WO1994010203A2 - Op-3-induced morphogenesis - Google Patents

Op-3-induced morphogenesis Download PDF

Info

Publication number
WO1994010203A2
WO1994010203A2 PCT/US1993/010520 US9310520W WO9410203A2 WO 1994010203 A2 WO1994010203 A2 WO 1994010203A2 US 9310520 W US9310520 W US 9310520W WO 9410203 A2 WO9410203 A2 WO 9410203A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
seq
species
tissue
leu
Prior art date
Application number
PCT/US1993/010520
Other languages
French (fr)
Other versions
WO1994010203A3 (en
Inventor
Hermann Oppermann
Engin Ozkaynak
Thangavel Kuberasampath
David C. Rueger
Roy H. L. Pang
Charles M. Cohen
Original Assignee
Creative Biomolecules, 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
Application filed by Creative Biomolecules, Inc. filed Critical Creative Biomolecules, Inc.
Priority to JP6511392A priority Critical patent/JPH08503198A/en
Priority to AU55900/94A priority patent/AU681362B2/en
Priority to EP94901244A priority patent/EP0672064A1/en
Publication of WO1994010203A2 publication Critical patent/WO1994010203A2/en
Publication of WO1994010203A3 publication Critical patent/WO1994010203A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K6/00Preparations for dentistry
    • A61K6/50Preparations specially adapted for dental root treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • A61L27/227Other specific proteins or polypeptides not covered by A61L27/222, A61L27/225 or A61L27/24
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/22Polypeptides or derivatives thereof, e.g. degradation products
    • A61L27/24Collagen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2310/00Prostheses classified in A61F2/28 or A61F2/30 - A61F2/44 being constructed from or coated with a particular material
    • A61F2310/00005The prosthesis being constructed from a particular material
    • A61F2310/00365Proteins; Polypeptides; Degradation products thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • This invention relates generally to the field of tissue morphogenesis and more particularly to a novel protein that induces tissue morphogenesis in mammals.
  • tissues can be divided into three broad categories: (1) tissues with static cell populations such as nerve and skeletal muscle where there is no cell division and most of the cells formed during early development persist throughout adult life; (2) tissues containing conditionally renewing populations such as liver where there is generally little cell division but, in response to an appropriate stimulus, cells can divide to produce daughters of the same differentially defined type; and (3) tissues with permanently renewing populations including blood, testes and stratified squamous epithelia which are characterized by rapid and
  • the terminally differentiated cells have a relatively short life span and are replaced through proliferation of a distinct subpopulation of cells, known as stem or progenitor cells.
  • tissue degenerative diseases including arthritis, emphysema, osteoporosis, cardiomyopathy, cirrhosis, and
  • TGF transforming growth factor
  • morphogenically active C-terminal domains including a conserved six or seven cysteine skeleton, are capable of inducing tissue-specific morphogenesis in a variety of organs and tissues, including bone, cartilage, liver, dentin, periodontal ligament, cementum, nerve tissue and the epithelial mucosa of the
  • the proteins apparently bind to surface receptors or otherwise contact and interact with progenitor cells, predisposing or stimulating the cells to proliferate and differentiate in a
  • the morphogens are capable of inducing the developmental cascade of cellular and molecular events that culminate in the formation of new organ-specific tissue, including any vascularization, connective tissue formation, and nerve ennervation as required by the naturally occurring tissue.
  • proteins useful in tissue morphogenesis are proteins originally identified as bone inductive proteins, such as the OP-1, (also referred to in related applications as “OP1”), OP-2 (also referred to in related applications as “OP2”), and the CBMP2 proteins, as well as amino acid sequence-related proteins such as BMP5, BMP6 and its murine homolog, Vgr-1, DPP and 60A (from Drosophila), Vgl (from
  • TGF- ⁇ superfamily members comprise a distinct subfamily of proteins different from other members of the TGF- ⁇ superfamily in that the family of morphogenic proteins are able to induce the full cascade of events that result in tissue morphogenesis, including stimulating cell proliferation and cell differentiation of progenitor cells, and supporting the growth and maintenance of differentiated cells.
  • the morphogenic proteins apparently can act as endocrine, paracrine or autocrine factors.
  • the endogenous morphogens may be synthesized by the cells on which they act, by neighboring cells, or by cells of a distant tissue, the secreted protein being
  • fibrotic (scar) tissue as, for example, TGF- ⁇ does.
  • the morphogens are synthesized in the cell as a precursor molecule approximately three times larger than the mature protein that is processed to yield mature disulfide-linked dimers comprising the
  • the proteins are inactive when reduced e.g., in monomeric form, but are active as oxidized homodimeric species as well as when oxidized in combination with other morphogens under conditions to produce heterodimers.
  • the proteins useful in tissue morphogenesis typically require a suitable environment enabling cells to migrate, proliferate and differentiate in a tissue-specific manner into, e.g., cartilage-producing
  • chondroblasts chondroblasts, bone-producing osteoblasts, hemopoietic cells, or liver cells, depending on the nature of the local environment.
  • OP-3 novel purified morphogenic protein
  • properties of OP-3 include the ability to induce proliferation and differentiation of progenitor cells, and the ability to support and maintain the
  • Still another object is to provide tissue-specific acellular matrices that may be used in
  • utilizing OP-3 to induce the developmental cascade of tissue morphogenesis in a mammal Specifically, methods are provided for utilizing OP-3 to induce the proliferation of uncommitted progenitor cells, to induce the differentiation of these stimulated
  • OP-3 can be utilized to initiate and maintain the developmental cascade of tissue morphogenesis in an appropriate, morphogenically permissive environment.
  • useful OP-3 morphogens include proteins encoded by the DNA sequence provided in Seq. ID No. 1 ("mOP-3") and allelic and species variants thereof, as well as other naturally-occurring and biosynthetic amino acid sequence variants, including chimeric proteins, that are morphogenically active as defined herein.
  • “Morphogenically active fragment” is understood to include all proteins and protein
  • a morphogen is a dimeric protein comprising a pair of polypeptide chains, wherein each polypeptide chain comprises at least the C-terminal six cysteine skeleton defined by residues 303 to 399 of Seq. ID No. 1 (or residues 335-431 of OP1, Seq. ID no. 3), including functionally equivalent arrangements of these cysteines (e.g., amino acid insertions or deletions which alter the linear arrangement of the cysteines in the sequence but not their relationship in the folded structure), such that, when the polypeptide chains are folded, the dimeric protein species comprising the pair of
  • polypeptide chains has the appropriate three-dimensional structure, including the appropriate intra- or inter-chain disulfide bonds such that the protein is capable of acting as a morphogen as defined herein.
  • the morphogens generally are capable of all of the following biological functions in a
  • morphogenically permissive environment stimulating proliferation of progenitor cells; stimulating the differentiation of progenitor cells; stimulating the proliferation of differentiated cells; and supporting the growth and maintenance of differentiated cells.
  • the morphogens of this invention comprise a morphogenically active dimeric species comprising a pair of polypeptide chains, wherein at least one of the polypeptide chains comprises the amino acid sequence defined by residues 303 to 399 of Seq. ID No. 1 including allelic, species and other amino acid sequence variants thereof.
  • at least one polypeptide chain comprises the sequence defined by residues 298-399, residues 261-399 or residues 264-399 of Seq. ID No. 1.
  • the amino acid sequence of both polypeptide chains may be defined by part or all of the amino acid sequence of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof, including
  • polypeptide chain preferably comprises at least the sequence defining the C-terminal six cysteine skeleton of any of the other known
  • morphogen family members including OP-1, OP-2, CBMP2A, CBMP2B, BMP3, BMP5, BMP6, Vgr-1, Vgl, 60A, DPP and GDF-1, described, for example, in US92/07432 (WO93/05751), including allelic, species and other amino acid
  • sequence variants thereof including chimeric variants.
  • Other useful sequences include biosynthetic constructs, such as are described in U.S. Pat. No. 5,011,691.
  • morphogens of this invention comprise morphogenically active proteins encoded by part or all of the genetic sequence listed in Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
  • the invention comprises morphogens encoded by nucleic acids that hybridize to part or all of the pro region of the OP-3 protein, bases 120 to 848 of Seq ID No. 1, under stringent hybridization conditions.
  • stringent hybridization conditions are defined as hybridization in 40% formamide, 5 ⁇ SSPE, 5 ⁇ Denhardt's Solution, and 0.1% SDS at 37°C
  • morphogenically active fragments of OP-3 are useful in the replacement of diseased or damaged tissue in a mammal, including, but not limited to, damaged lung tissue resulting from emphysema; cirrhotic tissue, including cirrhotic kidney or liver tissue; damaged heart or blood vessel tissue, as may result from cardiomyopathies and/or
  • damaged cartilage and bone tissue as may result from metabolic bone diseases and other bone remodeling disorders
  • damaged dentin, periodontal and/or cementum tissue as may result from disease or mechanical injury
  • in the replacement of damaged tissue as a result of inflammation and/or chronic inflammatory disease.
  • fragments of OP-3 are provided to a tissue-specific locus in vivo, to induce the developmental cascade of tissue morphogenesis at that site.
  • Cells stimulated ex vivo by contact with OP-3 also may be provided to the tissue locus.
  • the existing tissue provides the necessary matrix requirements, providing a suitable substratum or scaffold for the proliferating and differentiating cells in a morphogenically
  • proteins or stimulated cells also may be combined with a formulated matrix and implanted as a device at a locus in vivo.
  • formulated matrix should be a biocompatible, preferably biodegradable acellular matrix having the
  • the matrix preferably also is tissue-specific.
  • the members of the morphogen protein family also can control the body's cellular and humoral inflammatory response to a foreign object or an initial tissue injury.
  • the loss of tissue function results from the tissue destructive effects and the subsequent formation of scar tissue associated with the body's immune/inflammatory response to an initial or repeated injury to the tissue.
  • the degree of scar tissue formation generally depends on the regenerative properties of the injured tissue, and on the degree and type of tissue damage.
  • morphogenically active fragments of OP-3 may be used to prevent or to substantially inhibit the formation of scar tissue, including alleviating immune response-mediated tissue damage, by providing OP-3 or cells stimulated by exposure to OP-3 protein, to a newly injured tissue locus.
  • the OP-3 protein also may be provided as a prophylactic, provided to a site in anticipation of tissue injury, such as part of a surgical or other clinical procedure likely to produce tissue damage, and to induce an inflammatory/immune response.
  • OP-3 may be used as part of a transplant procedure, to enhance the tissue viability of the organ and/or tissue to be transplanted.
  • the morphogen may be provided to the organ and/or tissue to be transplanted prior to harvest, during its transport, and/or during
  • OP-3 also may be used to increase or regenerate a mesenchymal progenitor or stem cell population in vitro or in a mammal.
  • progenitor cells may be isolated from an individual's bone marrow, stimulated ex vivo with morphogenic OP-3 for a time and at a concentration sufficient to induce the cells to
  • progenitor cells that may be suitable include biocompatible cells obtained from a cultured cell line, stimulated in culture, and subsequently provided to the body.
  • OP-3 may be provided by systemic (e.g., oral or parenteral) administration, or it may be injected or otherwise provided to a progenitor cell population in an
  • a morphogenically active fragment of OP-3 may be provided to the cells in vivo, e.g., by systemic injection, to induce mitogenic activity.
  • a particular population of hemopoietic stem cells may be increased by exposure to OP-3, for example by perfusing (plasmaphoresing) an individual's blood to extract the cells of interest, stimulating these cells ex vivo, and returning the stimulated cells to the blood.
  • the morphogens of this invention also can inhibit proliferation of epithelial cell populations.
  • the ability to inhibit epithelial cell proliferation may be exploited to reduce tissue damage associated with psoriasis and dermatitis, and other inflammatory skin diseases, as well as ulcerative diseases of the
  • Morphogens may be used to particular advantage as a cytoprotective agent in clinical
  • chemotherapies that typically induce oral mucositis, hair loss and/or skin disorders.
