Asp Val Gln Asp Gln Cys 35 40 45 Ser Cys Cys Ser Pro Thr Arg Thr Glu Pro Met Gln Val Ala Leu His 50 55 60 Cys Thr Asn Gly Ser Val Val Tyr His Glu Val Leu Asn Ala Met Glu 65 70 75 80 Cys Lys Cys 131 80 PRT Homo sapiens 131 Cys Ser Thr Val Pro Val Thr Thr Glu Val Ser Tyr Ala Gly Cys Thr 1 5 10 15 Lys Thr Val Leu Met Asn His Cys Ser Gly Ser Cys Gly Thr Phe Val 20 25 30 Met Tyr Ser Ala Lys Ala Gln Ala Leu Asp His Ser Cys Ser Cys Cys 35 40 45 Lys Glu Glu Lys Thr Ser Gln Arg Glu Val Val Leu Ser Cys Pro Asn 50 55 60 Gly Gly Ser Leu Thr His Thr Tyr Thr His Ile Glu Ser Cys Gln Cys 65 70 75 80 132 80 PRT Homo sapiens 132 Cys Arg Thr Val Pro Phe Ser Gln Thr Ile Thr His Glu Gly Cys Glu 1 5 10 15 Lys Val Val Val Gln Asn Asn Leu Cys Phe Gly Lys Cys Gly Ser Val 20 25 30 His Phe Pro Gly Ala Ala Gln His Ser His Thr Ser Cys Ser His Cys 35 40 45 Leu Pro Ala Lys Phe Thr Thr Met His Leu Pro Leu Asn Cys Thr Glu 50 55 60 Leu Ser Ser Val Ile Lys Val Val Met Leu Val Glu Glu Cys Gln Cys 65 70 75 80 133 85 PRT Homo sapiens 133 Cys Lys Thr Gln Pro Leu Lys Gln Thr Ile His Glu Glu Gly Cys Asn 1 5 10 15 Ser Arg Thr Ile Ile Asn Arg Phe Cys Tyr Gly Gln Cys Asn Ser Phe 20 25 30 Tyr Ile Pro Arg His Ile Arg Lys Glu Glu Gly Ser Phe Gln Ser Cys 35 40 45 Ser Phe Cys Lys Pro Lys Lys Phe Thr Thr Met Met Val Thr Leu Asn 50 55 60 Cys Pro Glu Leu Gln Pro Pro Thr Lys Lys Lys Arg Val Thr Arg Val 65 70 75 80 Lys Gln Cys Arg Cys 85 134 86 PRT Homo sapiens 134 Cys Glu Ala Lys Asn Ile Thr Gln Ile Val Gly His Ser Gly Cys Glu 1 5 10 15 Ala Lys Ser Ile Gln Asn Arg Ala Cys Leu Gly Gln Cys Phe Ser Tyr 20 25 30 Ser Val Pro Asn Thr Phe Pro Gln Ser Thr Glu Ser Leu Val His Cys 35 40 45 Asp Ser Cys Met Pro Ala Gln Ser Met Trp Glu Ile Val Thr Leu Glu 50 55 60 Cys Pro Gly His Glu Glu Val Pro Arg Val Asp Lys Leu Val Glu Lys 65 70 75 80 Ile Leu His Cys Ser Cys 85 135 70 PRT Homo sapies 135 Cys Ile Arg Thr Pro Lys Ile Ser Lys Pro Ile Lys Phe Glu Leu Ser 1 5 10 15 Gly Cys Thr Ser Met Lys Thr Tyr Arg Ala Lys Phe Cys Gly Val Cys 20 25 30 Thr Asp Gly Arg Cys Cys Thr Pro His Arg Thr Thr Thr Leu Pro Val 35 40 45 Glu Phe Lys Cys Pro Asp Gly Glu Val Met Lys Lys Asn Met Met Phe 50 55 60 Ile Lys Thr Cys Ala Cys 65 70 136 70 PRT Homo sapiens 136 Cys Leu Arg Thr Lys Lys Ser Leu Lys Ala Ile His Leu Gln Phe Lys 1 5 10 15 Asn Cys Thr Ser Leu His Thr Tyr Lys Pro Arg Phe Cys Gly Val Cys 20 25 30 Ser Asp Gly Arg Cys Cys Thr Pro His Asn Thr Lys Thr Ile Gln Ala 35 40 45 Glu Phe Gln Cys Ser Pro Gly Gln Ile Val Lys Lys Pro Val Met Val 50 55 60 Ile Gly Thr Cys Thr Cys 65 70 137 70 PRT Homo sapiens 137 Cys Ser Lys Thr Lys Lys Ser Pro Glu Pro Val Arg Phe Thr Tyr Ala 1 5 10 15 Gly Cys Leu Ser Val Lys Lys Tyr Arg Pro Lys Tyr Cys Gly Ser Cys 20 25 30 Val Asp Gly Arg Cys Cys Thr Pro Gln Leu Thr Arg Thr Val Lys Met 35 40 45 Arg Phe Arg Cys Glu Asp Gly Glu Thr Phe Ser Lys Asn Val Met Met 50 55 60 Ile Gln Ser Cys Lys Cys 65 70 138 205 PRT Homo sapiens 138 Gln His Tyr Leu His Ile Arg Pro Ala Pro Ser Asp Asn Leu Pro Leu 1 5 10 15 Val Asp Leu Ile Glu His Pro Asp Pro Ile Phe Asp Pro Lys Glu Lys 20 25 30 Asp Leu Asn Glu Thr Leu Leu Arg Ser Leu Leu Gly Gly His Tyr Asp 35 40 45 Pro Gly Phe Met Ala Thr Ser Pro Pro Glu Asp Arg Pro Gly Gly Gly 50 55 60 Gly Gly Ala Ala Gly Gly Ala Glu Asp Leu Ala Glu Leu Asp Gln Leu 65 70 75 80 Leu Arg Gln Arg Pro Ser Gly Ala Met Pro Ser Glu Ile Lys Gly Leu 85 90 95 Glu Phe Ser Glu Gly Leu Ala Gln Gly Lys Lys Gln Arg Leu Ser Lys 100 105 110 Lys Leu Arg Arg Lys Leu Gln Met Trp Leu Trp Ser Gln Thr Phe Cys 115 120 125 Pro Val Leu Tyr Ala Trp Asn Asp Leu Gly Ser Arg Phe Trp Pro Arg 130 135 140 Tyr Val Lys Val Gly Ser Cys Phe Ser Lys Arg Ser Cys Ser Val Pro 145 150 155 160 Glu Gly Met Val Cys Lys Pro Ser Lys Ser Val His Leu Thr Val Leu 165 170 175 Arg Trp Arg Cys Gln Arg Arg Gly Gly Gln Arg Cys Gly Trp Ile Pro 180 185 190 Ile Gln Tyr Pro Ile Ile Ser Glu Cys Lys Cys Ser Cys 195 200 205 139 197 PRT Gallus gallus 139 Gln His Tyr Leu His Ile Arg Pro Ala Pro Ser Asp Asn Leu Pro Leu 1 5 10 15 Val Asp Leu Ile Glu His Pro Asp Pro Ile Phe Asp Pro Lys Glu Lys 20 25 30 Asp Leu Asn Glu Thr Leu Leu Arg Ser Leu Met Gly Gly His Phe Asp 35 40 45 Pro Asn Phe Met Ala Met Ser Leu Pro Glu Asp Arg Leu Gly Val Asp 50 55 60 Asp Leu Ala Glu Leu Asp Leu Leu Leu Arg Gln Arg Pro Ser Gly Ala 65 70 75 80 Met Pro Gly Glu Ile Lys Gly Leu Glu Phe Tyr Asp Gly Leu Gln Pro 85 90 95 Gly Lys Lys His Arg Leu Ser Lys Lys Leu Arg Arg Lys Leu Gln Met 100 105 110 Trp Leu Trp Ser Gln Thr Phe Cys Pro Val Leu Tyr Thr Trp Asn Asp 115 120 125 Leu Gly Ser Arg Phe Trp Pro Arg Tyr Val Lys Val Gly Ser Cys Tyr 130 135 140 Ser Lys Arg Ser Cys Ser Val Pro Glu Gly Met Val Cys Lys Pro Ala 145 150 155 160 Lys Ser Val His Leu Thr Ile Leu Arg Trp Arg Cys Gln Arg Arg Gly 165 170 175 Gly Gln Arg Cys Thr Trp Ile Pro Ile Gln Tyr Pro Ile Ile Ala Glu 180 185 190 Cys Lys Cys Ser Cys 195 140 196 PRT Xenopus laevis 140 Gln His Tyr Leu His Ile Arg Pro Ala Pro Ser Glu Asn Leu Pro Leu 1 5 10 15 Val Asp Leu Ile Glu His Pro Asp Pro Ile Tyr Asp Pro Lys Glu Lys 20 25 30 Asp Leu Asn Glu Thr Leu Leu Arg Thr Leu Met Val Gly His Phe Asp 35 40 45 Pro Asn Phe Met Ala Thr Ile Leu Pro Glu Glu Arg Leu Gly Val Glu 50 55 60 Asp Leu Gly Glu Leu Asp Leu Leu Leu Arg Gln Lys Pro Ser Gly Ala 65 70 75 80 Met Pro Ala Glu Ile Lys Gly Leu Glu Phe Tyr Glu Gly Leu Gln Ser 85 90 95 Lys Lys His Arg Leu Ser Lys Lys Leu Arg Arg Lys Leu Gln Met Trp 100 105 110 Leu Trp Ser Gln Thr Phe Cys Pro Val Leu Tyr Thr Trp Asn Asp Leu 115 120 125 Gly Thr Arg Phe Trp Pro Arg Tyr Val Lys Val Gly Ser Cys Tyr Ser 130 135 140 Lys Arg Ser Cys Ser Val Pro Glu Gly Met Val Cys Lys Ala Ala Lys 145 150 155 160 Ser Met His Leu Thr Ile Leu Arg Trp Arg Cys Gln Arg Arg Val Gln 165 170 175 Gln Lys Cys Ala Trp Ile Thr Ile Gln Tyr Pro Val Ile Ser Glu Cys 180 185 190 Lys Cys Ser Cys 195 141 195 PRT Takifugu rubripes 141 Gln Pro Tyr Tyr Leu Leu Arg Pro Ile Pro Ser Asp Ser Leu Pro Ile 1 5 10 15 Val Glu Leu Lys Glu Asp Pro Gly Pro Val Phe Asp Pro Lys Glu Arg 20 25 30 Asp Leu Asn Glu Thr Glu Leu Lys Ser Val Leu Gly Asp Phe Asp Ser 35 40 45 Arg Phe Leu Ser Val Leu Pro Pro Ala Glu Asp Gly His Ala Gly Asn 50 55 60 Asp Glu Leu Asp Asp Phe Asp Ala Gln Arg Trp Gly Gly Ala Leu Pro 65 70 75 80 Lys Glu Ile Arg Ala Val Asp Phe Asp Ala Pro Gln Leu Gly Lys Lys 85 90 95 His Lys Pro Ser Lys Lys Leu Lys Arg Arg Leu Gln Gln Trp Leu Trp 100 105 110 Ala Tyr Ser Phe Cys Pro Leu Ala His Ala Trp Thr Asp Leu Gly Ser 115 120 125 Arg Phe Trp Pro Arg Phe Val Arg Ala Gly Ser Cys Leu Ser Lys Arg 130 135 140 Ser Cys Ser Val Pro Glu Gly Met Thr Cys Lys Pro Ala Thr Ser Thr 145 150 155 160 His Leu Thr Ile Leu Arg Trp Arg Cys Val Gln Arg Lys Val Gly Leu 165 170 175 Lys Cys Ala Trp Ile Pro Met Gln Tyr Pro Val Ile Thr Asp Cys Lys 180 185 190 Cys Ser Cys 195 142 196 PRT Danio rerio 142 Gln His Tyr Tyr Leu Leu Arg Pro Ile Pro Ser Asp Ser Leu Pro Ile 1 5 10 15 Val Glu Leu Lys Glu Asp Pro Asp Pro Val Leu Asp Pro Lys Glu Arg 20 25 30 Asp Leu Asn Glu Thr Glu Leu Arg Ala Ile Leu Gly Ser His Phe Glu 35 40 45 Gln Asn Phe Met Ser Ile Asn Pro Pro Glu Asp Lys His Ala Gly Gln 50 55 60 Asp Glu Leu Asn Glu Ser Glu Leu Met Lys Gln Arg Pro Asn Gly Ile 65 70 75 80 Met Pro Lys Glu Ile Lys Ala Met Glu Phe Asp Ile Gln His Gly Lys 85 90 95 Lys His Lys Pro Ser Lys Lys Leu Arg Arg Arg Leu Gln Leu Trp Leu 100 105 110 Trp Ser Tyr Thr Phe Cys Pro Val Val His Thr Trp Gln Asp Leu Gly 115 120 125 Asn Arg Phe Trp Pro Arg Tyr Leu Lys Val Gly Ser Cys Tyr Asn Lys 130 135 140 Arg Ser Cys Ser Val Pro Glu Gly Met Val Cys Lys Pro Pro Lys Ser 145 150 155 160 Ser His Leu Thr Val Leu Arg Trp Arg Cys Val Gln Arg Lys Gly Gly 165 170 175 Leu Lys Cys Ala Trp Ile Pro Val Gln Tyr Pro Val Ile Ser Glu Cys 180 185 190 Lys Cys Ser Cys 195 143 188 PRT Mus musculus 143 Gln Gly Trp Gln Ala Phe Arg Asn Asp Ala Thr Glu Val Ile Pro Gly 1 5 10 15 Leu Gly Glu Tyr Pro Glu Pro Pro Pro Glu Asn Asn Gln Thr Met Asn 20 25 30 Arg Ala Glu Asn Gly Gly Arg Pro Pro His His Pro Tyr Asp Ala Lys 35 40 45 Gly Val Ser Glu Tyr Ser Cys Arg Glu Leu His Tyr Thr Arg Phe Leu 50 55 60 Thr Asp Gly Pro Cys Arg Ser Ala Lys Pro Val Thr Glu Leu Val Cys 65 70 75 80 Ser Gly Gln Cys Gly Pro Ala Arg Leu Leu Pro Asn Ala Ile Gly Arg 85 90 95 Val Lys Trp Trp Arg Pro Asn Gly Pro Asp Phe Arg Cys Ile Pro Asp 100 105 110 Arg Tyr Arg Ala Gln Arg Val Gln Leu Leu Cys Pro Gly Gly Ala Ala 115 120 125 Pro Arg Ser Arg Lys Val Arg Leu Val Ala Ser Cys Lys Cys Lys Arg 130 135 140 Leu Thr Arg Phe His Asn Gln Ser Glu Leu Lys Asp Phe Gly Pro Glu 145 150 155 160 Thr Ala Arg Pro Gln Lys Gly Arg Lys Pro Arg Pro Gly Ala Arg Gly 165 170 175 Ala Lys Ala Asn Gln Ala Glu Leu Glu Asn Ala Tyr 180 185

Claims (20)

We claim the following:
1. An antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide, said sclerostin polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the antibody competitively inhibits binding of the sclerostin polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in a sequence selected from the group consisting of GenBank Acc. Nos. NM004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); and AAB33865 (SEQ ID NO: 110) and wherein the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in a sequence selected from the group consisting of GenBank Ace. NOs. U25 110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); and NM001204 (SEQ ID NO: 113).
2. An antibody, or an antigen-binding fragment thereof, that binds specifically to a sclerostin polypeptide and that impairs formation of a sclerostin homodimer, wherein the sclerostin polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65.
3. The antibody of either claim 1 or claim 2, wherein the antibody is a polyclonal antibody.
4. The antibody of either claim 1 or claim 2, wherein the antibody is a monoclonal antibody.
5. The antibody of claim 4 wherein the monoclonal antibody is selected from the group consisting of a mouse monoclonal antibody, a human monoclonal antibody, a rat monoclonal antibody, and a hamster monoclonal antibody.
6. A hybridoma cell producing the antibody of claim 4.
7. A host cell that is capable of expressing the antibody of claim 4.
8. The antibody of either claim 1 or claim 2, wherein the antibody is a humanized antibody or a chimeric antibody.
9. A host cell that is capable of expressing the antibody of claim 8.
10. The antibody of either claim 1 or claim 2, wherein the antigen-binding fragment is selected from the group consisting of F(ab′)2, Fab′, Fab, Fd, and Fv.
11. The antibody of either claim 1 or claim 2 that comprises a single chain antibody.
12. A host cell that is capable of expressing the antibody of claim 11.
13. A composition comprising an antibody, or antigen-binding fragment thereof, according to either claim 1 or claim 2 and a physiologically acceptable carrier.
14. An immunogen comprising a peptide comprising at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site, wherein the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising an amino acid sequence set forth in a sequence selected from the group consisting of GenBank Acc. Nos. NM004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM 001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); and AAB33865 (SEQ ID NO: 110) and wherein the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in a sequence selected from the group consisting of GenBank Acc. NOs. U25110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); and NM001204 (SEQ ID NO: 113).
15. An immunogen comprising a peptide that comprises at least 21 consecutive amino acids and no more than 50 consecutive amino acids of a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, wherein the peptide is capable of eliciting in a non-human animal an antibody that binds specifically to the SOST polypeptide and that impairs formation of a SOST homodimer.
16. The immunogen of either claim 14 or claim 15 wherein the peptide is associated with a carrier molecule.
17. The immunogen of claim 16 wherein the carrier molecule is carrier polypeptide.
18. The immunogen of claim 17 wherein the carrier polypeptide is keyhole limpet hemocyanin.
19. A method for producing an antibody that specifically binds to a SOST polypeptide, comprising immunizing a non-human animal with an immunogen according to claim 14, wherein (a) the SOST polypeptide comprises an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65; (b) the antibody competitively inhibits binding of the SOST polypeptide to at least one of (i) a bone morphogenic protein (BMP) Type I Receptor binding site and (ii) a BMP Type II Receptor binding site; (c) the BMP Type I Receptor binding site is capable of binding to a BMP Type I Receptor polypeptide comprising the amino acid sequence set forth in a sequence selected from the group consisting of GenBank Acc. Nos. NM004329 (SEQ ID NO: 102); D89675 (SEQ ID NO: 103); NM001203 (SEQ ID NO: 104); S75359 (SEQ ID NO: 105); NM030849 (SEQ ID NO: 106); D38082 (SEQ ID NO: 107); NP001194 (SEQ ID NO: 108); BAA19765 (SEQ ID NO: 109); and AAB33865 (SEQ ID NO: 110); and (d) the BMP Type II Receptor binding site is capable of binding to a BMP Type II Receptor polypeptide comprising the amino acid sequence set forth in a sequence selected from the group consisting of GenBank Acc. NOs. U25110 (SEQ ID NO: 111); NM033346 (SEQ ID NO: 112); Z48923 (SEQ ID NO: 114); CAA88759 (SEQ ID NO: 115); and NM001204 (SEQ ID NO: 113).
20. A method for producing an antibody that specifically binds to a SOST polypeptide, said SOST polypeptide comprising an amino acid sequence set forth in SEQ ID NOS: 2, 6, 8, 14, 46, or 65, comprising immunizing a non-human animal with an immunogen according to claim 15, wherein the antibody impairs formation of a SOST homodimer.
US10/463,190 1998-11-27 2003-06-16 Compositions and methods for increasing bone mineralization Abandoned US20040009535A1 (en)

Priority Applications (34)