  • morphogenic OP-3 may be used to support the growth and maintenance of differentiated cells, inducing existing
  • differentiated cells to continue expressing their phenotype. It is anticipated that this activity will be particularly useful in the treatment of tissue disorders where loss of function is caused by reduced or lost metabolic function in which cells become senescent or quiescent, such as may occur in aging cells and/or may be manifested in osteoporosis and a number of nerve degenerative diseases, including
  • OP-3 directly to the cells to be treated, or providing it systemically, as by oral or parenteral administration, can stimulate these cells to continue expressing their phenotype, thereby significantly reversing the effects of the dysfunction.
  • a morphogenically active fragment of OP-3 also may be used in gene therapy protocols to stimulate the growth of quiescent cells, thereby potentially enhancing the ability of these cells to incorporate exogenous DNA.
  • morphogenically active fragment of OP-3 also may be used to induce "redifferentiation" of cells that have strayed from their differentiation pathway, such as can occur during tumorgenesis. It is anticipated that this activity will be particularly useful in treatments to reduce or substantially inhibit the growth of
  • a morphogenically active OP-3 fragment may be provided to the cells directly or systemically, stimulating these cells to revert back to a morphology and phenotype characteristic of untransformed cells.
  • OP-3 may be used to stimulate cell adhesion molecule (CAM) expression levels in a cell.
  • CAMs are molecules defined as carrying out cell-cell interactions
  • CAMs are believed to play a fundamental regulatory role in tissue
  • tissue boundary formation embryonic induction and migration, and tissue
  • Altered CAM levels have been implicated in a number of tissue disorders, including congenital defects, neoplasias, and
  • N-CAM expression is associated with normal neuronal cell development and differentiation, including retinal formation, synaptogenesis, and nervemuscle tissue adhesion. Inhibition of one or more of the N-CAM isoforms is known to prevent proper tissue development. Altered N-CAM expression levels also are associated with neoplasias, including neuroblastomas (see infra), as well as with a number of neuropathies, including normal pressure hydrocephalous and type II schizophrenia.
  • Application of the morphogen directly to the cells to be treated, or providing the morphogen to the mammal systemically, for example, parenterally, or indirectly by oral administration, may be used to induce cellular expression of one or more CAMs, particularly N-CAMs and L1.
  • CAMs also have been postulated as part of a
  • morphogens described herein may act as inducers of this pathway.
  • inventions may be derived from organ-specific tissue, or they may be formulated synthetically. In one
  • OP-3 (or a collection of progenitor cells stimulated by OP-3) is provided at a tissue-specific locus, e.g., by systemic
  • a formulated matrix may be provided externally together with the stimulated progenitor cells or morphogenically active OP-3
  • the matrix should be a biocompatible, suitably modified acellular matrix having dimensions such that it allows the differentiation and proliferation of migratory
  • progenitor cells and is capable of providing a
  • the matrix also preferably allows cellular attachment and is biodegradable. Where the necessary tissue-directing signals can not be provided endogenously, the matrix preferably also is tissue-specific.
  • Formulated matrices may be generated from
  • the matrix may be formulated synthetically using a biocompatible, preferably in vivo biodegradable, structural molecule, and may be formulated with suitable tissue-specific cell attachment factors.
  • the molecule may be a
  • polyglycolic acid and copolymers thereof.
  • preferred structural polymers comprise tissue-specific collagens.
  • preferred cell attachment factors include glycosaminoglycans and proteoglycans.
  • the matrix further may be treated with an agent or agents to increase the number of pores and micropits on its surfaces, so as to enhance the influx, proliferation and differentiation of migratory progenitor cells from the body of the mammal.
  • the invention thus relates to compositions and methods for the use of morphogenically active fragments of OP-3, a novel species variant of the generic family of morphogens disclosed in USSN 667,274 and USSN
  • Morphogenically active OP-3 and protein fragments can be isolated from
  • Active OP-3 useful in the compositions and methods of this invention may include forms having varying glycosylation patterns, varying N-termini and active truncated forms, e.g., produced by recombinant DNA techniques.
  • Active OP-3 proteins also include chimeric constructs as described below, comprising both an OP-3 active domain and a non-OP-3 sequence as, for example, the pro domain and/or the N-terminal region of the mature protein.
  • OP-3 protein can be expressed from intact or truncated cDNA or from synthetic DNAs in procaryotic or eucaryotic host cells, and purified, cleaved, refolded, and dimerized to form
  • Useful host cells include procaryotes, including E. coli, and eucaryotic cells, including mammalian cells, such as CHO, COS, melanoma or BSC cells, or the insect/baculovirus system.
  • procaryotes including E. coli
  • eucaryotic cells including mammalian cells, such as CHO, COS, melanoma or BSC cells, or the insect/baculovirus system.
  • OP-3 capable of inducing tissue-specific cell differentiation and tissue morphogenesis in a variety of mammals, including humans.
  • Figure 1 is a nucleotide sequence comparison of the mouse cDNA sequence of OP-2 and OP-3. Exon boundaries are indicated by bars beneath the sequence; diamonds indicate nucleotide differences within exons 2 and 3; and
  • Figure 2 is an immunoblot comparing mammalian cell expression of an OP1/OP3 chimeric protein construct (lanes 4-8) with that of authentic, recombinant OP1 ( lane 1). Detailed Description
  • the invention provides a novel genetic sequence, mOP-3, encoding a novel protein, OP-3, having
  • the invention provides methods for identifying and isolating the gene from other species. As will be appreciated by those skilled in the art, the methods described herein also may be used to isolate the OP-3 gene from other libraries, including genomic libraries. The invention also provides means for producing the OP-3 genetic sequence and the encoded protein. The invention further provides methods and compositions for inducing the developmental cascade of tissue
  • morphogenesis in a mammal utilizing morphogenically active fragments of OP-3.
  • the methods and compositions provided herein may be utilized in a range of
  • the morphogenic OP-3 species of the invention are novel members of the family of morphogens disclosed in
  • OP-3 may be isolated from natural sources or constructed biosynthetically utilizing conventional recombinant DNA technology or constructed synthetically using standard chemical techniques.
  • Morphogenically active fragments of OP-3 are useful for initiating and maintaining the tissue-specific developmental cascade in a variety of tissues, including, but not limited to, bone, cartilage, dentin, neural tissue, liver, periodontal ligament, cementum, lung, heart, kidney and numerous tissues of the
  • OP-3 can induce the proliferation and differentiation of these progenitor cells.
  • OP-3 is capable of reproducing the cascade of cellular and molecular events that occur during embryonic development to yield functional tissue.
  • the protein can induce the de novo formation of cartilage and endochondral bone, including inducing the proliferation and differentiation of progenitor cells into chondrocytes and osteoblasts, inducing appropriate mineralization and bone remodeling,
  • nucleic acid and amino acid sequences which describe OP-3 proteins useful in the compositions and methods of this invention, including a description of how to make them, and methods and means for their therapeutic administration. Also provided are numerous,
  • Example 9 a method for screening compounds to identify morphogen stimulating agents capable of stimulating endogenous OP-3 expression and/or secretion.
  • OP-3 stimulating agents then may be used in any of the therapeutic applications described herein in place of, or in addition to, OP-3 protein administration.
  • a protein is morphogenic if it is capable of inducing the developmental cascade of cellular and molecular events that culminate in the formation of new, organ-specific tissue and comprises at least the conserved C-terminal six cysteine skeleton or its functional equivalent (see supra).
  • the morphogens generally are capable of all of the following biological functions in a
  • morphogenically permissive environment stimulating proliferation of progenitor cells; stimulating the differentiation of progenitor cells; stimulating the proliferation of differentiated cells; and supporting the growth and maintenance of differentiated cells.
  • the morphogens may be purified from naturally-sourced material or recombinantly produced from procaryotic or eucaryotic host cells, preferably as described therein.
  • novel morphogenic sequences may be identified following the procedures disclosed therein.
  • Particularly useful morphogens identified to date include OP-1, OP-2, CBMP2A and CBMP2B (the
  • BMP2A and BMP2B morphogenically active domains of proteins referred to in the art as BMP2A and BMP2B, or BMP2 and BMP4, respectively
  • BMP3, BMP5, BMP6, Vgr-1, GDF-1, Vgl, DPP and 60A including their allelic and species variants, as well as other amino acid sequence variants,
  • Morphogenically active biosynthetic constructs such as those disclosed in U.S. Pat. No. 5,011,691, (e.g., COP-1, COP-3, COP-4, COP-5, COP-7, and COP-16) also are envisioned to be useful.
  • the novel morphogen OP-3 and its genetic sequence now have been identified.
  • the OP-3 proteins useful in the invention include any morphogenically active fragment of the OP-3 amino acid sequence present in Seq. ID No. 1, or allelic, species or other amino acid sequence variants thereof.
  • the morphogenically active fragment of OP-3 also may include any morphogenically active protein encoded by part or all of the nucleic acid sequence presented in Seq. ID No. 1.
  • morphogenic protein also may comprise a protein encoded by part or all of a nucleic acid which hybridizes to at least part of the nucleic acid sequence encoding the "pro" region of the OP-3 protein, e.g., bases 120-848 of Seq. ID No. 1, under stringent conditions.
  • the mOP-3 gene encodes a protein (“mOP-3") first expressed as an immature translation product that is 399 amino acids in length. This precursor form, referred to herein as the "prepro" form, (Seq. ID.
  • amino acid residues 1-399 includes an
  • N-terminal signal peptide sequence typically less than about 20 residues, followed by a "pro" domain that is cleaved to yield the mature sequence.
  • the "pro” form of the protein includes the pro domain and the mature domain, and forms a soluble species that appears to be the primary form secreted from cultured mammalian cells.
  • the signal peptide anticipated to include residues 1-17 for mOP3, is cleaved rapidly upon
  • morphogenically active OP-3 protein comprises a processed sequence, including fragments thereof, appropriately dimerized and disulfide bonded.
  • the protein preferably comprises both the mature domain, or an active portion thereof, and part or all of the pro domain.
  • the pro domain may be cleaved at residues 260-263 to yield a shorter sequence 135 amino acids in length (Seq. ID No. 1, amino acid residues 264-399).
  • All morphogens including OP-1, OP-2 and the OP-3 proteins disclosed herein, comprise at least a conserved six cysteine skeleton in the amino acid sequence C-terminal domain and, preferably, a conserved seven cysteine skeleton (see, for example, US92/01968 (W092/15323).
  • the conserved six cysteine skeleton in mOP-3 (Seq. ID No. 1) is defined by amino acid residues 303-399; the conserved seven cysteine skeleton is defined by amino acid residues 298-399.
  • the OP-3 proteins like the OP-2 proteins, also has one
  • cysteine residue 338 of Seq. ID No. 1 in the conserved C-terminal domain.
  • the mature sequence of OP-3 shares significant amino acid sequence homology with the morphogens identified to date. Specifically, the seven cysteine fragment shows greater than 79% amino acid identity with the corresponding mOP-2 and hOP-2 sequences, and greater than 66% identity with the corresponding OP-1 sequences.
  • OP-3 has an eighth cysteine within the seven cysteine domain (e.g., at position 338 of Seq. ID No. 1).
  • OP-3 is unique among the morphogens identified to date in that the residue at position 9 in the conserved seven cysteine domain (e.g., residue 315 of Seq. ID No. 1) is a serine, whereas other morphogens typically have a tryptophan at this location (see Table I below, and Table II in
  • amino acid sequence homology is understood to mean amino acid sequence similarity, and homologous sequences share identical or similar amino acids, where similar amino acids are conserved amino acids as defined by Dayoff et al.. Atlas of Protein Sequence and Structure; vol.5, Suppl.3, pp.345-362 (M.O. Dayoff, ed., Nat'l BioMed. Research Fdn.,
  • a candidate sequence sharing 70% amino acid homology with a reference sequence requires that, following alignment of the candidate sequence with the reference sequence, 70% of the amino acids in the candidate sequence are identical to the corresponding amino acid in the reference sequence, or constitute a conserved amino acid change thereto.
  • Amino acid sequence identity is understood to require identical amino acids between two aligned sequences.