Application Number Priority Date Filing Date Title
US10/463,190 US20040009535A1 (en) 1998-11-27 2003-06-16 Compositions and methods for increasing bone mineralization
EP10013087A EP2338906A1 (en) 2003-06-16 2004-06-15 Compostion and methods for increasing bone mineralization
NZ544617A NZ544617A (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
SI200432101T SI1638999T1 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
CN2012105588665A CN103319596A (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
EP04776552.4A EP1638999B1 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
EA201001528A EA022991B1 (en) 2003-06-16 2004-06-15 Sclerostin antibodies and uses thereof
CA2529578A CA2529578C (en) 2003-06-16 2004-06-15 Antibodies for use in increasing bone mineralization
CN2004800233260A CN1835968B (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
ES04776552T ES2428767T3 (en) 2003-06-16 2004-06-15 Compositions and procedures to increase bone mineralization
YU20050932A RS52769B (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
PCT/US2004/018910 WO2005003158A2 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
JP2006517259A JP4818107B2 (en) 2003-06-16 2004-06-15 Compositions and methods for enhancing bone mineralization
BRPI0411535-0A BRPI0411535A (en) 2003-06-16 2004-06-15 compositions and methods for increasing bone mineralization
DK04776552.4T DK1638999T3 (en) 2003-06-16 2004-06-15 COMPOSITIONS AND PROCEDURES FOR INCREASING BONE MINERALIZATION
EA200600037A EA014525B1 (en) 2003-06-16 2004-06-15 Antibodies to sclerostin and method for use thereof
PL04776552T PL1638999T3 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
KR1020057024173A KR101221147B1 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
MEP-31/08A MEP3108A (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
SG2008068264A SG185824A1 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
EP10013086A EP2341071A1 (en) 2003-06-16 2004-06-15 Compostion and methods for increasing bone mineralization
PT47765524T PT1638999E (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
AU2004253870A AU2004253870B2 (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
MEP-2008-31A ME00023B (en) 2003-06-16 2004-06-15 Compositions and methods for increasing bone mineralization
NO20060235A NO20060235L (en) 2003-06-16 2005-01-16 Compositions and Methods for Increasing Bone Mineralization
IL172599A IL172599A (en) 2003-06-16 2005-12-15 Antibodies and antigen-binding fragments thereof that bind to a sclerostin polypeptide, compositions containing the same and methods for the production thereof
ZA200600384A ZA200600384B (en) 2003-06-16 2006-01-13 Compositions and methods for increasing bone mineralization
US11/399,210 US7578999B2 (en) 1998-11-27 2006-04-05 Antibodies specific for sclerostin
HK06109124.4A HK1088907A1 (en) 2003-06-16 2006-08-16 Compositions and methods for increasing bone mineralization
US12/504,296 US7985834B2 (en) 2003-06-16 2009-07-16 Compositions and methods for increasing bone mineralization
JP2011157675A JP2011246482A (en) 2003-06-16 2011-07-19 Composition and method for increasing bone mineralization
IL222756A IL222756A0 (en) 2003-06-16 2012-10-29 Antibodies and antigen-binding fragments thereof that bind to a sclerostin polypeptide, compositions containing the same and methods for the production thereof
CY20131100883T CY1114701T1 (en) 2003-06-16 2013-10-09 COMPOSITIONS AND METHODS FOR INCREASING BONE BALANCING
HRP20131014TT HRP20131014T1 (en) 2003-06-16 2013-10-24 Compositions and methods for increasing bone mineralization

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US11028398P 1998-11-27 1998-11-27
US09/449,218 US6395511B1 (en) 1998-11-27 1999-11-24 Nucleic acids encoding a novel family of TGF-β binding proteins from humans
US10/095,248 US7572899B2 (en) 1998-11-27 2002-03-07 Compositions and methods for increasing bone mineralization
US10/463,190 US20040009535A1 (en) 1998-11-27 2003-06-16 Compositions and methods for increasing bone mineralization

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/095,248 Continuation-In-Part US7572899B2 (en) 1998-11-27 2002-03-07 Compositions and methods for increasing bone mineralization

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/399,210 Continuation US7578999B2 (en) 1998-11-27 2006-04-05 Antibodies specific for sclerostin

Publications (1)

Publication Number Publication Date
US20040009535A1 true US20040009535A1 (en) 2004-01-15

Family

ID=33563705

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/463,190 Abandoned US20040009535A1 (en) 1998-11-27 2003-06-16 Compositions and methods for increasing bone mineralization
US11/399,210 Expired - Lifetime US7578999B2 (en) 1998-11-27 2006-04-05 Antibodies specific for sclerostin
US12/504,296 Expired - Lifetime US7985834B2 (en) 2003-06-16 2009-07-16 Compositions and methods for increasing bone mineralization

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/399,210 Expired - Lifetime US7578999B2 (en) 1998-11-27 2006-04-05 Antibodies specific for sclerostin
US12/504,296 Expired - Lifetime US7985834B2 (en) 2003-06-16 2009-07-16 Compositions and methods for increasing bone mineralization

Country Status (25)

Country Link
US (3) US20040009535A1 (en)
EP (3) EP1638999B1 (en)
JP (2) JP4818107B2 (en)
KR (1) KR101221147B1 (en)
CN (2) CN1835968B (en)
AU (1) AU2004253870B2 (en)
BR (1) BRPI0411535A (en)
CA (1) CA2529578C (en)
CY (1) CY1114701T1 (en)
DK (1) DK1638999T3 (en)
EA (2) EA014525B1 (en)
ES (1) ES2428767T3 (en)
HK (1) HK1088907A1 (en)
HR (1) HRP20131014T1 (en)
IL (2) IL172599A (en)
ME (2) ME00023B (en)
NO (1) NO20060235L (en)
NZ (1) NZ544617A (en)
PL (1) PL1638999T3 (en)
PT (1) PT1638999E (en)
RS (1) RS52769B (en)
SG (1) SG185824A1 (en)
SI (1) SI1638999T1 (en)
WO (1) WO2005003158A2 (en)
ZA (1) ZA200600384B (en)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040058321A1 (en) * 1998-11-27 2004-03-25 Darwin Discovery Ltd. Compositions and methods for increasing bone mineralization
US20050196349A1 (en) * 2003-09-22 2005-09-08 Dianqing Wu Compositions and methods for bone formation and remodeling
US20050261181A1 (en) * 2004-05-19 2005-11-24 Dianqing Wu Compositions and methods for the stimulation or enhancement of bone formation and the self-renewal of cells
US20060030523A1 (en) * 2003-09-22 2006-02-09 Wu Dianqing D Sclerostin and the inhibition of WNT signaling and bone formation
US20060237679A1 (en) * 2005-04-22 2006-10-26 Effebi S.P.A. Valve-actuator connection plate
WO2006119062A2 (en) 2005-05-03 2006-11-09 Ucb Pharma S.A. Sclerostin epitopes
US20070031487A1 (en) * 2005-08-04 2007-02-08 Squashic Steven A Nutritional supplement for women
US20070031486A1 (en) * 2005-08-04 2007-02-08 Squashic Steven A Nutritional supplement for use under physiologically stressful conditions
US20070110747A1 (en) * 2005-05-03 2007-05-17 Ucb S.A. Binding agents
US20080160060A1 (en) * 2006-12-29 2008-07-03 Osteogenex Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
US20080226746A1 (en) * 2005-08-04 2008-09-18 Vertical Pharmaceuticals, Inc. Nutritional supplement for women
US20090074763A1 (en) * 2007-09-17 2009-03-19 Amgen Inc. Method for inhibiting bone resorption
US20100003324A1 (en) * 2008-07-03 2010-01-07 Osteogenex Inc. Vinpocetine and eburn amonine derivatives for promoting bone growth
US20100015665A1 (en) * 2006-11-10 2010-01-21 Ucb Pharma S.A. Antibodies and diagnostics
US20100036091A1 (en) * 2006-11-10 2010-02-11 Amgen Inc. Antibody-based diagnostics and therapeutics
US20100041599A1 (en) * 2006-11-14 2010-02-18 Dakai Liu Compositions and methods for bone formation, bone remodeling and toxin protection
US7799523B2 (en) 2002-04-03 2010-09-21 Celltech R & D, Inc. Association of polymorphisms in the SOST gene region with bone mineral density
US20100298308A1 (en) * 2003-09-22 2010-11-25 Wu Dianqing Dan Compositions and methods for bone formation and remodeling
US7868134B2 (en) 2003-06-16 2011-01-11 Ucb Manufacturing, Inc. Immunogenic peptides derived from sclerostin
US20110044978A1 (en) * 2007-12-14 2011-02-24 Amgen Inc. Method for treating bone fracture
US20110159055A1 (en) * 2005-08-04 2011-06-30 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US7985834B2 (en) 2003-06-16 2011-07-26 Celltech R & D, Inc. Compositions and methods for increasing bone mineralization
WO2011156252A2 (en) 2010-06-07 2011-12-15 Enzo Biochem. Inc. Sulfation of wnt pathway proteins
WO2012145417A1 (en) 2011-04-19 2012-10-26 Amgen Inc. Method for treating osteoporosis
WO2013003178A1 (en) 2011-06-28 2013-01-03 Enzo Biochem, Inc. Sulfation of wnt pathway proteins
WO2013019954A1 (en) 2011-08-04 2013-02-07 Amgen Inc. Method for treating bone gap defects
WO2013032527A1 (en) 2011-09-02 2013-03-07 The Regents Of The University Of California Llp2a-bisphosphonate conjugates for osteoporosis treatment
EP2586796A1 (en) 2007-10-12 2013-05-01 Novartis AG Compositions and methods for use for antibodies against sclerostin
WO2013101451A1 (en) 2011-12-28 2013-07-04 Amgen Inc. Method of treating alvelor bone loss through the use of anti-sclerostin antibodies
CN103432568A (en) * 2005-11-23 2013-12-11 阿塞勒隆制药公司 Activin-ActRIIa antagonists and uses for promoting bone growth
WO2014006100A1 (en) 2012-07-05 2014-01-09 Ucb Pharma S.A. Treatment for bone diseases
WO2014144817A2 (en) 2013-03-15 2014-09-18 Amgen Inc. Inhibitory polypeptides specific to wnt inhibitors
US9046537B2 (en) 2003-09-22 2015-06-02 Enzo Biochem, Inc. Method for treating inflammation by administering a compound which binds LDL-receptor-related protein (LRP) ligand binding domain
US9052324B2 (en) 2004-05-19 2015-06-09 Enzo Biochem, Inc. Compounds and assays for controlling Wnt activity
US9133272B2 (en) 2011-03-01 2015-09-15 Amgen Inc. Bispecific binding agents
US9145457B2 (en) 2011-03-25 2015-09-29 Amgen Inc. Sclerostin antibody crystals and formulations thereof
US9352043B2 (en) 2010-05-14 2016-05-31 Amgen Inc. High concentration antibody formulations
US9617323B2 (en) 2010-06-07 2017-04-11 Joshua Rabbani Sulfonated sclerostin, antibodies, epitopes and methods for identification and use therefor
US9822173B2 (en) 2012-11-21 2017-11-21 Amgen Inc. Heterodimeric immunoglobulins
WO2018031454A1 (en) 2016-08-08 2018-02-15 Amgen Inc. Method of improving connective tissue attachment using anti-sclerostin antibodies
WO2018115879A1 (en) 2016-12-21 2018-06-28 Mereo Biopharma 3 Limited Use of anti-sclerostin antibodies in the treatment of osteogenesis imperfecta
WO2018115880A1 (en) 2016-12-21 2018-06-28 Mereo Biopharma 3 Limited Use of anti-sclerostin antibodies in the treatment of osteogenesis imperfecta
WO2019191534A1 (en) 2018-03-30 2019-10-03 Amgen Inc. C-terminal antibody variants
WO2020033788A1 (en) 2018-08-10 2020-02-13 Amgen Inc. Method of preparing an antibody pharmaceutical formulation
WO2021030179A1 (en) 2019-08-12 2021-02-18 Amgen Inc. Anti-sclerostin antibody formulations
US11167011B2 (en) 2010-06-07 2021-11-09 Enzo Biochem, Inc. Methods for treating bone loss using sclerostin peptides
US11576970B2 (en) 2016-03-10 2023-02-14 UCB Biopharma SRL Pharmaceutical formulations
US11851483B2 (en) 2014-12-12 2023-12-26 Amgen Inc. Anti-sclerostin antibodies and their use to treat bone disorders as part of a regimen