  • a candidate sequence sharing 60% amino acid identity with a reference sequence requires that, following alignment of the candidate sequence with the reference sequence , 60% of the amino acids in the candidate sequence are identical to the
  • useful OP-3 variants include, but are not limited to, amino acid sequences derived from Seq. ID No. 1 and wherein the cysteine at position 338 is replaced with another amino acid, preferably a
  • Still other useful OP-3 variants include proteins wherein the serine at position 315 is replaced with another amino acid, preferably a tryptophan and conservative substitutions thereof.
  • Generic Sequence 7 (Seq. ID No. 12) and Generic Sequence 8 (Seq. ID No. 13) disclosed below,
  • Sequence 8 providing an appropriate cysteine skeleton where inter- or intramolecular disulfide bonds can form, and containing certain critical amino acids which influence the tertiary structure of the proteins.
  • each Xaa is independently selected from a group of one or more specified amino acids defined as
  • Generic Sequence 8 (Seq. ID No. 13) includes all of Generic Sequence 7 and in addition includes the following sequence at its N-terminus: Cys Xaa Xaa Xaa Xaa Xaa
  • each "Xaa” in Generic Seq. 8 is a specified amino acid defined as for Generic Seq. 7, with the distinction that each residue number described for Generic Sequence 7 is shifted by five in Generic Seq. 8.
  • Table I compares the C-terminal amino acid sequences defining the seven cysteine skeleton of human OP-1, mouse OP-1, human OP-2, mouse OP-2, and mouse OP-3 (mOP-3, Seq. ID No. 1).
  • sequences are aligned essentially following the method of Needleman et al. (1970) J. Mol. Biol., 48: 443-453, calculated using the Align Program
  • the morphogens described herein may be provided to an individual by any suitable means, preferably
  • morphogen is to be provided directly (e.g., locally, as by injection, to a desired tissue site), or parenterally, such as by intravenous, subcutaneous, intramuscular, intraorbital, ophthalmic,
  • the morphogen preferably comprises part of an aqueous solution.
  • the solution is
  • the aqueous medium for the morphogen thus may comprise normal physiologic saline (0.9% NaCl, 0.15M), pH 7-7.4.
  • the aqueous solution containing the morphogen can be made, for example, by dissolving the protein in 50% ethanol, or acetonitrile containing 0.1% trifluoroacetic acid (TFA) or 0.1% HCl, or equivalent solvents.
  • TFA trifluoroacetic acid
  • HSA human serum albumin
  • the resultant solution preferably is vortexed extensively.
  • a given morphogen may be made more soluble by association with a suitable molecule.
  • association of the mature dimer with the pro domain of the morphogen increases solubility of the protein significantly.
  • the pro form of OP-3 comprises a species that is soluble in
  • the endogenous protein is thought to be transported (e.g., secreted and circulated) to particular tissues in this form.
  • This soluble form of the protein may be obtained from the culture medium of morphogen-secreting
  • a soluble species may be formulated by complexing the mature dimer (or an active fragment thereof) with part or all of a pro domain.
  • Another molecule capable of enhancing the mature dimer (or an active fragment thereof) may be formulated by complexing the mature dimer (or an active fragment thereof) with part or all of a pro domain.
  • casein For example, addition of 0.2% casein increases solubility of the mature active form of OP-1 by 80%.
  • Other components found in milk and/or various serum proteins also may be useful.
  • administration may be prepared by any of the methods well known in the pharmaceutical art, described, for example, in Remington's Pharmaceutical Sciences,
  • Formulations may include, for example, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin,
  • Formulations for direct administration may include glycerol and other compositions of high viscosity.
  • Biocompatible, preferably bioresorbable polymers including, for example, hyaluronic acid, collagen, tricalcium phosphate, polybutyrate, polylactide, polyglycolide and lactide/glycolide copolymers, may be useful excipients to control the release of the
  • Formulations for inhalation administration may contain as excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally.
  • the morphogens described herein may be administered orally.
  • proteins as therapeutics generally is not practiced as most proteins readily are degraded by digestive enzymes and acids in the mammalian digestive system before they can be absorbed into the bloodstream.
  • the morphogens described herein typically are acid-stable and protease-resistant (see, for example, U.S. Pat. No. 4,968,590.)
  • at least one morphogen, OP-1 has been identified in bovine mammary gland extract, colostrum and milk, as well as saliva.
  • the OP-1 purified from mammary gland extract is
  • this protein induces endochondral bone formation in mammals when implanted subcutaneously in association with a suitable matrix material, using a standard in vivo bone assay, such as is disclosed in U.S. Pat. No. 4,968,590.
  • endogenous morphogen also is detected in human serum.
  • the compounds provided herein also may be associated with molecules capable of enhancing their solubility in vitro or in vivo, including, for example, part or all of a morphogen pro domain, as described below, and casein, as described above.
  • tetracycline and diphosphonates are known to bind to bone mineral, particularly at zones of bone remodeling, when they are provided systemically in a mammal. Accordingly, these molecules may be included as useful agents for targeting OP-3 to bone tissue. Alternatively, an antibody or other binding protein that interacts specifically with a surface molecule on the desired target tissue cells also may be used. Such targeting molecules further may be covalently
  • Useful targeting molecules may be designed, for example, using the single chain binding site technology disclosed, for example, in U.S. Pat. No. 5,091,513.
  • the morphogen family members share significant sequence homology in the C-terminal active domains.
  • the sequences diverge significantly in the sequences which define the pro domain and the N-terminal 39 amino acids of the mature protein.
  • the pro domain and/or N-terminal sequence may be morphogen-specific.
  • the various morphogens identified to date are differentially expressed in the different tissues. Accordingly, without being limited to any given theory, it is likely that, under natural conditions in the body, selected morphogens typically act on a given tissue. Accordingly, part or all of morphogen-specific sequences may serve as targeting molecules for the morphogens described herein.
  • the pro domains may interact specifically with one or more molecules at the target tissue to direct the morphogen associated with the pro domain to that tissue.
  • another useful targeting molecule for targeting OP-3 to bone tissue may include part or all of a morphogen-specific sequence, such as part or all of a pro domain and/or the N-terminus of the mature protein.
  • Particularly useful are the morphogen-specific sequences of OP-1, BMP2 or BMP4, all of which proteins are found naturally associated with bone tissue (see, for example, US Pat. No. 5,011,691).
  • the morphogen-specific sequences of GDF-1 may be used to target morphogenic OP-3 to nerve tissue, particularly brain tissue where GDF-1 appears to be primarily expressed (see, for example, Lee,
  • pro forms of the proteins may be obtained from the culture medium of morphogen-secreting mammalian cells.
  • a suitable species may be formulated by complexing the mature dimer (or an active fragment thereof) with part or all of a pro domain.
  • Chimeric OP-3 proteins comprising, for example, non-OP-3 pro domains and/or non-OP-3 N-termini, may be synthesized using standard recombinant DNA methodology and/or automated chemical nucleic acid synthesis methodology well described in the art and as disclosed below.
  • OP-3 proteins provided herein may be administered alone or in combination with other agents.
  • useful cofactois for stimulating bone tissue growth in osteoporotic individuals include but are not limited to, vitamin D 3 , calcitonin, prostaglandins, parathyroid hormone, dexamethasone, estrogen and IGF-I or IGF-II.
  • useful cofactors for nerve tissue repair and regeneration may include nerve growth factors.
  • Other useful cofactors include
  • symptom-alleviating cofactors including antiseptics, antibiotics, antiviral and antifungal agents and analgesics and anesthetics.
  • compositions can be formulated into pharmaceutical compositions by admixture with pharmaceutically acceptable nontoxic excipients and carriers.
  • compositions may be prepared for parenteral administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets or capsules; or intranasally, particularly in the form of powders, nasal drops or aerosols.
  • the composition may include the morphogen dispersed in a fibrinogen-thrombin composition or other bioadhesive such as is disclosed, for example in PCT US91/09275, (WO92/10567).
  • the composition then may be painted, sprayed or otherwise applied to the desired tissue surface.
  • compositions can be formulated for parenteral or oral administration to humans or other mammals in therapeutically effective amounts, e.g., amounts which provide appropriate concentrations of OP-3 to target tissue for a time sufficient to induce morphogenesis, including particular steps thereof, as described above.
  • OP-3 is to be used as part of a transplant procedure, the morphogen may be provided to the living tissue or organ to be transplanted prior to removal of tissue or organ from the donor.
  • OP-3 may be provided to the donor host directly, as by injection of a formulation comprising OP-3 into the tissue, or
  • the organ or living tissue may be placed in a preservation solution containing OP-3.
  • the recipient also preferably is provided with the morphogen just prior to, or concommitant with,
  • OP-3 may be
  • OP-3 comprises part of a tissue or organ preservation solution
  • any commercially available preservation solution may be used to advantage.
  • useful solutions known in the art include
  • an organ preservation solution usually possesses one or more of the following properties: (a) an osmotic pressure substantially equal to that of the inside of a mammalian cell, (solutions typically are hyperosmolar and have K+ and/or Mg++ ions present in an amount sufficient to produce an osmotic pressure slightly higher than the inside of a mammalian cell); (b) the solution typically is capable of maintaining substantially normal ATP levels in the cells; and (c) the solution usually allows optimum maintenance of glucose metabolism in the cells.
  • Organ preservation solutions also may contain anticoagulants, energy sources such as glucose, fructose and other sugars, metabolites, heavy metal chelators, glycerol and other materials of high viscosity to enhance survival at low temperatures, free oxygen radical inhibiting and/or scavenging agents and a pH indicator.
  • energy sources such as glucose, fructose and other sugars, metabolites, heavy metal chelators, glycerol and other materials of high viscosity to enhance survival at low temperatures, free oxygen radical inhibiting and/or scavenging agents and a pH indicator.
  • OP-3 is envisioned to be useful in enhancing viability of any organ or living tissue to be
  • the morphogens may be used to particular advantage in lung, heart, liver, kidney or pancreas transplants, as well as in the transplantation and/or grafting of bone marrow, skin, gastrointestinal mucosa, and other living tissues.
  • the concentration of the compounds described in a therapeutic composition will vary depending upon a number of factors, including the dosage of the drug to be administered, the chemical characteristics (e.g., hydrophobicity) of the compounds employed, and the route of administration.
  • the preferred dosage of drug to be administered also is likely to depend on such variables as the type and extent of tissue loss or defect, the overall health status of the particular patient, the relative biological efficacy of the compound selected, the formulation of the compound, the presence and types of excipients in the formulation, and the route of administration.
  • the compounds of this invention may be provided in an aqueous physiological buffer solution containing about 0.001 to 10% w/v compound for parenteral
  • Typical dose ranges are from about 10 ng/kg to about 1 g/kg of body weight per day; a
  • preferred dose range is from about 0.1 ⁇ g/kg to
  • morphogen-induced pathological lesions are induced when mature morphogen (e.g., OP-1, 20 ⁇ g ) is administered daily to normal growing rats for 21 consecutive days.
  • mature morphogen e.g., OP-1, 20 ⁇ g
  • 10 ⁇ g systemic injections of morphogen (e.g., OP-1) injected daily for 10 days into normal newborn mice does not produce any gross abnormalities.
  • a morphogenically active fragment of OP-3 may be implanted surgically, dispersed in a biocompatible, preferably in vivo biodegradable matrix appropriately modified to provide a structure or scaffold in which the OP-3 may be dispersed and which allows the
  • the matrix also may provide signals capable of directing the tissue specificity of the differentiating cells, as well as providing a
  • the formulated matrix may be shaped as desired in anticipation of surgery or may be shaped by the
  • the material may be used in topical, subcutaneous,
  • the matrix preferably is biodegradable in vivo, being slowly absorbed by the body and replaced by new tissue growth, in the shape or very nearly in the shape of the
  • the matrix also may be particulate in nature.
  • Suitable biocompatible, in vivo biodegradable acellular matrices may be prepared from naturally-occurring tissue. The tissue is treated with suitable agents to substantially extract the cellular,
  • the agents also should be capable of extracting any morphogenesis inhibiting components associated with the tissue.
  • the resulting material is a porous, acellular matrix, substantially depleted in nonstructurally-associated components.
  • the matrix also may be further treated with agents that modify the matrix, increasing the number of pores and micropits on its surfaces.
  • agents that modify the matrix, increasing the number of pores and micropits on its surfaces.
  • Structural tissues such as cartilage and dentin where collagen is a primary proteinaceous component may be demineralized and extracted with guanidinium hydrochloride, essentially following the method of Sampath et al. (1983) PNAS 80: 6591-6595.
  • pulverized and demineralized dentin is
  • the matrix particles may further be treated with a collagen fibril-modifying agent that extracts potentially unwanted components from the matrix, and alters the surface structure of the matrix material.
  • Useful agents include acids, organic solvents or heated aqueous media.
  • the currently most preferred agent is a heated aqueous fibril-modifying medium such as water, to increase the matrix particle surface area and porosity.
  • the currently most preferred aqueous medium is an acidic aqueous medium having a pH of less than about 4.5, e.g., within the range of about pH 2 - pH 4 which may help to "swell" the collagen before heating. 0.1% acetic acid, which has a pH of about 3, currently is most preferred. 0.1 M acetic acid also may be used.
  • aqueous medium lg matrix/30ml aqueous medium
  • Preferred treatment times are about one hour, although exposure times of between about 0.5 to two hours appear acceptable.
  • the temperature employed is held constant at a temperature within the range of about 37°C to 65°C.
  • the currently preferred heat treatment temperature is within the range of about 45°C to 60°C.
  • the matrix After the heat treatment, the matrix is filtered, washed, lyophilized and used for implant. Where an acidic aqueous medium is used, the matrix also is preferably neutralized prior to washing and
  • a currently preferred neutralization buffer is a 200mM sodium phosphate buffer, pH 7.0.
  • the matrix preferably first is allowed to cool following thermal treatment, the acidic aqueous medium (e.g., 0.1% acetic acid) then is removed and replaced with the neutralization buffer and the matrix agitated for about 30 minutes.
  • the acidic aqueous medium e.g. 0.1% acetic acid
  • neutralization buffer then may be removed and the matrix washed and lyophilized.
  • Other useful fibril-modifying treatments include acid treatments (e.g., trifluoroacetic acid and
  • the treated matrix may be washed to remove any extracted components, following a form of the procedure set forth below: 1. Suspend matrix preparation in TBS (Tris-buffered saline) 1g/200 ml and stir at 4°C for 2 hrs; or in 6 M urea, 50 mM Tris-HCl, 500 mM NaCl, pH 7.0 (UTBS) or water and stir at room temperature (RT) for 30 minutes (sufficient time to neutralize the pH);
  • TBS Tris-buffered saline
  • suitable matrix materials may be obtained commercially.
  • an extracellular matrix extract such as MatrigelTM, (Collaborative
  • mouse sarcoma cells may be used to advantage.
  • tissue-specific matrices may be formulated synthetically.
  • porous biocompatible, in vivo biodegradable synthetic matrices are disclosed in PCT publication US91/03603, published December 12, 1991 (W091/18558). Briefly, the matrix comprises a porous crosslinked structural polymer of biocompatible, biodegradable collagen and appropriate, tissue-specific
  • Collagen derived from a number of sources may be suitable for use in these synthetic matrices, including insoluble collagen, acid-soluble collagen, collagen soluble in neutral or basic aqueous solutions, as well as those collagens which are commercially available.
  • Glycosammoglycans or mucopolysaccharides are hexosamine-containing polysaccharides of animal origin that have a tissue specific distribution, and therefore may be used to help determine the tissue specificity of the morphogen-stimulated differentiating cells. Reaction with the GAGs also provides collagen with another valuable property, i.e., inability to provoke an immune reaction (foreign body reaction) from an animal host.
  • GAGs are made up of residues of
  • hexosamines glycosidically bound and alternating in a more-or-less regular manner with either hexouronic acid or hexose moieties (see, e.g., Dodgson et al. in
  • GAGs include hyaluronic acid, heparin, heparin sulfate, chondroitin 6-sulfate, chondroitin 4-sulfate, dermatan sulfate, and keratin sulfate.
  • Other GAGs are suitable for forming the matrix described herein, and those skilled in the art will either know or be able to ascertain other suitable GAGs using no more than routine experimentation.
  • chondroitin-6-sulfate can be used where endochondral bone formation is desired.
  • Heparin sulfate may be used to formulate synthetic matrices for use in lung tissue repair.
  • Collagen can be reacted with a GAG in aqueous acidic solutions, preferably in diluted acetic acid solutions.
  • a GAG aqueous acidic solutions
  • coprecipitates of tangled collagen fibrils coated with GAG results.
  • This tangled mass of fibers then can be homogenized to form a homogeneous dispersion of fine fibers and then filtered and dried.
  • Insolubility of the collagen-GAG products can be raised to the desired degree by covalently cross-linking these materials, which also serves to raise the resistance to resorption of these materials.
  • any covalent cross-linking method suitable for cross-linking collagen also is suitable for cross-linking these composite materials, although
  • crosslinking by a dehydrothermal process is preferred.
  • the crosslinked particles are essentially spherical, with diameters of about 500 ⁇ m.
  • Scanning electron miscroscopy shows pores of about 20 ⁇ m on the surface and 40 ⁇ m on the interior.
  • the interior is made up of both fibrous and sheet-like structures, providing surfaces for cell attachment.
  • the voids interconnect, providing access to the cells throughout the interior of the particle.
  • the material appears to be roughly 99.5% void volume, making the material very efficient in terms of the potential cell mass that can be grown per gram of microcarrier.
  • Another useful synthetic matrix is one formulated from biocompatible, in vivo biodegradable synthetic polymers, such as those composed of glycolic acid, lactic acid and/or butyric acid, including copolymers and derivatives thereof. These polymers are well described in the art and are available commercially. For example, polymers composed of polyactic acid (e.g., MW 100 kDa), 80% polylactide/20% glycoside or poly 3-hydroxybutyric acid (e.g., MW 30 kDa) all may be purchased from PolySciences, Inc. The polymer
  • compositions generally are obtained in particulate form.
  • the steps preferably are performed under conditions which avoid hydrolysis of the polymers (e.g., non-aqueous conditions such as in an ethanol-trifluoro-acetic acid solution).
  • OP-3 proteins described herein can be combined and dispersed in a suitable matrix using any of the methods described below:
  • Matrix is added to the morphogen dissolved in guanidine-HCl. Samples are vortexed and incubated at a low temperature. Samples are then further vortexed. Cold absolute ethanol is added to the mixture which is then stirred and incubated. After centrifugation
  • a preparation of a morphogenically active fragment of OP-3 in physiological saline also may be vortexed with the matrix and lyophilized to produce
  • Tissue morphogenesis requires a morphogenically permissive environment. Clearly, in fully-functioning healthy tissue that is not composed of a permanently renewing cell population, there must exist signals to prevent continued tissue growth. Thus, it is
  • the bone-derived carrier is not demineralized but rather is washed only in low salt, for example, induction of endochondral bone formation is inhibited, suggesting the presence of one or more inhibiting factors within the carrier.
  • compositions of this invention may be purified from natural sources or produced using standard recombinant methodology.
  • a genetic sequence encoding the morphogenic OP-3 protein was identified using a 0.3 kb EcoRI-BamHI OP-2 fragment from a mouse OP-2 cDNA as a hybridization probe, specific to the mid-pro region of OP-2
  • the 32 P-labeled probe was prepared using the random hexanucleotide priming method, and the hybridizations were performed using the following conditions: 40% formamide, 5 X SSPE, 5 X Denhardt's Solution, 0.1% SDS, at 37°C overnight, and washing in 0.1 X SSPE, 0.1% SDS at 50°C.
  • the plasmid DNA containing the cDNAs was obtained using the lambda zapll excision process following manufacturer's directions. Three of the four clones were shown by DNA sequencing to encode OP-3. The DNA sequence, referred to herein as mOP-3 and described in Seq. ID No. 1, was identified by this procedure.
  • the isolated mOP-3 DNA sequence in accordance with other known morphogens, encodes a protein comprising a "pro" region (defined essentially by residues 20-260 or 20-263 of Seq. ID No. 1) and a mature region (defined essentially by residues 261-399 or 264-399 of Seq. ID No. 1), including a functional domain comprising the conserved cysteine skeleton.
  • OP-3 is marked by an eighth cysteine within the seven cysteine domain (e.g., at position 338 of Seq. ID No.l).
  • the extra cysteine likely helps stabilize the folded structure, possibly by providing inter-molecular disulfide bonding.
  • the extra cysteine also allows for heterodimer formation between OP-3 and another morphogen comprising the "eighth" cysteine, like OP-2 for example, or a modified OP-1, wherein an extra cysteine has been inserted at the appropriate location.
  • the extra cysteine also may allow tetramer formation.
  • the extra cysteine does not inhibit
  • a primary glycosylation site occurs just C terminal to the extra cysteine in both OP-2 and OP-3, which may provide a protective effect.
  • the cDNA sequences for both human and mouse OP-2 are provided in Seq. ID Nos. 7 and 9, and the genomic sequence for human OP-2 is provided in Seq. ID No. 11, wherein the exons defining the coding region of these proteins are indicated. The exon boundaries also are indicated in Fig. 1, described below.
  • the human OP-2 locus was isolated from a genomic library (Clontech, EMBL-3 #HL1067J) on three overlapping phage clones, using standard cloning procedures. The OP-2 coding information was spread over 27 kb and, like OP-1, contains 7 exons. A comparison of exon-intron
  • the first OP-2 exon contains 334 bp of coding sequence (111 amino acids), including the signal peptide, and is followed by the largest intron (14.6 kb).
  • the second exon 190 bp, 64 amino acids
  • the second exon is separated by a short intron (0.4 kb) from exon 3 (149 bp, 49 amino acids). It follows a large third intron of 9.5 kb.
  • the fourth exon (195 bp, 65 amino acids) encodes the maturation site
  • exon-Ala (“OP-2-Ala”) and is followed by a 0.8 kb intron.
  • the 7 cysteine domains resides on exons 5 to 7: exon 5 (80 bp, 27 amino acids) encodes the first cysteine of mature OP-2 and is followed by intron 5 (0.5 kb in length), exon 6 (111 bp, 37 amino acids) is separated by a 2.5 kb intron from the seventh, last exon with 147 bp (49 amino acids) of coding sequence.
  • exon-intron boundaries are conserved between human OP-1 and OP-2, two different members of the morphogen family of proteins.
  • the exonintron boundaries between human and mouse OP-2, two species variants of a morphogen are anticipated to be conserved as well.
  • Figure 1 shows the alignment of the murine OP-2 and murine OP-3 coding regions of the cDNA. The exon boundaries are indicated by bars beneath the sequence. Both sequences have the same number of nucleotides.
  • nucleotide sequence is about 80% conserved in the N-terminal and C-terminal regions.
  • nucleotide identity between the sequences is indicated by stippling.
  • the central region of the sequence is highly conserved and this conserved region falls into the boundaries of exon 2 and 3.
  • the high degree of conservation in the nucleotide sequences indicates that OP-2 and OP-3 likely share the nucleotide sequence of exon 2 and 3.
  • the different proteins may result from alternatively spliced
  • Intron 1 which lies upstream of exon 2 in OP-2 (see Seq. ID No.11) is large (14.6kb) and could include the start of the OP-3 gene and/or its first exon sequence.
  • one or more of the introns of these morphogens may include sequences having a transcription regulatory function.
  • OP-3 genetic sequences from other species and other libraries may be isolated.
  • a probe to the N-terminal region of the mature protein, or the 3' noncoding region flanking and immediately following the stop codon also may be used to screen for other OP-3 species variants.
  • OP-3 Mammalian cell expression of OP-3 readily can be achieved using COS (simian kidney ATCC, CRL-1650) or CHO (Chinese hamster ovary) cells (e.g., CHO-DXBII, from Lawrence Chasin, Columbia University, NY).
  • COS simian kidney ATCC, CRL-1650
  • CHO Choinese hamster ovary
  • Other useful eukaryotic cell systems include the insect/baculovirus system or the mammalian complement system.