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2476410C (en) * 2002-03-01 2013-09-24 Celltech R & D, Inc. Methods to increase or decrease bone density
US7193069B2 (en) 2002-03-22 2007-03-20 Research Association For Biotechnology Full-length cDNA
US7893218B2 (en) * 2003-06-16 2011-02-22 Stowers Institute For Medical Research Antibodies that specifically bind SOST peptides
US7585501B2 (en) 2002-06-14 2009-09-08 Stowers Institute For Medical Research Compositions and methods for treating kidney disease
WO2003106657A2 (en) * 2002-06-14 2003-12-24 Stowers Institute For Medical Research Wise/sost nucleic acid sequences and amino acid sequences
US9045553B2 (en) 2004-05-27 2015-06-02 Acceleron Pharma, Inc. Cerberus/Coco derivatives and uses thereof
CA2567810A1 (en) 2004-05-27 2005-12-08 Acceleron Pharma Inc. Cerberus/coco derivatives and uses thereof
US7709605B2 (en) 2004-07-23 2010-05-04 Acceleron Pharma Inc. ActRII receptor polypeptides, methods and compositions
US8128933B2 (en) 2005-11-23 2012-03-06 Acceleron Pharma, Inc. Method of promoting bone growth by an anti-activin B antibody
US20090203041A1 (en) * 2006-04-21 2009-08-13 Wei Shi Bmp4 inhibitors
CA2671983A1 (en) 2006-12-08 2009-06-19 Acceleron Pharma Inc. Uses of cerberus, coco and derivatives thereof
US8895016B2 (en) 2006-12-18 2014-11-25 Acceleron Pharma, Inc. Antagonists of activin-actriia and uses for increasing red blood cell levels
ES2415666T3 (en) 2007-02-01 2013-07-26 Acceleron Pharma, Inc. Pharmaceutical compositions comprising Activin-ActRIIa antagonists for use in the prevention or treatment of breast cancer metastases or bone loss related to breast cancer
TW201940502A (en) 2007-02-02 2019-10-16 美商艾瑟勒朗法瑪公司 Variants derived from ActRIIB and uses therefor
TWI667038B (en) 2007-02-09 2019-08-01 美商艾瑟勒朗法瑪公司 Pharmaceutical composition comprising an actriia-fc fusion protein; use of an actriia-fc fusion protein for treatment or prevention of cancer-related bone loss; use of an actriia-fc fusion protein for the treatment or prevention of multiple myeloma
ES2446293T3 (en) 2007-03-20 2014-03-07 Eli Lilly & Company Anti sclerostin antibodies
US8133553B2 (en) 2007-06-18 2012-03-13 Zimmer, Inc. Process for forming a ceramic layer
US8309521B2 (en) 2007-06-19 2012-11-13 Zimmer, Inc. Spacer with a coating thereon for use with an implant device
EP3243524A1 (en) 2007-09-18 2017-11-15 Acceleron Pharma Inc. Activin-actriia antagonists and uses for decreasing or inhibiting fsh secretion
US8608049B2 (en) 2007-10-10 2013-12-17 Zimmer, Inc. Method for bonding a tantalum structure to a cobalt-alloy substrate
JPWO2009110215A1 (en) * 2008-03-03 2011-07-14 独立行政法人科学技術振興機構 Ciliary cell differentiation induction method
US8216997B2 (en) 2008-08-14 2012-07-10 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating anemia using a combination of GDF traps and erythropoietin receptor activators
PT3750552T (en) 2008-08-14 2023-06-28 Acceleron Pharma Inc Gdf traps
WO2010083034A1 (en) 2009-01-13 2010-07-22 Acceleron Pharma Inc. Methods for increasing adiponectin
NZ594481A (en) * 2009-02-06 2012-09-28 Pepscan Systems Bv Truncated cystine-knot proteins
EP2414043B1 (en) * 2009-03-30 2016-01-13 Acceleron Pharma Inc. Bmp-alk3 antagonists and uses for promoting bone growth
WO2010115932A1 (en) 2009-04-08 2010-10-14 Novartis Ag Combination for the treatment of bone loss
WO2010130830A2 (en) * 2009-05-15 2010-11-18 Ablynx N.V. Amino acid sequences directed against sclerostin and polypeptides comprising the same for the treatment of bone diseases and disorders
AU2010258931B2 (en) 2009-06-08 2015-04-23 Acceleron Pharma Inc. Methods for increasing thermogenic adipocytes
MX2011013364A (en) 2009-06-12 2012-03-16 Acceleron Pharma Inc Truncated actriib-fc fusion proteins.
JP6267425B2 (en) 2009-11-17 2018-01-24 アクセルロン ファーマ, インコーポレイテッド ACTRIIB protein and its variants and uses thereof for utrophin induction for the treatment of muscular dystrophy
EP2558106A1 (en) 2010-04-16 2013-02-20 Novartis AG Methods and compositions for improving implant osseointegration
LT3111954T (en) 2010-11-05 2019-07-10 Novartis Ag Methods of treating ankylosing spondylitis using anti-il-17 antibodies
EP2638065A4 (en) 2010-11-08 2014-04-09 Acceleron Pharma Inc Actriia binding agents and uses thereof
FR2974061B1 (en) 2011-04-13 2013-06-07 Michelin Soc Tech METHOD AND APPARATUS FOR COUNTING THE NUMBER OF LANDINGS OF A TIRE OF AN AIRCRAFT
WO2012149246A1 (en) 2011-04-29 2012-11-01 Novartis Ag Methods of treating squamous cell carcinoma related applications
WO2013010254A1 (en) * 2011-07-19 2013-01-24 University Health Network Biomarkers for ankylosing spondylitis
KR102279522B1 (en) 2012-11-02 2021-07-19 셀진 코포레이션 Activin-actrii antagonists and uses for treating bone and other disorders
WO2014118705A1 (en) 2013-01-31 2014-08-07 Novartis Ag Methods of treating chronic kidney disease-mineral and bone disorder using sclerostin antagonists
LT3312195T (en) * 2013-03-20 2020-01-10 Genzyme Corporation Methods for treating osteogenesis imperfecta
WO2014155278A2 (en) 2013-03-26 2014-10-02 Novartis Ag Methods of treating autoimmune diseases using il-17 antagonists
TN2016000553A1 (en) 2014-06-13 2018-04-04 Acceleron Pharma Inc Methods and compositions for treating ulcers
MA41052A (en) 2014-10-09 2017-08-15 Celgene Corp TREATMENT OF CARDIOVASCULAR DISEASE USING ACTRII LIGAND TRAPS
WO2016090077A1 (en) 2014-12-03 2016-06-09 Celgene Corporation Activin-actrii antagonists and uses for treating anemia
GB201510758D0 (en) 2015-06-18 2015-08-05 Ucb Biopharma Sprl Novel TNFa structure for use in therapy
GB201621907D0 (en) * 2016-12-21 2017-02-01 Ucb Biopharma Sprl And Sanofi Antibody epitope
RU2651091C1 (en) * 2016-12-22 2018-04-18 Федеральное Государственное бюджетное образовательное учреждение высшего профессионального образования "Санкт-Петербургский государственный педиатрический медицинский университет" Министерства здравоохранения Российской Федерации (ФГБОУ ВО СПбГПМУ Минздрава России) Method of diagnostics of osteomyelitis in children
WO2019154410A1 (en) * 2018-02-12 2019-08-15 翼高生物科技有限公司 Aptamer for sclerostin and use thereof
GB201810746D0 (en) 2018-06-29 2018-08-15 Mereo Biopharma 3 Ltd Use of sclerostin antagonist

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6395511B1 (en) * 1998-11-27 2002-05-28 Darwin Discovery, Ltd. Nucleic acids encoding a novel family of TGF-β binding proteins from humans