  • the OP-3 DNA is subcloned into an insertion site of a suitable, commercially available pUC-type vector (e.g., pUC-19, ATCC #37254, Rockville, MD), along with a suitable promoter/enhancer sequences and 3' termination
  • promoter/enhancer sequences are the CMV-MIE promoter (human
  • cytomegalovirus major intermediate-early promoter preferably the intron-free or "short” form of the promoter
  • mMTV mouse mammary tumor virus promoter
  • mMTV mouse mammary tumor virus promoter
  • the plasmid also preferably contains a selectable marker, most
  • an amplifiable marker such as DHFR, e.g., under SV40 early promoter control (ATCC #37148).
  • Transfection, cell culturing, gene amplification and protein expression conditions are standard conditions, well known in the art, such as are described, for example in Ausubel et al., ed.. Current Protocols in Molecular Biology, John Wiley & Sons, NY (1989).
  • transfected cells are cultured in medium containing 0.1-0.5% dialyzed fetal calf serum (FCS), stably transfected high expression cell lines obtained by subcloning and evaluated by standard Northern blot. Southern blots also are used to assess the state of integrated OP-3 sequences and the extent of their copy number amplification .
  • Chimeric OP-3 morphogens e.g., comprising an OP-3 active domain and, for example, part or all of a pro domain from another, different morphogen may be
  • Useful chimeras include those wherein the non-OP-3 sequence is joined to the OP-3 sequence encoding the mature OP-3 protein, and the non-OP-3 sequence encodes part or all of the sequence between the signal peptide processing site and the
  • the non-OP-3 sequence may be joined to an OP-3 sequence encoding, for example, the 6 or 7 cysteine skeletons, wherein the non-OP-3 sequence includes the sequence encoding the N-terminus of the mature protein.
  • the non-OP-3 sequences may be composed of sequences from one or morphogens and/or may comprise novel biosynthetic sequences.
  • a construct encoding a chimeric OP1-OP3 polypeptide chain is demonstrated in the immunoblot presented in Fig. 2.
  • a vector carrying the construct under CMV promoter control was transfected into CHO cells (CHO-DXB11) using standard procedures and as described herein.
  • a chimeric gene was constructed by replacing the conserved seven cysteine domain of OP-1 with that of OP-3.
  • the resulting chimeric gene contains the entire pre-pro-domain of human OP-1 and the region of mature OP-1 between the maturation site and the first cysteine of the conserved C-terminal seven cysteine domain, fused to the conserved seven cysteine domain of mouse OP-3, but with two arginine residues in place of the native lysine residues found in OP-3 at the start of the seven cysteine domain.
  • the gene fusion was accomplished by splicing the Sad site of OP-3 (near the first cysteine of the seven cysteine domain) with a newly created Sad site in OP-1, created at the matching residues by silent mutagenesis.
  • the Sad site encodes the Glu-Leu
  • the chimeric gene was placed downstream of the CMV (Cytomegalovirus) MIE "short" (intron-free) promoter and upstream of the SV40 transcriptional terminator in a pUC vector.
  • This plasmid was cotransfected with DNA encoding the DHFR marker and viral trans-activating elements (e.g., VA1, E1A) into a CHO dhfr(-) host and subjected to Methotrexate selection and one round of amplification at 1 mM Methotrexate including
  • the 10 ⁇ l harvested medium was briefly heated with concentrated SDS sample buffer, containing ⁇ -mercapto ethanol (5%) and directly analysed by electrophoresis on a 15% SDS- polyacrylamide gel (in the buffer system of Laemmli) along with a set of prestained molecular weight standards (Bio-rad, Richmond, CA). Proteins were transferred from the gel to Immobilon membrane by the "Western blot" procedure.
  • the chimeric OP-1/OP-3 protein was detected by reaction with rabbit serum raised against a synthetic peptide representing the first 17 amino acids of mature OP-1, starting with serine-threonine-glycine-serine-.
  • recombinant OP-1 expressed in CHO cells was included for comparison.
  • lanes were as follows: lane 1: OP-1; lanes 4, 5, 6, 7, and 8:
  • the expressed protein then can be purified as follows. For a typical 2L preparation of transfected mammalian cells conditioned in 0.5% FCS, for example, the total protein is typically about 700 mg.
  • the amount of OP-3 in the media estimated by Western blot, is between about 0.1-5.0 mg.
  • OP-3 media then is diluted in a low salt, physiologically buffered 6M urea solution, and loaded onto an S-Sepharose column, which acts as a strong cation exchanger. OP-3 binds to the column in low salt, and serum proteins are removed.
  • the column subsequently is developed with an NaCl gradient, e.g., 0.1M NaCl-1.0M NaCl, in 6M urea, 20mM HEPES, pH 7.0.
  • OP-3 is eluted primarily at a higher salt concentration.
  • the sample then is loaded onto a phenyl-Sepharose column (hydrophobic interaction chromatography). OP-3 binds phenyl-Sepharose in the presence of high
  • a weak chaotropic salt e.g., IM (NH 4 ) 2 SO 4 in a physiologically buffered 6M urea
  • the column is developed with a decreasing ammonium sulfate gradient, e.g., 0.6M-0.0M (NH 4 ) 2 SO 4 gradient in a physiologically buffered, 6M urea solution. Again, most contaminants are removed at the start of the gradient, and OP-3 elutes primarily at low or no ammonium sulfate
  • the 0P-3 eluted from the phenyl-Sepharose column then is dialyzed against water, and prepared for loading onto a reverse phase chromatography column (e.g., C-18 HPLC), for example, by dialyzing against 30% acetonitrile, 0.1% TFA.
  • a reverse phase chromatography column e.g., C-18 HPLC
  • An alternative chromatography protocol is to perform the S-Sepharose chromatography in the absence of 6 M urea.
  • the bound proteins then are eluted with salt step elutions (e.g., 0.1-0.6M NaCl). Remaining OP-3 then can be eluted in the presence of 6M urea.
  • the 6M urea elution also may be used in place of the non-urea elution to achieve maximum recovery in one step.
  • OP-3 may be eluted from the phenylSepharose column in 38% ethanol-0.01% TFA, thereby eliminating the need to dialyze the eluent before applying it to the C-18 column.
  • multiple C-18 columns may be used (e.g., three), to further enhance purification and concentration of the protein.
  • OP-3 also will bind hydroxyapatite efficiently, typically in the absence of 6 M urea and at low
  • Bound OP-3 can be removed from the column with an elution gradient of about .001-0.5M step elution of phosphate in a physiologically buffered solution.
  • urea (6M) may be added during the elution step.
  • Other related chromatography methods also may be useful in purifying OP-3 from eucaryotic cell culture systems.
  • heparin-Sepharose may be used in combination with the S-Sepharose column.
  • IMAC chromatography
  • physiologically buffered phosphate solution may be used to advantage.
  • a currently preferred form of the OP-3 morphogen useful in therapeutic formulations having improved solubility in aqueous solutions and consisting
  • morphogenic protein essentially of amino acids, is a dimeric morphogenic protein comprising at least the 100 amino acid peptide sequence having the pattern of seven or more cysteine residues characteristic of the morphogen family
  • the dimeric morphogenic protein is
  • morphogenic protein preferably is noncovalently
  • the pro region peptides also preferably comprise at least the N-terminal eighteen amino acids that define the OP-3 morphogen pro region (e.g., residues 18-35 of Seq. ID No. 1). In a most preferred embodiment, peptides defining substantially the full length pro region are used.
  • soluble forms of morphogens include dimers of the uncleaved pro forms of these proteins, as well as "hemi-dimers" wherein one subunit of the dimer is an uncleaved pro form of the protein, and the other subunit comprises the mature form of the protein, including truncated forms thereof, preferably
  • useful pro domains include the full length pro regions, as well as various truncated forms hereof, particularly truncated forms cleaved at proteolytic Arg-Xaa-Xaa-Arg cleavage sites.
  • possible pro sequences cleaved at Arg-Xaa-Xaa-Arg sites include sequences defined by residues 18-260 of Seq. ID No. 1 (anticipated full length form); or by residues 18-263.
  • currently preferred pro sequences are those encoding the full length form of the pro region for OP-3 or another, known morphogen.
  • Other pro sequences contemplated to have utility include
  • biosynthetic pro sequences particularly those that incorporate a sequence derived from the N-terminal portion of one or more morphogen pro sequences.
  • useful sequences encoding the pro region may be obtained from genetic sequences encoding known morphogens.
  • chimeric pro regions can be constructed from the sequences of one or more known morphogens.
  • Still another option is to create a synthetic sequence variant of one or more known pro region sequences.
  • useful pro region peptides include polypeptide chains comprising an amino acid sequence encoded by a nucleic acid that hybridizes under stringent conditions with a DNA or RNA sequence encoding at least the N-terminal eighteen amino acids of the pro region sequence for OP-3 e.g., nucleotides 120-173 of Seq. ID No. 1.
  • useful pro region peptides include polypeptide chains comprising an amino acid sequence encoded by a nucleic acid that hybridizes under stringent conditions with a DNA or RNA sequence encoding at least the N-terminal eighteen amino acids of the pro region sequence for OP1 or OP2, e.g., nucleotides 136-192 and 152-211 of Seq. ID No. 3 and 7, respectively.
  • Morphogens are expressed from mammalian cells as soluble complexes. Typically, however the complex is disassociated during purification, generally by
  • soluble proteins from conditioned media (or, optionally, a body fluid such as serum, cerebro-spinal or peritoneal fluid), under non-denaturing conditions.
  • the method is rapid, reproducible and yields isolated soluble proteins
  • Soluble OP-3 morphogen complexes can be isolated from conditioned media using a simple, three step chromatographic protocol performed in the absence of denaturants.
  • the protocol involves running the media (or body fluid) over an affinity column, followed by ion exchange and gel filtration chromatographies.
  • the affinity column described below is a Zn-IMAC column.
  • the present protocol has general applicability to the purification of a variety of morphogens, all of which are anticipated to be isolatable using only minor modifications of the protocol described below.
  • An alternative protocol also envisioned to have utility an immunoaffinity column, created using standard
  • OP-1 was expressed in mammalian CHO (Chinese hamster ovary) cells as described in the art (see, for example, international application
  • the Zn-IMAC step separates the soluble OP-1 from the bulk of the contaminating serum proteins that elute in the flow through and 35 mM imidazole wash fractions.
  • the Zn-IMAC purified soluble OP-1 is next applied to an S-Sepharose cation-exchange column equilibrated in 20 mM NaPO 4 (pH 7.0) with 50 mM NaCl.
  • This S-Sepharose step serves to further purify and concentrate the soluble OP-1 complex in preparation for the following gel filtration step.
  • the protein was applied to a Sephacryl S-200HR column equilibrated in TBS.
  • soluble morphogens also may be isolated from one or more body fluids, including serum, cerebro-spinal fluid or peritoneal fluid. IMAC was performed using Chelating-Sepharose
  • the soluble OP-1 complex then is eluted with 50 mM imidazole (pH 8.0) in 20 mM HEPES and 500 mM NaCl.
  • the 50 mM imidazole eluate containing the soluble OP-1 complex was diluted with nine volumes of 20 mM NaPO. (pH 7.0) and applied to an S-Sepharose
  • ADH alcohol dehydrogenase
  • BSA bovine serum albumin
  • CA carbonic anhydrase
  • cytochrome C cytochrome C
  • the soluble OP-1 complex elutes with an apparent molecular weight of 110 kDa. This agrees well with the predicted composition of the soluble OP-1 complex with one mature OP-1 dimer (35-36 kDa) associated with two pro-domains (39 kDa each). Purity of the final complex can be verified by running the appropriate fraction in a reduced 15% polyacrylamide gel.
  • the complex components can be verified by running the complex-containing fraction from the S-200 or S- 200HR columns over a reverse phase C18 HPLC column and eluting in an acetonitrile gradient (in 0.1% TFA), using standard procedures.
  • the complex is dissociated by this step, and the pro domain and mature species elute as separate species. These separate species then can be subjected to N-terminal sequencing using
  • N-terminal sequencing of the isolated pro domain from mammalian cell produced OP-1 revealed 2 forms of the pro region, the intact form (beginning at residue 30 of Seq. ID No. 16) and a truncated form, (beginning at residue 48 of Seq. ID No. 16.)
  • N-terminal sequencing of the polypeptide subunit of the isolated mature species reveals a range of
  • soluble complexes may be formulated from purified pro domains and mature dimeric species. Successful complex formation
  • the denaturing conditions mimic the environment of an intracelluiar vesicle sufficiently such that the cleaved pro domain has an opportunity to associate with the mature dimeric species under relaxed folding conditions.
  • concentration of denaturant in the solution then is decreased in a controlled, preferably step-wise manner, so as to allow proper refolding of the dimer and pro regions while maintaining the association of the pro domain with the dimer.
  • Useful denaturants include 4-6M urea or guanidine hydrochloride (GuHCl), in buffered solutions of pH 4-10, preferably pH 6-8.
  • the soluble complex then is formed by controlled dialysis or dilution into a solution having a final denaturant concentration of less than 0.1-2M urea or GuHCl, preferably 1-2 M urea of GuHCl, which then preferably can be diluted into a physiological buffer. Protein purification/renaturing procedures and considerations are well described in the art, and details for
  • the stability of the highly purified soluble morphogen complex in a physiological buffer can be enhanced by any of a number of means.
  • a physiological buffer e.g., tris-buffered saline (TBS) and phosphate-buffered saline (PBS)
  • TBS tris-buffered saline
  • PBS phosphate-buffered saline
  • a pro region that comprises at least the first 18 amino acids of the pro sequence (e.g., residues 18-35 of Seq. ID NO. 1 for OP-3), and preferably is the full length pro region.
  • Residues 18-35 show sequence homology to the N-terminal portion of other morphogens and are believed to have particular utility in enhancing complex stability for all morphogens.
  • additives include basic amino acids (e.g., L-arginine, lysine and betaine); nonionic detergents (e.g., Tween 80 or Nonidet P-120); and carrier proteins (e.g., serum albumin and casein).
  • useful concentrations of these additives include 1-100 mM, preferably 10-70 mM, including 50 mM, basic amino acid;, 0.01-1.0%, preferably 0.05-0.2%, including 0.1% (v/v) nonionic detergent;, and 0.01-1.0%, preferably 0.05-0.2%, including 0.1% (w/v) carrier protein.
  • osteoblasts in vitro using the following assay.
  • rat osteoblast-enriched primary cultures preferably are used. Although these cultures are heterogeneous in that the individual cells are at different stages of differentiation, the culture is believed to more accurately reflect the metabolism and function of osteoblasts in vivo than osteoblast cultures obtained from established cell lines. Unless otherwise
  • Rat osteoblast-enriched primary cultures are prepared by sequential collagenase digestion of newborn suture-free rat calvaria (e.g., from 1-2 day-old animals, Long-Evans strain, Charles River Laboratories, Wilmington, MA), following standard procedures, such as are described, for example, in Wong et al., (1975) PNAS 72:3167-3171. Rat osteoblast single cell suspensions then are plated onto a multi-well plate (e.g., a
  • the cultured cells are divided into three groups: (1) wells which receive, for example, 0.1, 1.0, 10.0, 40 and 80.0 ng of OP-3; (2) wells which receive 0.1, 1.0, 10.0 and 40 ng of a local-acting growth factor (e.g., TGF- ⁇ ); and (3) the control group, which receive no growth factors.
  • the cells then are incubated for an additional 18 hours after which the wells are pulsed with 2 ⁇ Ci/well of 3 H-thymidine and incubated for six more hours.
  • the excess label then is washed off with a cold solution of 0.15 M NaCl and then 250 ⁇ l of 10% tricholoracetic acid is added to each well and the wells incubated at room temperature for 30 minutes.
  • the cells then are washed three times with cold
  • TGF- ⁇ incorporation into DNA, and thus promote osteoblast cell proliferation.
  • the effect of TGF- ⁇ is transient and biphasic. At high concentrations, TGF- ⁇ has no significant effect on osteoblast cell proliferation.
  • Useful naive stem cells include pluripotential stem cells, which may be isolated from bone marrow or umbilical cord blood using conventional methodologies, (see, for example. Faradji et al.,
  • a cultured mesodermal cell line may be useful.
  • Another method for obtaining progenitor cells and for determining the ability of OP-3 fragments to stimulate cell proliferation is to capture progenitor cells from an in vivo source. For example, a
  • biocompatible matrix material able to allow the influx of migratory progenitor cells may be implanted at an in vivo site long enough to allow the influx of migratory progenitor cells.
  • a bone-derived, guanidine-extracted matrix formulated as disclosed for example in Sampath et al. ((1983) PNAS 80:6591-6595), or U.S. Patent No. 4,975,526, may be implanted into a rat at a subcutaneous site, essentially following the method of Sampath et al. After three days the implant is removed, and the progenitor cells associated with the matrix dispersed and cultured.
  • Progenitor cells are incubated in vitro with OP-3 under standard cell culture conditions well described in the art and described hereinabove. In the absence of external stimuli, the progenitor cells do not, or only
  • progenitor cells cultured in the presence of a morphogenically active fragment of OP-3 are anticipated to proliferate.
  • Cell growth can be determined visually or spectrophotometrically using standard methods well known in the art.
  • Morphogenically active fragments of OP-3 can be utilized to induce cell differentiation.
  • the ability of OP-3 to induce cell differentiation can be
  • rat mesenchymal cells destined to become mandibular bone when separated from the overlying epithelial cells at stage 11 and cultured in vitro under standard tissue culture conditions, e.g., in a chemically defined, serum-free medium, containing for example, 67% DMEM (Dulbecco's modified Eagle's medium), 22% F-12 medium, 10mM Hepes pH 7, 2mM
  • aminopterin 12 ⁇ M thymidine
  • vascularized environment e.g., a vascularized environment.
  • Stage 11 mesenchymal cells cultured in vitro in the presence of OP-3, e.g., 10-100 ng/ml, are
  • osteoblast differentiation may be demonstrated in vitro using primary osteoblast cultures, or osteoblast-like cells lines, and assaying for a variety of bone cell markers that are specific markers for the
  • differentiated osteoblast phenotype e.g., alkaline phosphatase activity, parathyroid hormone-mediated cyclic AMP (cAMP) production, osteocalcin synthesis, and enhanced mineralization rates.
  • cAMP parathyroid hormone-mediated cyclic AMP
  • Osteoblasts by OP-3 The cultured cells in serum-free medium are
  • OP-3 concentrations for example, 0.1, 1.0, 10.0, 40.0 or 80.0 ng OP-3/ml medium; or with a similar range of TGF- ⁇
  • the cell layer is extracted with 0.5 ml of 1% Triton X-100.
  • the resultant cell extract then, is
  • PNPP paranitrosophenylphospate
  • the samples then are run through a plate reader (e.g., Dynatech MR700 plate reader, and absorbance measured at 400 nm, using p-nitrophenol as a standard) to determine the presence and amount of alkaline phosphate activity. Protein concentrations are determined by the Biorad method.
  • Rat osteoblasts are prepared and cultured in multiwell plates as described above. In this example six sets of 24 well plates are plated with 50,000 rat osteoblasts per well. The wells in each plate,
  • Example 3.1 using paranitroso-phenylphosphate (PNPP).
  • PNPP paranitroso-phenylphosphate
  • Rat osteoblasts are prepared and cultured in a multiwell plate as described above.
  • the cultured cells then are divided into three groups: (1) wells which receive, for example, 1.0, 10.0 and 40.0 ng OP-3/ml medium); (2) wells which receive for example, TGF- ⁇ , at similar concentration ranges; and (3) a control group which receives no growth factors.
  • the plate is then incubated for another 72 hours. At the end of the 72 hours the cells are treated with medium containing 0.5% bovine serum albumin (BSA) and ImM 3-isobutyl-1-methylxanthine for 20 minutes followed by the addition into half of the wells of human recombinant parathyroid hormone (hPTH, Sigma, St. Louis) at a concentration of 200 ng/ml for 10 minutes.
  • BSA bovine serum albumin
  • hPTH human recombinant parathyroid hormone
  • cAMP levels then are determined using a radioimmunoassay kit (e.g., Amersham, Arlington
  • Osteocalcin is a bone-specific protein synthesized by osteoblasts which plays an integral role in the rate of bone mineralization in vivo. Circulating levels of osteocalcin in serum are used as a marker for
  • Rat osteoblasts are prepared and cultured in a multi-well plate as above.
  • the medium is supplemented with 10%FBS, and on day 2, cells are fed with fresh medium supplemented with fresh 10 mM ⁇ -glycerophosphate (Sigma, Inc.). Beginning on day 5 and twice weekly thereafter, cells are fed with a complete mineralization medium containing all of the above components plus fresh L(+)-ascorbate, at a final concentration of 50 ⁇ g/ml medium.
  • OP-3 then is added to the wells directly, e.g., in 50% acetonitrile (or 50% ethanol) containing 0.1% trifluoroacetic acid (TFA), at no more than 5 ⁇ l morphogen/ml medium. Control wells receive solvent vehicle only.
  • the cells then are re-fed and the conditioned medium sample diluted 1:1 in standard radioimmunoassay buffer containing standard protease inhibitors and stored at -20° C until assayed for osteocalcin. Osteocalcin synthesis is measured by standard radioimmunoassay using a commercially
  • osteocalcin-specific antibody mineralization is determined on long term cultures (13 day) using a modified von Kossa staining technique on fixed cell layers: cells are fixed in fresh 4% paraformaldehyde at 23° C for 10 min, following rinsing cold 0.9% NaCl. Fixed cells then are stained for endogenous alkaline phosphatase at pH 9.5 for 10 min, using a commercially available kit (Sigma, Inc.)
  • OP-3 stimulates osteocalcin synthesis in osteoblast cultures.
  • the increased osteocalcin synthesis in response to OP-3 is dose dependent and shows a
  • the enhanced osteocalcin synthesis also can be confirmed by detecting the elevated osteocalcin mRNA message (20-fold increase) using a rat
  • osteocalcin-specific probe In addition, the increase in osteoclacin synthesis correlates with increased mineralization in long term osteoblast cultures as determined by the appearance of mineral nodules. OP-3 increases the initial mineralization rate significantly compared to untreated cultures. 3.5 Morphogen-Induced CAM Expression
  • the morphogens described herein induce CAM
  • CAMs are morphoregulatory molecules identified in all tissues as an essential step in tissue development.
  • N-CAMs which comprise at least 3 isoforms (N-CAM-180, N-CAM-140 and N-CAM-120, where "180", “140” and “120” indicate the apparent molecular weights of the isoforms as measured by SDS
  • polyacrylamide gel electrophoresis are expressed at least transiently in developing tissues, and
  • N-CAM-180 and N-CAM-140 isoforms are expressed in both developing and adult tissue.
  • the N-CAM-120 isoform is found only in adult tissue.
  • Another neural CAM is L1.
  • NG108-15 cells are a transformed hybrid cell line (neuroblastoma ⁇ glioma, America Type Tissue
  • NG108-15 cells exhibit a fibroblastic, or minimally differentiated, morphology and express only the 180 and 140 isoforms of N-CAM normally associated with a developing cell.
  • N-CAM isoforms are detected with an antibody which
  • Control NG108-15 cells express both the 140 kDa and the 180 kDa isoforms, but not the 120 kDa, as determined by Western blot analyses using up to 100 ⁇ g of protein.
  • Treatment of NG108-15 cells with OP-3 results in a dose-dependent increase in the expression of the 180 kDa and 140 kDa isoforms, as well as the induction of the 120 kDa isoform induced.
  • OP-3-induced CAM expression correlates with cell aggregation, as determined by histology.
  • the OP-3 morphogens described herein also can induce redifferentiation of transformed cells to a morphology characteristic of untransformed cells.