Family Cites Families (162)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US751929A (en) 1904-02-09 Ash-pan
US793398A (en) 1905-03-24 1905-06-27 Herschel A Schermerhorn Fastening for wagon eng-gates.
US4018884A (en) 1975-06-26 1977-04-19 Hoffmann-La Roche Inc. Fluorogenic materials and labeling techniques
US4704362A (en) 1977-11-08 1987-11-03 Genentech, Inc. Recombinant cloning vehicle microbial polypeptide expression
US4528266A (en) 1979-10-01 1985-07-09 George Pieczenik Method of inserting unique DNA sequences into DNA vectors
US4359535A (en) 1979-10-01 1982-11-16 George Pieczenik Autonomously replicating DNA containing inserted DNA sequences
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4486530A (en) 1980-08-04 1984-12-04 Hybritech Incorporated Immunometric assays using monoclonal antibodies
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4411993A (en) 1981-04-29 1983-10-25 Steven Gillis Hybridoma antibody which inhibits interleukin 2 activity
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4427115A (en) 1981-10-19 1984-01-24 Laipply Thomas C One piece alcohol preparation device
US4579821A (en) 1981-11-23 1986-04-01 University Patents, Inc. Control of DNA sequence transcription
USRE32011E (en) 1981-12-14 1985-10-22 Scripps Clinic And Research Foundation Ultrapurification of factor VIII using monoclonal antibodies
JPS58117537A (en) 1982-01-06 1983-07-13 Toray Ind Inc Photosensitive resin composition
US4766075A (en) 1982-07-14 1988-08-23 Genentech, Inc. Human tissue plasminogen activator
US4543439A (en) 1982-12-13 1985-09-24 Massachusetts Institute Of Technology Production and use of monoclonal antibodies to phosphotyrosine-containing proteins
US6054561A (en) 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
DE3417525C1 (en) 1984-05-11 1986-01-09 Matter + Siegmann Ag, Wohlen Device for the quantitative and qualitative detection of hydrocarbon-containing suspended particles in gases
US5288641A (en) 1984-06-04 1994-02-22 Arch Development Corporation Herpes Simplex virus as a vector
US5087571A (en) 1984-06-22 1992-02-11 President And Fellows Of Harvard College Method for providing a cell culture from a transgenic non-human mammal
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
US5272254A (en) 1984-10-02 1993-12-21 Biogen Inc. Production of streptavidin-like polypeptides
WO1986002077A1 (en) 1984-10-02 1986-04-10 Meade Harry M Production of streptavidin-like polypeptides
US4801542A (en) 1984-10-12 1989-01-31 Zymogenetics, Inc. Expression of biologically active PDGF analogs in eucaryotic cells
SG64329A1 (en) 1984-11-16 1999-04-27 American Cyanamid Co Antitumor antibiotics (LL-E33288 complex)
US4902614A (en) 1984-12-03 1990-02-20 Teijin Limited Monoclonal antibody to human protein C
US4897255A (en) 1985-01-14 1990-01-30 Neorx Corporation Metal radionuclide labeled proteins for diagnosis and therapy
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
CA1293460C (en) 1985-10-07 1991-12-24 Brian Lee Sauer Site-specific recombination of dna in yeast
US4744981A (en) 1985-10-17 1988-05-17 Neorx Corporation Trichothecene antibody conjugates
US4935349A (en) 1986-01-17 1990-06-19 Zymogenetics, Inc. Expression of higher eucaryotic genes in aspergillus
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US4851341A (en) 1986-12-19 1989-07-25 Immunex Corporation Immunoaffinity purification system
US5079233A (en) 1987-01-30 1992-01-07 American Cyanamid Company N-acyl derivatives of the LL-E33288 antitumor antibiotics, composition and methods for using the same
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
EP0289121B1 (en) 1987-05-01 1995-12-27 Stratagene Mutagenesis testing using transgenic non-human animals carrying test DNA sequences
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
DE3856559T2 (en) 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
GB8719042D0 (en) 1987-08-12 1987-09-16 Parker D Conjugate compounds
GB8723661D0 (en) 1987-10-08 1987-11-11 British Bio Technology Synthetic gene
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
EP0313874B1 (en) 1987-10-30 1994-04-13 American Cyanamid Company Disulfur analogs of LL-E33288 antitumor agents
US5254678A (en) 1987-12-15 1993-10-19 Gene Shears Pty. Limited Ribozymes
EP0329184A3 (en) 1988-02-19 1990-05-23 Neorx Corporation Antimers and antimeric conjugation
US4988496A (en) 1988-05-31 1991-01-29 Neorx Corporation Metal radionuclide chelating compounds for improved chelation kinetics
US5221778A (en) 1988-08-24 1993-06-22 Yale University Multiplex gene regulation
AU4308689A (en) 1988-09-02 1990-04-02 Protein Engineering Corporation Generation and selection of recombinant varied binding proteins
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA1340323C (en) 1988-09-20 1999-01-19 Arnold E. Hampel Rna catalyst for cleaving specific rna sequences
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5096815A (en) 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
EP0454781B1 (en) 1989-01-23 1998-12-16 Chiron Corporation Recombinant cells for therapies of infection and hyperproliferative disorders and preparation thereof
GB8903022D0 (en) 1989-02-10 1989-03-30 Celltech Ltd Chemical compounds
US5175383A (en) 1989-02-17 1992-12-29 President And Fellows Of Harvard College Animal model for benign prostatic disease
US5549910A (en) 1989-03-31 1996-08-27 The Regents Of The University Of California Preparation of liposome and lipid complex compositions
DE69024953T3 (en) 1989-05-04 2005-01-27 Southern Research Institute, Birmingham encapsulation
GB8914543D0 (en) 1989-06-23 1989-08-09 Parker David Chemical compounds
EP1001032A3 (en) 1989-08-18 2005-02-23 Chiron Corporation Recombinant retroviruses delivering vector constructs to target cells
US5464764A (en) 1989-08-22 1995-11-07 University Of Utah Research Foundation Positive-negative selection methods and vectors
KR920007887B1 (en) 1989-08-29 1992-09-18 스즈키 지도오샤 고오교오 가부시키가이샤 Exhaust gas cleaning device for internal combustion engine
GB8919607D0 (en) 1989-08-30 1989-10-11 Wellcome Found Novel entities for cancer therapy
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5359051A (en) 1990-01-11 1994-10-25 Isis Pharmaceuticals Compounds useful in the synthesis of nucleic acids capable of cleaning RNA
ATE149841T1 (en) 1990-01-26 1997-03-15 Immunomedics Inc VACCINES AGAINST CANCER AND INFECTIOUS DISEASES
US5177197A (en) 1990-02-27 1993-01-05 Ludwig Institute For Cancer Research Isolated nucleotide sequence expressing human transforming growth factor-β1-binding protein
US5466468A (en) 1990-04-03 1995-11-14 Ciba-Geigy Corporation Parenterally administrable liposome formulation comprising synthetic lipids
ATE260974T1 (en) 1990-06-15 2004-03-15 Scios Inc TRANSGENIC NON-HUMAN MAMMAL EXHIBITING THE AMYLOID-FORMING PATHOLOGY OF ALZHEIMER'S DISEASE
JP3218637B2 (en) 1990-07-26 2001-10-15 大正製薬株式会社 Stable aqueous liposome suspension
CA2090126C (en) 1990-08-02 2002-10-22 John W. Schrader Methods for the production of proteins with a desired function
JP2958076B2 (en) 1990-08-27 1999-10-06 株式会社ビタミン研究所 Multilamellar liposome for gene transfer and gene-captured multilamellar liposome preparation and method for producing the same
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
JPH04141095A (en) 1990-10-02 1992-05-14 Chemo Sero Therapeut Res Inst Hiv-resistant recombined modified antibody and method for preparing modified antibody
US5070108A (en) 1990-10-12 1991-12-03 Trustees Of The University Of Pennsylvania Methods of treating osteoporosis, increasing bone mineral content and preventing the occurrence of compression fractures in a mammal
WO1992006693A1 (en) 1990-10-22 1992-04-30 Fox Chase Cancer Center Dna construct for providing rna therapy
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
CA2105300C (en) 1991-03-01 2008-12-23 Robert C. Ladner Process for the development of binding mini-proteins
US6072039A (en) 1991-04-19 2000-06-06 Rohm And Haas Company Hybrid polypeptide comparing a biotinylated avidin binding polypeptide fused to a polypeptide of interest
GB9112536D0 (en) 1991-06-11 1991-07-31 Celltech Ltd Chemical compounds
GB9120467D0 (en) 1991-09-26 1991-11-06 Celltech Ltd Anti-hmfg antibodies and process for their production
DE4135543A1 (en) 1991-10-28 1993-04-29 Boehringer Mannheim Gmbh RECOMBINANT CORE STREPTAVIDINE
US5203975A (en) 1991-10-29 1993-04-20 E. I. Du Pont De Nemours And Company Process for cathodic electrodeposition of a clear coating over a conductive paint layer
WO1993010218A1 (en) 1991-11-14 1993-05-27 The United States Government As Represented By The Secretary Of The Department Of Health And Human Services Vectors including foreign genes and negative selective markers
GB9125623D0 (en) 1991-12-02 1992-01-29 Dynal As Cell modification
ES2102007T3 (en) 1992-01-23 1997-07-16 Merck Patent Gmbh MONOMER AND DIMER ANTIBODY FRAGMENT FUSION PROTEINS.
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
EP0578515A3 (en) 1992-05-26 1995-05-10 Bristol Myers Squibb Co Humanized monoclonal antibodies.
EP0644938A1 (en) 1992-05-29 1995-03-29 E.I. Du Pont De Nemours And Company PRODUCTION OF STREPTAVIDIN FROM $i(BACILLUS SUBTILIS)
WO1993025234A1 (en) 1992-06-08 1993-12-23 The Regents Of The University Of California Methods and compositions for targeting specific tissue
EP0644946A4 (en) 1992-06-10 1997-03-12 Us Health Vector particles resistant to inactivation by human serum.
GB2269175A (en) 1992-07-31 1994-02-02 Imperial College Retroviral vectors
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
CA2153391C (en) 1993-01-11 2000-08-15 Kenneth Rock Inducing cytotoxic t lymphocyte responses
IL108719A0 (en) 1993-02-25 1994-08-26 Ortho Pharma Corp Expression constructs containing hiv inhibitory antisense and other nucleotide sequences, retroviralvectors and recombinant retroviruses containing them
FR2702160B1 (en) 1993-03-02 1995-06-02 Biovecteurs As Synthetic particulate vectors and method of preparation.
FR2704145B1 (en) 1993-04-21 1995-07-21 Pasteur Institut Particulate vector and pharmaceutical composition containing it.
FR2705361B1 (en) 1993-05-18 1995-08-04 Centre Nat Rech Scient Viral vectors and use in gene therapy.
AU700371B2 (en) 1993-06-07 1999-01-07 Genentech Inc. Hiv envelope polypeptides
JPH09506857A (en) 1993-07-16 1997-07-08 オントーゲン コーポレーション Synthesis of combinatorial arrays of organic compounds by multi-component combinatorial array synthesis
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5453492A (en) 1993-07-28 1995-09-26 La Jolla Cancer Research Foundation 60 kDa transforming growth factor-β-binding protein and its use to detect or purify TGF-β
WO1995004277A1 (en) 1993-08-03 1995-02-09 Sphinx Pharmaceuticals Corporation A method for preparing and selecting pharmaceutically useful non-peptide compounds from a structurally diverse universal library
US5624803A (en) 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
AU688428B2 (en) 1993-11-09 1998-03-12 Johns Hopkins University, The Generation of high titers of recombinant AAV vectors
JPH09508355A (en) 1993-12-09 1997-08-26 チバ−ガイギー アクチェンゲゼルシャフト How to make a combinatorial compound library
JPH09506611A (en) 1993-12-15 1997-06-30 コンビケム,インコーポレイテッド Combinatorial library and its use
WO1995016712A1 (en) 1993-12-15 1995-06-22 Smithkline Beecham Corporation Compounds and methods
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
ATE239680T1 (en) 1994-03-11 2003-05-15 Pharmacopeia Inc SULFONAMIDE DERIVATIVES AND THEIR USE
ATE378407T1 (en) 1994-04-29 2007-11-15 Curis Inc MORPHOGENE PROTEIN-SPECIFIC CELL SURFACE RECEPTORS AND THEIR USES
JPH10500112A (en) 1994-05-06 1998-01-06 ファーマコピーア,インコーポレイテッド Combination dihydrobenzopyran library
US5549974A (en) 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US5792456A (en) 1994-08-04 1998-08-11 Bristol-Myers Squibb Company Mutant BR96 antibodies reactive with human carcinomas
FR2723849B1 (en) 1994-08-31 1997-04-11 Biovector Therapeutics Sa PROCESS FOR INCREASING IMMUNOGENICITY, PRODUCT OBTAINED AND PHARMACEUTICAL COMPOSITION
US5463564A (en) 1994-09-16 1995-10-31 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5846770A (en) 1994-11-22 1998-12-08 Genetics Institute, Inc. DNA molecules encoding human chordin
US6057421A (en) 1994-11-30 2000-05-02 Immpheron, Inc. Variable heavy and light chain regions of murine monoclonal antibody 1F7
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
IE80468B1 (en) 1995-04-04 1998-07-29 Elan Corp Plc Controlled release biodegradable nanoparticles containing insulin
EP0871705A4 (en) 1995-06-05 2000-01-26 Human Genome Sciences Inc Human ccn-like growth factor
US5738868A (en) 1995-07-18 1998-04-14 Lipogenics Ltd. Liposome compositions and kits therefor
DK0912738T3 (en) 1996-05-22 2008-11-17 Viventia Biotech Inc Antigen-binding fragments that specifically detect cancer cells, nucleotides encoding the fragments and their use for the prophylaxis and detection of cancers
US6958239B2 (en) 1996-11-21 2005-10-25 Oligos Etc Inc. Three component chimeric antisense oligonucleotides
US6133426A (en) 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US5989909A (en) 1997-09-26 1999-11-23 Millennium Biotherapeutics, Inc. Huchordin and uses thereof
AU6959898A (en) 1997-04-11 1998-11-11 David J. Grainger Compounds and therapies for the prevention of vascular and non-vascular pathol ogies
EP0985039B1 (en) 1997-06-12 2008-02-20 Novartis International Pharmaceutical Ltd. Artificial antibody polypeptides
US6075007A (en) 1997-07-17 2000-06-13 Regeneron Pharmaceuticals, Inc. Modified noggin polypeptide and compositions
EP1000152A2 (en) 1997-08-01 2000-05-17 Genset 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES
US6815201B2 (en) 1997-09-08 2004-11-09 The Public Health Research Institute Of The City Of New York, Inc. HIV-1 gp120 V1/V2 domain epitopes capable of generating neutralizing antibodies
WO2002024888A2 (en) 2000-09-01 2002-03-28 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6231864B1 (en) 1998-02-12 2001-05-15 Immune Complex Corporation Strategically modified hepatitis B core proteins and their derivatives
US6544485B1 (en) 2001-01-29 2003-04-08 Sharper Image Corporation Electro-kinetic device with enhanced anti-microorganism capability
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
PT1150918E (en) 1999-02-03 2005-01-31 Biosante Pharmaceuticals Inc METHOD OF MANUFACTURE OF CALCIUM PHOSPHATE THERAPEUTIC PARTICLES
ES2331657T3 (en) 1999-06-09 2010-01-12 Genentech, Inc. METHODS FOR TUMORS DIAGNOSIS.
JP4141095B2 (en) 1999-10-29 2008-08-27 三洋電機株式会社 Semiconductor device and manufacturing method thereof
WO2001064885A1 (en) 2000-03-02 2001-09-07 Amgen, Inc. Chordin-like-2 molecules and uses thereof
CA2410912A1 (en) * 2000-06-01 2001-12-06 Amgen, Inc. Cystine-knot polypeptides: cloaked-2 molecules and uses thereof
JP2004520005A (en) 2000-06-19 2004-07-08 エフ.ホフマン−ラ ロシュ アーゲー Osteolevin gene polymorphism
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
CA2374027A1 (en) 2001-03-13 2002-09-13 The Minister Of National Defence Cloning, expression, sequencing, and functional enhancement of monoclonal scfv antibody against venezuelan equine encephalitis virus(vee)
US20030186915A1 (en) 2002-02-11 2003-10-02 Yang Pan Regulatory polynucleotides and uses thereof
FR2838379B1 (en) 2002-04-12 2005-06-24 Valeo Climatisation DEVICE FOR PURIFYING THE AIR OF THE CABIN OF A MOTOR VEHICLE
WO2003106657A2 (en) * 2002-06-14 2003-12-24 Stowers Institute For Medical Research Wise/sost nucleic acid sequences and amino acid sequences
US7893218B2 (en) 2003-06-16 2011-02-22 Stowers Institute For Medical Research Antibodies that specifically bind SOST peptides
DE10255152A1 (en) 2002-11-26 2004-06-03 Von Langen Ursula Lang pollutant sucker
US20040141875A1 (en) 2003-01-15 2004-07-22 Rajiv Doshi System and method for treating microorganisms within motor vehicle heating, ventilation, and air conditioning units
US7381409B2 (en) * 2003-06-16 2008-06-03 Celltech R&D, Inc. Antibodies specific for sclerostin and methods of screening and use therefor
US7592429B2 (en) * 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
ES2446293T3 (en) * 2007-03-20 2014-03-07 Eli Lilly & Company Anti sclerostin antibodies
AR068767A1 (en) * 2007-10-12 2009-12-02 Novartis Ag ANTIBODIES AGAINST SCLEROSTIN, COMPOSITIONS AND METHODS OF USE OF THESE ANTIBODIES TO TREAT A PATHOLOGICAL DISORDER MEDIATIONED BY SCLEROSTIN