  • the examples provided below detail morphogen-induced redifferentiation of a transformed human cell line of neuronal origin (NG108-15); as well as mouse
  • neuroblastoma cells N1E-115
  • human embryo N1E-115
  • NG108-15 is a transformed hybrid cell line produced by fusing neuroblastoma ⁇ glioma cells (obtained from ATTC, Rockville, MD), and exhibiting a morphology characteristic of transformed embryonic neurons, e.g., having a fibroblastic
  • the cells have polygonal cell bodies, short, spike-like processes and make few contacts with neighboring cells (see copending
  • NG108-15 cells cultured in a chemically defined, serum-free medium, with 0.1 to 300 ng/ml of morphogen (e.g; OP-3) for four hours is anticipated to induce an orderly, dose-dependent change in cell morphology.
  • morphogen e.g; OP-3
  • NG108-15 cells are subcultured on poly-L-lysine coated 6 well plates. Each well contains 40-50,000 cells in 2.5 ml of chemically defined medium.
  • 2.5 ⁇ l of morphogen (e.g., OP-3) in 60% ethanol containing 0.025% trifluoroacetic is added to each well.
  • Morphogenic OP-3 of varying
  • concentrations are tested (typically, concentration ranges of 0-300 ng/ml are tested).
  • the media is changed daily with new aliquots of morphogen.
  • OP-3 is anticipated to induce a dose-dependent
  • morphogen-induced redifferentiation occurs without any associated changes in DNA synthesis, cell division, or cell viability, making it unlikely that the morphologic changes are secondary to cell differentiation or a toxic effect of the morphogen.
  • the morphogen-induced redifferentiation does not inhibit cell division, as determined by
  • 3H-thymidine uptake unlike other molecules which have been shown to stimulate differentiation of transformed cells, such as butyrate, DMSO, retanoic acid or
  • the OP-3 morphogens described herein accordingly provide useful therapeutic agents for the treatment of neoplasias and neoplastic lesions of the nervous system, particularly in the treatment of
  • neuroblastomas including retinoblastomas, and gliomas.
  • the ability of OP-3 to induce the "redifferentiation" of transformed human cells may be demonstrated using the following assay. Specifically, the effect of OP-3 on human EC cells (embryo carcinoma cells, e.g., NTERA-Z CL.D1,
  • ATCC ATCC, Rockville, MD
  • these cells may be maintained as undifferentiated stem cells, and can be induced to grow in serum free media (SFM).
  • SFM serum free media
  • the cells proliferate rampantly and are anchorage-independent.
  • EC cells grow as flattened cells, becoming anchorage dependent and forming
  • the cells are induced to differentiate.
  • varying concentrations of OP-3 e.g., 0-300 ng/ml
  • cultured cells e.g., 40-50,000 cells in
  • Morphogenically active fragments of OP-3 also may be used to maintain a cell's differentiated phenotype. This application is particularly useful for inducing the continued expression of phenotype in senescent or quiescent cells.
  • alkaline phosphatase activity of cultured osteoblasts is significantly reduced after multiple passages in vitro.
  • the cells are cultivated in the presence of OP-3, alkaline phosphatase activity should be maintained over extended periods of time.
  • phenotypic expression of myocytes also is maintained in the presence of a morphogen.
  • osteoblasts are cultured as described in Example 2.
  • the cells are divided into groups, incubated with varying concentrations of OP-3 (e.g., 0-300 ng/ml) and passaged multiple times (e.g., 3-5 times) using OP-3 (e.g., 0-300 ng/ml) and passaged multiple times (e.g., 3-5 times) using OP-3 (e.g., 0-300 ng/ml) and passaged multiple times (e.g., 3-5 times) using
  • Phenotypic maintenance capability also may be demonstrated in vivo, using a rat model for
  • rats are systemically provided with phosphate buffered saline (PBS) or morphogen, (e.g., OP-3, 1-100 ⁇ g) for 21 days (e.g., by daily tail vein injection.)
  • PBS phosphate buffered saline
  • morphogen e.g., OP-3, 1-100 ⁇ g
  • the rats then are sacrificed and serum alkaline phosphatase levels, serum calcium levels, and serum osteocalcin levels are determined, using standard methodologies as described therein and above. Elevated levels of osteocalcin and alkaline phosphatase should be observed in the rats treated with an effective amount of OP-3.
  • histomorphometric analysis on the tibial diasypheal bone is anticipated to show improved bone mass in
  • OP-3-treated animals as compared with untreated, ovariectomized rats.
  • the bone mass of OP-3-animals is anticipated to be comparable to (e.g., approaches) that of the sham-operated (e.g.,
  • Progenitor cells may be stimulated to proliferate in vivo or ex vivo.
  • the cells may be stimulated in vivo by injecting or otherwise providing a sterile preparation containing the morphogenically active fragment of OP-3 into the individual.
  • the hemopoietic pluripotential stem cell population of an individual may be stimulated to proliferate by
  • Progenitor cells may be stimulated ex vivo by contacting progenitor cells of the population to be enhanced with a morphogenically active fragment of OP-3 under sterile conditions at a concentration and for a time sufficient to stimulate proliferation of the cells. Suitable concentrations and stimulation times may be determined empirically, essentially following the procedure described in Example 2, above.
  • morphogen concentration of between about 0.1-100 ng/ml and a stimulation period of from about 10 minutes to about 72 hours, or, more generally, about 24 hours, typically should be sufficient to stimulate a cell population of about 10 4 to 10 6 cells.
  • the stimulated cells then are provided to the individual as, for example, by injecting the cells to an appropriate in vivo locus.
  • Suitable biocompatible progenitor cells may be obtained by any of the methods known in the art or described hereinabove.
  • OP-3 may be used to repair diseased or damaged mammalian tissue.
  • the tissue to be repaired preferably is assessed first, and excess necrotic or interfering scar tissue removed as needed, e.g., by ablation or by surgical, chemical, or other methods known in the medical arts.
  • OP-3 then may be provided directly to the tissue locus as part of a sterile, biocompatible composition, either by surgical implantation or injection.
  • the morphogen also may be provided systemically, as by oral or parenteral administration. Alternatively, a
  • progenitor cells stimulated by a morphogenically active fragment of OP-3 may be provided to the tissue locus.
  • the existing tissue at the locus whether diseased or damaged, provides the appropriate matrix to allow the proliferation and tissue-specific differentiation of progenitor cells.
  • a damaged or diseased tissue locus particularly one that has been further assaulted by surgical means, provides a morphogenically permissive environment.
  • Systemic provision of OP-3 should be sufficient for certain applications (e.g., in the treatment of osteoporosis and other disorders of the bone remodeling cycle, as an example). In some circumstances, particularly where tissue damage is extensive, the tissue may not be capable of providing a sufficient matrix for cell influx and proliferation.
  • OP-3 or progenitor cells stimulated by OP-3 may be necessary to provide OP-3 or progenitor cells stimulated by OP-3 to the tissue locus in association with a suitable, biocompatible, formulated matrix, prepared by any of the means described below.
  • the matrix preferably is in vivo biodegradable.
  • the matrix also may be
  • tissue-specific and/or may comprise porous particles having dimensions within the range of 70-850 ⁇ m, most preferably 150-420 ⁇ m.
  • OP-3 also may be used to prevent or substantially inhibit immune/inflammatory response-mediated tissue damage and scar tissue formation following an injury.
  • OP-3 is provided to a newly injured tissue locus, to induce tissue morphogenesis at the locus, preventing the aggregation of migrating fibroblasts into nondifferentiated connective tissue.
  • OP-3 preferably is provided as a sterile pharmaceutical preparation injected into the tissue locus within five hours of the injury. Where an immune/inflammatory response is unavoidably or deliberately induced, as part of, for example, a surgical or other aggressive clinical therapy, OP-3 preferably is provided prophylactically to the patient, prior to, or concomitant with, the therapy.
  • a particularly useful mammalian tissue model system for demonstrating and evaluating the morphogenic activity of a protein is the endochondral bone tissue morphogenesis model known in the art and described, for example, in U.S. Pat. No. 4,968,590.
  • the ability to induce endochondral bone formation includes the ability to induce the proliferation of progenitor cells into chondroblasts and osteoblasts, the ability to induce cartilage matrix formation, cartilage calcification, and bone remodeling, and the ability to induce
  • the local environment in which the morphogenic material is placed is important for tissue
  • local environment is understood to include the tissue structural matrix and the environment surrounding the tissue. For example, in addition to needing an appropriate anchoring
  • vascularization of new tissue requires a local environment which supports vascularization.
  • compositions may be injected or surgically implanted in a mammal, following any of a number of procedures well known in the art.
  • surgical implant bioassays may be
  • the stages include:
  • cartilage calcification on day eight cartilage calcification on day eight
  • appearance of osteoclastic cells and the commencement of bone remodeling and dissolution of the implanted matrix on days twelve to eighteen
  • tissue markers may be used as markers for tissue
  • Useful markers include tissue-specific enzymes whose activities may be assayed (e.g.,
  • alkaline phosphatase activity may be used as a marker for osteogenesis.
  • OP-3 may be followed using tagged fragments (e.g., radioactively labelled) and determining their localization in the new tissue, and/or by monitoring their disappearance from the circulatory system using a standard labeling protocol and pulse-chase procedure.
  • OP-3 also may be provided with a tissue-specific molecular tag, whose uptake may be monitored and correlated with the
  • OP-3 concentration of OP-3 provided.
  • ovary removal in female rats results in reduced bone alkaline phosphatase activity, and renders the rats predisposed to osteoporosis (as described in Example 5). If the female rats now are provided with OP-3, a reduction in the systemic concentration of calcium should be seen, which correlates with the presence of the provided OP-3 and which is anticipated to correspond with increased alkaline phosphatase activity.
  • morphogenesis of substantially injured liver tissue following a partial hepatectomy utilizing OP-3 is presented. Variations on this general protocol may be used to test morphogen activity of OP-3 in other different tissues.
  • the general method involves
  • liver has a potential to regenerate upon injury during post-fetal life.
  • OP-3 e.g., 1 mg/ml
  • a biocompatible solution for example, (e.g., a purified recombinant mature form of OP-3, is solubilized in 50% ethanol, or compatible solvent, containing 0.1% trifluoroacetic acid, or compatible acid.
  • the mature protein may be solubilized by association with a pro domain.
  • the injectable OP-3 solution is prepared, e.g., by diluting one volume of OP-3 solvent-acid stock solution with 9 volumes of 0.2% rat serum albumin in sterile PBS (phosphate-buffered saline).
  • OP-3 phosphate buffered saline/rat serum albumin
  • the OP-3 fragment-injected group is anticipated to show, e.g., complete liver tissue regeneration with no sign remaining of any cut in the liver.
  • the control group into which only PBS is injected show only minimal regeneration with the incision remaining in the sample.
  • Previous experiments with other morphogens e.g., OP-1) show these morphogens alone induce liver tissue
  • Pulp treatments used may include: a
  • OP-3 morphogenically active fragment of OP-3 dispersed in a carrier matrix; carrier matrix alone, and no treatment. Twelve teeth per animal (four for each treatment) are prepared, and two animals are used. At four weeks, teeth are extracted and processed histologically for analysis of dentin formation, and/or ground to analyze dentin mineralization. The effect of OP-3 on a carrier matrix; carrier matrix alone, and no treatment. Twelve teeth per animal (four for each treatment) are prepared, and two animals are used. At four weeks, teeth are extracted and processed histologically for analysis of dentin formation, and/or ground to analyze dentin mineralization. The effect of OP-3 on
  • osteodentin reparation may be observed visually by comparing control samples treatment (PBS) with OP-3.
  • OP-3 plus a carrier matrix induces formation of
  • untreated teeth or teeth treated with carrier vehicle alone do not induce periodontal tissue growth.
  • regenerative effects on central nervous system (CNS) repair by a morphogenically active fragment of OP-3, may be demonstrated using a rat brain stab model.
  • CNS central nervous system
  • male Long Evans rats are anesthetized and the head area prepared for surgery.