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6395511B1 (en) * 1998-11-27 2002-05-28 Darwin Discovery, Ltd. Nucleic acids encoding a novel family of TGF-β binding proteins from humans
US6495736B1 (en) * 1998-11-27 2002-12-17 Darwin Discovery, Ltd. Compositions and methods for increasing bone mineralization

Cited By (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8986685B2 (en) 1998-11-27 2015-03-24 Ucb Pharma S.A. Compositions and methods for increasing bone mineralization
US20040158045A1 (en) * 1998-11-27 2004-08-12 Darwin Discovery Ltd. Antibodies associated with alterations in bone density
US20080182788A1 (en) * 1998-11-27 2008-07-31 Darwin Discovery Limited Compositions and Methods for Increasing Bone Mineralization
US20080234219A1 (en) * 1998-11-27 2008-09-25 Darwin Discovery Limited Compositions and Methods for Increasing Bone Mineralization
US20110150866A1 (en) * 1998-11-27 2011-06-23 Darwin Discovery Limited Compositions and Methods for Increasing Bone Mineralization
US7977312B2 (en) 1998-11-27 2011-07-12 Darwin Discovery Limited Compositions and methods for increasing bone mineralization
US9791462B2 (en) 1998-11-27 2017-10-17 Ucb Pharma, S.A. Compositions and methods for increasing bone mineralization
US7994299B2 (en) 1998-11-27 2011-08-09 Darwin Discovery Limited Compositions and methods for increasing bone mineralization
US20040058321A1 (en) * 1998-11-27 2004-03-25 Darwin Discovery Ltd. Compositions and methods for increasing bone mineralization
US7758858B2 (en) 1998-11-27 2010-07-20 Darwin Discovery Ltd. Antibodies associated with alterations in bone density
US7572899B2 (en) 1998-11-27 2009-08-11 Ucb Sa Compositions and methods for increasing bone mineralization
US7799523B2 (en) 2002-04-03 2010-09-21 Celltech R & D, Inc. Association of polymorphisms in the SOST gene region with bone mineral density
US8563271B2 (en) 2003-06-16 2013-10-22 Ucb Manufacturing, Inc. Antibodies specific for sclerostin and methods for increasing bone mineralization
US9657095B2 (en) 2003-06-16 2017-05-23 Ucb Pharma S.A. Antibodies specific for sclerostin and methods for increasing bone mineralization
US8992911B2 (en) 2003-06-16 2015-03-31 Ucb Pharma S.A. Antibodies specific for sclerostin and methods for increasing bone mineralization
US7985834B2 (en) 2003-06-16 2011-07-26 Celltech R & D, Inc. Compositions and methods for increasing bone mineralization
US11702468B2 (en) 2003-06-16 2023-07-18 Ucb Pharma, S.A. Antibodies specific for sclerostin and methods for increasing bone mineralization
US9011856B2 (en) 2003-06-16 2015-04-21 Ucb Pharma S.A. Antibodies specific for sclerostin and methods for increasing bone mineralization
US7868134B2 (en) 2003-06-16 2011-01-11 Ucb Manufacturing, Inc. Immunogenic peptides derived from sclerostin
US20110195453A1 (en) * 2003-06-16 2011-08-11 Ucb Manufacturing, Inc. Antibodies Specific for Sclerostin and Methods for Increasing Bone Mineralization
US20110206678A1 (en) * 2003-06-16 2011-08-25 Ucb Manufacturing, Inc. Antibodies Specific for Sclerostin and Methods for Increasing Bone Mineralization
US9492419B2 (en) 2003-09-22 2016-11-15 Enzo Biochem, Inc. Method for treating crohn'S disease or ulcerative colitis by administering a compound which binds LDL-receptor-related protein (LRP) ligand binding domain
US8367822B2 (en) 2003-09-22 2013-02-05 Enzo Therapeutics, Inc. Compositions and methods for bone formation and remodeling
US8969339B2 (en) 2003-09-22 2015-03-03 Enzo Biochem, Inc. Compositions and methods for bone formation and modeling
US9046537B2 (en) 2003-09-22 2015-06-02 Enzo Biochem, Inc. Method for treating inflammation by administering a compound which binds LDL-receptor-related protein (LRP) ligand binding domain
US20050196349A1 (en) * 2003-09-22 2005-09-08 Dianqing Wu Compositions and methods for bone formation and remodeling
US8637506B2 (en) 2003-09-22 2014-01-28 Enzo Biochem, Inc. Compositions and methods for bone formation and remodeling
US20060030523A1 (en) * 2003-09-22 2006-02-09 Wu Dianqing D Sclerostin and the inhibition of WNT signaling and bone formation
US8461155B2 (en) * 2003-09-22 2013-06-11 University Of Connecticut Sclerostin and the inhibition of WNT signaling and bone formation
US20100298308A1 (en) * 2003-09-22 2010-11-25 Wu Dianqing Dan Compositions and methods for bone formation and remodeling
US20060198791A2 (en) * 2003-09-22 2006-09-07 Dianqing Wu Compositions and Methods for Bone Formation and Remodeling
US8343922B2 (en) 2004-05-19 2013-01-01 Enzo Biochem, Inc. Compositions and methods for the stimulation or enhancement of bone formation and the self-renewal of cells
US20050261181A1 (en) * 2004-05-19 2005-11-24 Dianqing Wu Compositions and methods for the stimulation or enhancement of bone formation and the self-renewal of cells
US9052324B2 (en) 2004-05-19 2015-06-09 Enzo Biochem, Inc. Compounds and assays for controlling Wnt activity
WO2006102070A3 (en) * 2005-03-18 2009-04-23 Enzo Biochem Inc Sclerostin and the inhibition of wnt signaling and bone formation
US20060237679A1 (en) * 2005-04-22 2006-10-26 Effebi S.P.A. Valve-actuator connection plate
US20110097342A1 (en) * 2005-05-03 2011-04-28 Amgen Inc. Binding agents
US9296812B2 (en) 2005-05-03 2016-03-29 Amgen Inc. Sclerostin binding antibodies
EP2298800A1 (en) 2005-05-03 2011-03-23 UCB Pharma, S.A. Sclerostin binding antibodies
EP2325200A1 (en) 2005-05-03 2011-05-25 UCB Pharma S.A. Sclerostin binding agents
EP2325199A1 (en) 2005-05-03 2011-05-25 UCB Pharma S.A. Sclerostin binding agents
EP2336163A1 (en) 2005-05-03 2011-06-22 UCB Pharma S.A. Sclerostin binding agents
EP2295448A1 (en) 2005-05-03 2011-03-16 UCB Pharma, S.A. Sclerostin epitopes
WO2006119062A2 (en) 2005-05-03 2006-11-09 Ucb Pharma S.A. Sclerostin epitopes
US8637643B2 (en) 2005-05-03 2014-01-28 Ucb Pharma, S.A. Sclerostin binding antibody
US20070072797A1 (en) * 2005-05-03 2007-03-29 Ucb S.A. Epitopes
EP2345668A1 (en) 2005-05-03 2011-07-20 UCB Pharma, S.A. Sclerostin epitopes
US7872106B2 (en) 2005-05-03 2011-01-18 Amgen Inc. Sclerostin-binding antibodies
EP2264063A1 (en) 2005-05-03 2010-12-22 UCB Pharma, S.A. Sclerostin binding antibody
EP2251351A1 (en) 2005-05-03 2010-11-17 UCB Pharma, S.A. Sclerostin epitopes
US20070110747A1 (en) * 2005-05-03 2007-05-17 Ucb S.A. Binding agents
US8003108B2 (en) * 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
US10273293B2 (en) 2005-05-03 2019-04-30 Amgen Inc. Method for inhibiting bone resorption
US10562964B2 (en) 2005-05-03 2020-02-18 Amgen Inc. Methods for isolating antibodies that bind sclerostin
EP3670528A1 (en) 2005-05-03 2020-06-24 UCB Pharma S.A. Sclerostin binding agents
US20110305702A1 (en) * 2005-05-03 2011-12-15 Amgen Inc. Epitopes
US9353174B2 (en) * 2005-05-03 2016-05-31 Amgen Inc. Epitopes
EP2301961A1 (en) 2005-05-03 2011-03-30 UCB Pharma, S.A. Sclerostin epitopes
US8715663B2 (en) * 2005-05-03 2014-05-06 Amgen Inc. Epitopes
US9089553B2 (en) 2005-05-03 2015-07-28 Amgen Inc. Method for inhibiting bone resorption
US20140248666A1 (en) * 2005-05-03 2014-09-04 Amgen Inc. Epitopes
US8383801B2 (en) 2005-05-03 2013-02-26 Amgen Inc. Polynucleotide encoding a sclerostin-binding antibody
US11939372B2 (en) 2005-05-03 2024-03-26 Amgen Inc. Binding agents
EA037044B1 (en) * 2005-05-03 2021-01-29 Юсб Фарма С.А. Sclerostin binding antibody and use thereof
US20090304713A1 (en) * 2005-05-03 2009-12-10 Amgen Inc. Binding agents
US7592429B2 (en) 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
EP3985015A1 (en) 2005-05-03 2022-04-20 UCB Pharma S.A. Sclerostin binding agents
US8263137B2 (en) 2005-08-04 2012-09-11 Vertical Pharmaceuticals, Inc. Nutritional supplement for women
US20080226746A1 (en) * 2005-08-04 2008-09-18 Vertical Pharmaceuticals, Inc. Nutritional supplement for women
US20070031486A1 (en) * 2005-08-04 2007-02-08 Squashic Steven A Nutritional supplement for use under physiologically stressful conditions
US8263667B2 (en) 2005-08-04 2012-09-11 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US20070031487A1 (en) * 2005-08-04 2007-02-08 Squashic Steven A Nutritional supplement for women
US8202546B2 (en) 2005-08-04 2012-06-19 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US8197854B2 (en) 2005-08-04 2012-06-12 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US7998500B2 (en) 2005-08-04 2011-08-16 Vertical Pharmaceuticals, Inc. Nutritional supplement for women
US7901710B2 (en) 2005-08-04 2011-03-08 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US20110159055A1 (en) * 2005-08-04 2011-06-30 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
US20110151021A1 (en) * 2005-08-04 2011-06-23 Vertical Pharmaceuticals, Inc. Nutritional supplement for use under physiologically stressful conditions
CN103432568A (en) * 2005-11-23 2013-12-11 阿塞勒隆制药公司 Activin-ActRIIa antagonists and uses for promoting bone growth
EP2423226A2 (en) 2006-11-10 2012-02-29 Amgen Inc. Antibody-based diagnostics and therapeutics
US20100036091A1 (en) * 2006-11-10 2010-02-11 Amgen Inc. Antibody-based diagnostics and therapeutics
US20100015665A1 (en) * 2006-11-10 2010-01-21 Ucb Pharma S.A. Antibodies and diagnostics
US20100041599A1 (en) * 2006-11-14 2010-02-18 Dakai Liu Compositions and methods for bone formation, bone remodeling and toxin protection
US20080160060A1 (en) * 2006-12-29 2008-07-03 Osteogenex Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
EP3345607A1 (en) 2006-12-29 2018-07-11 Osteoqc Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
US11891438B2 (en) 2006-12-29 2024-02-06 Ossifi-Mab Llc Methods of altering bone growth by administration of Sost or Wise antagonist or agonist
US8877196B2 (en) 2006-12-29 2014-11-04 Ossifi Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
US11807681B2 (en) 2006-12-29 2023-11-07 Ossifi-Mab Llc Methods of altering bone growth by administration of Sost or Wise antagonist or agonist
US11608373B2 (en) 2006-12-29 2023-03-21 Ossifi-Mab Llc Methods of altering bone growth by administration of Sost or Wise antagonist or agonist
US8178099B2 (en) 2006-12-29 2012-05-15 Osteogenex Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
EP4176884A1 (en) 2006-12-29 2023-05-10 Ossifi-Mab LLC Methods of altering bone growth by administration of sost or wise antagonist or agonist
EP3345607B1 (en) 2006-12-29 2022-10-26 Ossifi-Mab LLC Methods of altering bone growth by administration of sost or wise antagonist or agonist
EP3085386A1 (en) 2007-09-17 2016-10-26 Amgen Inc. Method for inhibiting bone resorption
US8017120B2 (en) 2007-09-17 2011-09-13 Amgen Inc. Method for inhibiting bone resorption
US8329176B2 (en) 2007-09-17 2012-12-11 Amgen Inc. Method for inhibiting bone resorption
WO2009039175A2 (en) 2007-09-17 2009-03-26 Amgen Inc. Method for inhibiting bone resorption
US20090074763A1 (en) * 2007-09-17 2009-03-19 Amgen Inc. Method for inhibiting bone resorption
EP2556841A1 (en) 2007-09-17 2013-02-13 Amgen, Inc Method for inhibiting bone resorption
US8440193B2 (en) 2007-09-17 2013-05-14 Amgen Inc. Method for inhibiting bone resorption
US11091537B2 (en) 2007-09-17 2021-08-17 Amgen Inc. Method for inhibiting bone resorption
EP4335510A2 (en) 2007-09-17 2024-03-13 Amgen Inc. Method for inhibiting bone resorption
EP3725329A1 (en) 2007-09-17 2020-10-21 Amgen, Inc Method for inhibiting bone resorption
EP2586796A1 (en) 2007-10-12 2013-05-01 Novartis AG Compositions and methods for use for antibodies against sclerostin
US20110044978A1 (en) * 2007-12-14 2011-02-24 Amgen Inc. Method for treating bone fracture
EP2628486A2 (en) 2007-12-14 2013-08-21 Amgen, Inc. Method for treating bone fracture with anti-sclerostin antibodies
US20100003324A1 (en) * 2008-07-03 2010-01-07 Osteogenex Inc. Vinpocetine and eburn amonine derivatives for promoting bone growth
US8198292B2 (en) 2008-07-03 2012-06-12 Osteogenex, Inc. Vinpocetine and eburnamonine derivatives for promoting bone growth, treating renal damage and cancer, and devices thereof
US11040102B2 (en) 2010-05-14 2021-06-22 Amgen Inc. High concentration antibody formulations
US9352043B2 (en) 2010-05-14 2016-05-31 Amgen Inc. High concentration antibody formulations
US10064946B2 (en) 2010-05-14 2018-09-04 Amgen Inc. High concentration antibody formulations
US11167011B2 (en) 2010-06-07 2021-11-09 Enzo Biochem, Inc. Methods for treating bone loss using sclerostin peptides
US11639382B2 (en) 2010-06-07 2023-05-02 Enzo Biochem, Inc. Antibodies specific for sulfation sites of Wnt pathway proteins
US11938167B2 (en) 2010-06-07 2024-03-26 Enzo Biochem, Inc. Pharmaceutical compositions and methods targeting Wnt pathway proteins
US11667703B2 (en) 2010-06-07 2023-06-06 Enzo Biochem, Inc. Sulfonated sclerostin, antibodies, epitopes and methods for identification and use therefor
US9403882B2 (en) 2010-06-07 2016-08-02 Joshua Rabbani Sulfation of Wnt pathway proteins
WO2011156252A2 (en) 2010-06-07 2011-12-15 Enzo Biochem. Inc. Sulfation of wnt pathway proteins
US9493541B2 (en) 2010-06-07 2016-11-15 Joshua Rabbani Antibodies specific for sulfated sclerostin
US10899827B2 (en) 2010-06-07 2021-01-26 Enzo Biochem, Inc. Antibodies specific for sulfation sites of sclerostin
US9617323B2 (en) 2010-06-07 2017-04-11 Joshua Rabbani Sulfonated sclerostin, antibodies, epitopes and methods for identification and use therefor
US10626170B2 (en) 2010-06-07 2020-04-21 Enzo Biochem, Inc. Sulfonated sclerostin, antibodies, epitopes and methods for identification and use therefor
US10328120B2 (en) 2010-06-07 2019-06-25 Enzo Biochem, Inc. Pharmaceutical compositions targeting WNT pathway proteins
US9133272B2 (en) 2011-03-01 2015-09-15 Amgen Inc. Bispecific binding agents
US9617333B2 (en) 2011-03-25 2017-04-11 Amgen Inc. Sclerostin antibody crystals and formulations thereof
US9920114B2 (en) 2011-03-25 2018-03-20 Amgen Inc. Antibody formulations
US9145457B2 (en) 2011-03-25 2015-09-29 Amgen Inc. Sclerostin antibody crystals and formulations thereof
WO2012145417A1 (en) 2011-04-19 2012-10-26 Amgen Inc. Method for treating osteoporosis
EP3404041A1 (en) 2011-04-19 2018-11-21 Amgen, Inc Method for treating osteoporosis
WO2013003178A1 (en) 2011-06-28 2013-01-03 Enzo Biochem, Inc. Sulfation of wnt pathway proteins
EP3428183A1 (en) 2011-06-28 2019-01-16 Enzo Biochem, Inc. Sulfation of wnt pathway proteins
US10538584B2 (en) 2011-08-04 2020-01-21 Amgen Inc. Methods for treating bone gap defects
WO2013019954A1 (en) 2011-08-04 2013-02-07 Amgen Inc. Method for treating bone gap defects
EP2753626A1 (en) 2011-09-02 2014-07-16 The Regents of The University of California Llp2a-bisphosphonate conjugates for osteoporosis treatment
EP3453711A1 (en) 2011-09-02 2019-03-13 The Regents of the University of California Llp2a- bisphosphonate conjugates for osteoporosis treatment
WO2013032527A1 (en) 2011-09-02 2013-03-07 The Regents Of The University Of California Llp2a-bisphosphonate conjugates for osteoporosis treatment
US9657090B2 (en) 2011-12-28 2017-05-23 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
WO2013101451A1 (en) 2011-12-28 2013-07-04 Amgen Inc. Method of treating alvelor bone loss through the use of anti-sclerostin antibodies
EP3712168A2 (en) 2011-12-28 2020-09-23 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
US9913900B2 (en) 2011-12-28 2018-03-13 Amgen Inc. Method of treating alvelor bone loss through the use of anti-sclerostin antibodies
US10799583B2 (en) 2012-07-05 2020-10-13 Ucb Pharma, S.A. Treatment for bone diseases
US11896667B2 (en) 2012-07-05 2024-02-13 Ucb Pharma S.A. Treatment for bone diseases
WO2014006100A1 (en) 2012-07-05 2014-01-09 Ucb Pharma S.A. Treatment for bone diseases
EP3626267A1 (en) 2012-07-05 2020-03-25 UCB Pharma, S.A. Treatment for bone diseases
US9925260B2 (en) 2012-07-05 2018-03-27 Ucb Pharma S.A. Treatment for bone diseases
US11466078B2 (en) 2012-11-21 2022-10-11 Amgen Inc. Heterodimeric immunoglobulins
EP3587451A1 (en) 2012-11-21 2020-01-01 Amgen Inc. Heterodimeric immunoglobulins
US10233237B2 (en) 2012-11-21 2019-03-19 Amgen Inc. Heterodimeric immunoglobulins
US9822173B2 (en) 2012-11-21 2017-11-21 Amgen Inc. Heterodimeric immunoglobulins
WO2014144817A2 (en) 2013-03-15 2014-09-18 Amgen Inc. Inhibitory polypeptides specific to wnt inhibitors
US11851483B2 (en) 2014-12-12 2023-12-26 Amgen Inc. Anti-sclerostin antibodies and their use to treat bone disorders as part of a regimen
US11576970B2 (en) 2016-03-10 2023-02-14 UCB Biopharma SRL Pharmaceutical formulations
WO2018031454A1 (en) 2016-08-08 2018-02-15 Amgen Inc. Method of improving connective tissue attachment using anti-sclerostin antibodies
WO2018115879A1 (en) 2016-12-21 2018-06-28 Mereo Biopharma 3 Limited Use of anti-sclerostin antibodies in the treatment of osteogenesis imperfecta
EP3868780A1 (en) 2016-12-21 2021-08-25 Mereo Biopharma 3 Limited Use of anti-sclerostin antibodies in the treatment of osteogenesis imperfecta
WO2018115880A1 (en) 2016-12-21 2018-06-28 Mereo Biopharma 3 Limited Use of anti-sclerostin antibodies in the treatment of osteogenesis imperfecta
WO2019191534A1 (en) 2018-03-30 2019-10-03 Amgen Inc. C-terminal antibody variants
US11858983B2 (en) 2018-03-30 2024-01-02 Amgen Inc. C-terminal anti-sclerostin antibody variants
US11466079B2 (en) 2018-03-30 2022-10-11 Amgen Inc. C-terminal antibody variants
WO2020033788A1 (en) 2018-08-10 2020-02-13 Amgen Inc. Method of preparing an antibody pharmaceutical formulation
WO2021030179A1 (en) 2019-08-12 2021-02-18 Amgen Inc. Anti-sclerostin antibody formulations

Also Published As

Publication number Publication date
PL1638999T3 (en) 2014-04-30
EP2338906A1 (en) 2011-06-29
IL222756A0 (en) 2012-12-31
CY1114701T1 (en) 2016-10-05
KR20060064568A (en) 2006-06-13
MEP3108A (en) 2010-02-10
CN1835968A (en) 2006-09-20
CA2529578A1 (en) 2005-01-13
SG185824A1 (en) 2012-12-28
AU2004253870B2 (en) 2011-11-10
PT1638999E (en) 2013-09-03
CA2529578C (en) 2014-11-18
SI1638999T1 (en) 2013-12-31
RS52769B (en) 2013-10-31
ME00023B (en) 2010-06-10
ZA200600384B (en) 2008-10-29
US7985834B2 (en) 2011-07-26
JP2011246482A (en) 2011-12-08
KR101221147B1 (en) 2013-01-10
EA014525B1 (en) 2010-12-30
EA201001528A1 (en) 2011-06-30
IL172599A0 (en) 2006-04-10
US20060233801A1 (en) 2006-10-19
ES2428767T3 (en) 2013-11-11
HK1088907A1 (en) 2006-11-17
JP2008505843A (en) 2008-02-28
RS20050932A (en) 2008-11-28
BRPI0411535A (en) 2006-08-01
EA200600037A1 (en) 2006-06-30
AU2004253870A1 (en) 2005-01-13
EP1638999A2 (en) 2006-03-29
IL172599A (en) 2016-05-31
NZ544617A (en) 2009-11-27
US7578999B2 (en) 2009-08-25
DK1638999T3 (en) 2013-10-14
NO20060235L (en) 2006-03-14
EP2341071A1 (en) 2011-07-06
CN1835968B (en) 2013-01-30
US20100151524A1 (en) 2010-06-17
HRP20131014T1 (en) 2013-12-20
WO2005003158A3 (en) 2005-05-26
EA022991B1 (en) 2016-04-29
JP4818107B2 (en) 2011-11-16
CN103319596A (en) 2013-09-25
EP1638999B1 (en) 2013-07-24
WO2005003158A2 (en) 2005-01-13

Similar Documents

Publication Publication Date Title
KR101221147B1 (en) Compositions and methods for increasing bone mineralization
US6489445B1 (en) Polypeptides associated with alterations in bone density
KR101210187B1 (en) Antibodies specific for sclerostin and methods for increasing bone mineralization
AU2003271274B2 (en) Compositions and methods for increasing bone mineralization

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLTECH R&D, INC., WASHINGTON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRUNKOW, MARY E.;GALAS, DAVID J.;KOVACEVICH, BRIAN;AND OTHERS;REEL/FRAME:014536/0127;SIGNING DATES FROM 20030814 TO 20030819

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: UCB MANUFACTURING, INC., NEW YORK

Free format text: MERGER;ASSIGNOR:DARWIN MOLECULAR CORPORATION;REEL/FRAME:025051/0957

Effective date: 20051228

Owner name: DARWIN MOLECULAR CORPORATION, WASHINGTON

Free format text: DISSOLUTION OF CELLTECH R&D, INC. AND SUBSEQUENT DISTRIBUTION OF ASSETS INTO DARWIN MOLECULAR CORPORATION;ASSIGNOR:CELLTECH R&D, INC.;REEL/FRAME:025051/0949

Effective date: 20041228

AS Assignment

Owner name: UCB PHARMA S.A., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UCB MANUFACTURING, INC.;REEL/FRAME:033515/0904

Effective date: 20140805