  • the calvariae is exposed using standard surgical procedures and a hole drilled toward the center of each lobe using a 0.035K wire, just piercing the calvariae. 25 ⁇ l
  • solutions containing either morphogen (e.g., OP-3, 25 ⁇ g) or PBS then is provided to each of the holes by Hamilton syringe. Solutions are delivered to a depth approximately 3 mm below the surface, into the
  • glial fibrillary acidic protein a marker protein for glial scarring, to qualitatively determine the degree of scar formation. Sections also are probed with OP-3-specific antibody to determine the presence of the protein. Reduced levels of glial fibrillary acidic protein are anticipated to be observed in the tissue sections of animals treated with OP-3,
  • OP-3 to stimulate peripheral nervous system axonal growth over extended distances may be demonstrated using the following model.
  • Neurons of the peripheral nervous system can sprout new processes on their own following injury, but without guidance these sproutings typically fail to connect appropriately and die. Where the break is extensive, e.g., greater than 5 or 10 mm, regeneration is poor or nonexistent.
  • the rat sciatic nerve can regenerate spontaneously across a 5 mm gap, and occasionally across a 10 mm gap, provided that the severed ends are inserted in a saline-filled nerve guidance channel. In this experiment, nerve regeneration across at least a 12mm gap is tested.
  • avascular intermuscular plane between vastus lateralis and hamstring muscles are entered and followed to the loose fibroareolar tissue surrounding the sciatic nerve.
  • the loose tissue is divided longitudinally thereby freeing the sciatic nerve over its full extent without devascularizing any portion.
  • the sciatic nerves are transected with microscissors at mid-thigh and grafted with a OP-3 gel graft that separates the nerve stumps by 12 mm.
  • the graft region is encased in a silicone tube 20 mm in length with a 1.5 mm inner diameter, the interior of which is filled with the morphogen solution.
  • the central 12 mm of the tube consists of an OP-3 gel prepared by mixing 1 to 5 ⁇ g of
  • extracellular matrix extract derived from mouse sarcoma tissue, and containing solubilized tissue basement membrane, including laminin, type IV collagen, heparin sulfate, proteoglycan and entactin, in phosphatebuffered saline.
  • the morphogen-filled tube then is implanted directly into the defect site, allowing 4 mm on each end to insert the nerve stumps . Each stump is abutted against the morphogen gel and is secured in the silicone tube by three stitches of commercially
  • Frozen sections then are cut throughout the graft site, and examined for axonal regeneration by immunofluorescent staining using anti-neurofilament antibodies labeled with flurocein (obtained, for example, from Sigma Chemical Co., St. Louis).
  • Determining the tissue distribution of morphogens may be used to identify different morphogens expressed in a given tissue, as well as to identify new, related morphogens. Tissue distribution also may be used to identify useful morphogen-producing tissue for use in screening and identifying candidate morphogen-stimulating agents.
  • the morphogens (or their mRNA transcripts) readily are identified in different tissues using standard methodologies and minor
  • protein distribution may be determined using standard Western blot analysis or immunofluorescent techniques, and antibodies specific to the morphogen or morphogens of interest.
  • distribution of morphogen transcripts may be determined using standard Northern hybridization protocols and transcript-specific probes.
  • any probe capable of hybridizing specifically to a transcript, and distinguishing the transcript of interest from other, related transcripts may be used. Because the morphogens described herein share such high sequence homology in their active, C-terminal domains, the tissue distribution of a specific morphogen
  • transcript may best be determined using a probe
  • Another useful sequence is the 3' non-coding region flanking and immediately following the stop codon.
  • OP-3-specific probe sequence is one derived from a portion of the 3' untranslated sequence, e.g., nucleotides 1310-1674 of Seq. ID No. 1, which shares little or no homology with other morphogen sequences, including OP-2.
  • the chosen fragment then is labelled using standard means well known and described in the art.
  • morphogen transcripts can be identified in mammalian tissue, using standard methodologies well known to those having ordinary skill in the art. A detailed description of a suitable hybridization protocol is described in Ozkaynak, et al., (1991)
  • RNA is prepared from various tissues (e.g., murine embryo and developing and adult liver, kidney, testis, heart, brain, thymus, stomach) by a standard methodology such as by the method of Chomczyaski et al. ((1987) Anal. Biochem
  • Poly (A)+ RNA is prepared by using oligo (dT)-cellulose chromatography (e.g.. Type 7, from Pharmacia LKB Biotechnology, Inc.). Poly (A)+ RNA (generally 15 ⁇ g) from each tissue is fractionated on a 1% agarose/formaldehyde gel and transferred onto a Nytran membrane (Schleicher &
  • the membrane is baked at 80°C and the RNA is cross-linked under UV light (generally 30 seconds at 1 mW/cm 2 ) .
  • the appropriate probe is denatured by heating. The hybridization is carried out in a lucite cylinder rotating in a roller bottle apparatus at approximately 1 rev/min for approximately 15 hours at 37°C using a hybridization mix of 40% formamide,
  • An OP-3-specific 0.5 kb probe was made from a StuI-BglII fragment of OP-3 cDNA.
  • the fragment contains the 3' untranslated sequence from nucleotides 1310-1674, plus an additional 140 bases.
  • the fragment was labelled using standard techniques and the
  • OP-3 like OP-2, appears to be expressed primarily in early embryonic tissue. Specifically, Northern blots of murine embryos show abundant OP-3 expression in 8-day embryos, demonstrated by a strong band at 2.9 kb and a weaker band at 2.3 kb.
  • Candidate compound(s) which may be administered to affect the level of endogenous OP-3 morphogen may be found using the following screening assay, in which the level of OP-3 production by a cell type which produces measurable levels of the morphogen is determined with and without incubating the cell in culture with the compound, in order to assess the effects of the
  • kidneys may be explanted from neonatal or new born or young or adult rodents (mouse or rat) and used in organ culture as whole or sliced (1-4 mm) tissues.
  • Primary tissue cultures and established cell lines, also derived from kidney, adrenals, urinary, bladder, brain, mammary, or other tissues may be established in multiwell plates (6 well or 24 well) according to conventional cell culture techniques, and are cultured in the absence or presence of serum for a period of time (1-7 days).
  • Cells may be cultured, for example, in Dulbecco's Modified Eagle medium (Gibco, Long Island, NY) containing serum (e.g., fetal calf serum at 1%-10%, Gibco) or in serum-deprived medium, as desired, or in defined medium (e.g., Dulbecco's Modified Eagle medium (Gibco, Long Island, NY) containing serum (e.g., fetal calf serum at 1%-10%, Gibco) or in serum-deprived medium, as desired, or in defined medium (e.g.,
  • an immunoassay may be performed to detect the morphogen using a polyclonal or monoclonal antibody specific for that protein.
  • OP-3 may be detected using a polyclonal antibody specific for OP-3 in an ELISA, as follows.
  • 96-well plate and incubated at 37 °C for an hour.
  • the wells are washed four times with 0.167M sodium borate buffer with 0.15 M NaCl (BSB), pH 8.2, containing 0.1% Tween 20.
  • BSA bovine serum albumin
  • amplification system kit is added and incubated for another 15 min at room temperature. The reaction is stopped by the addition of 50 ⁇ l 0.3 M sulphuric acid. The OD at 490 nm of the solution in each well is recorded. To quantitate OP-3 in culture media, an OP-3 standard curve is performed in parallel with the test samples.
  • Polyclonal antibody may be prepared as follows. Each rabbit is given a primary immunization of 100 ug/500 ⁇ l recombinantly-produced OP-3 protein or protein fragment in 0.1% SDS mixed with 500 ⁇ l
  • Complete Freund's Adjuvant The antigen is injected subcutaneously at multiple sites on the back and flanks of the animal. The rabbit is boosted after a month in the same manner using incomplete Freund's Adjuvant. Test bleeds are taken from the ear vein seven days later. Two additional boosts and test bleeds are performed at monthly intervals until antibody against OP-3 is detected in the serum using an ELISA assay. Then, the rabbit is boosted monthly with 100 ⁇ g of antigen and bled (15 ml per bleed) at days seven and ten after boosting.
  • Monoclonal antibody specific for a given morphogen may be prepared as follows. A mouse is given two injections of OP-3 protein or a protein fragment specific for OP-3.
  • the protein preferably is
  • the first injection contains lOO ⁇ g of OP-3 in complete Freund's adjuvant and is given subcutaneously.
  • the second injection contains 50 ⁇ g of OP-3 in incomplete adjuvant and is given
  • the mouse then receives a total of 230 ⁇ g of OP-3 in four intraperitoneal injections at various times over an eight month period.
  • OP-3 e.g., 100 ⁇ g
  • an OP-3-specific peptide e.g.,
  • mice spleen cells then are fused to commercially available myeloma cells at a ratio of 1:1 using PEG 1500 (Boeringer Mannheim,
  • CTGCAGCAAG TGACCTCGGG TCGTGGACCG CTGCCCTGCC CCCTCCGCTG CCACCTGGGG 60
  • AAGCATGTAA GGGTTCCAGA AACCTGAGCG TGCAGCAGCT GATGAGCGCC CTTTCCTTCT 1593

Abstract

Disclosed are (1) nucleic acid and amino acid sequences for a novel morphogenic protein; (2) methods for producing and expressing the protein in a biologically active form; and (3) methods for utilizing the protein to induce tissue morphogenesis in a mammal, including methods for increasing a progenitor cell population in a mammal, methods for stimulating progenitor cells to differentiate and maintain their differentiated phenotype in vivo or in vitro, methods for inducing tissue-specific growth in vivo and methods for the replacement of diseased or damaged tissue in vivo.

Description

OP-3-INDUCED MORPHOGENESIS
Field of the Invention
This invention relates generally to the field of tissue morphogenesis and more particularly to a novel protein that induces tissue morphogenesis in mammals.
Background of the Invention
Cell differentiation is the central characteristic of morphogenesis which initiates in the embryo, and continues to various degrees throughout the life of an organism in adult tissue repair and regeneration mechanisms. The degree of morphogenesis in adult tissue varies among different tissues and is related, among other things, to the degree of cell turnover in a given tissue. On this basis, tissues can be divided into three broad categories: (1) tissues with static cell populations such as nerve and skeletal muscle where there is no cell division and most of the cells formed during early development persist throughout adult life; (2) tissues containing conditionally renewing populations such as liver where there is generally little cell division but, in response to an appropriate stimulus, cells can divide to produce daughters of the same differentially defined type; and (3) tissues with permanently renewing populations including blood, testes and stratified squamous epithelia which are characterized by rapid and
continuous cell turnover in the adult. Here, the terminally differentiated cells have a relatively short life span and are replaced through proliferation of a distinct subpopulation of cells, known as stem or progenitor cells.
The cellular and molecular events which govern the stimulus for differentiation of these cells is an area of intensive research. In the medical field, it is anticipated that the discovery of factor(s) which control cell differentiation and tissue morphogenesis will advance significantly medicine's ability to repair and regenerate diseased or damaged mammalian tissues and organs. Particularly useful areas include
reconstructive surgery and in the treatment of tissue degenerative diseases including arthritis, emphysema, osteoporosis, cardiomyopathy, cirrhosis, and
degenerative nerve diseases.
A number of different factors have been isolated in recent years which appear to play a role in cell differentiation. Recently, various members of the structurally related proteins of the transforming growth factor (TGF)-β superfamily of proteins have been identified as true morphogens.
This "family" of proteins, sharing substantial amino acid sequence homology within their
morphogenically active C-terminal domains, including a conserved six or seven cysteine skeleton, are capable of inducing tissue-specific morphogenesis in a variety of organs and tissues, including bone, cartilage, liver, dentin, periodontal ligament, cementum, nerve tissue and the epithelial mucosa of the
gastrointestinal tract. The proteins apparently bind to surface receptors or otherwise contact and interact with progenitor cells, predisposing or stimulating the cells to proliferate and differentiate in a
morphogenically permissive environment. The morphogens are capable of inducing the developmental cascade of cellular and molecular events that culminate in the formation of new organ-specific tissue, including any vascularization, connective tissue formation, and nerve ennervation as required by the naturally occurring tissue.
Among the proteins useful in tissue morphogenesis are proteins originally identified as bone inductive proteins, such as the OP-1, (also referred to in related applications as "OP1"), OP-2 (also referred to in related applications as "OP2"), and the CBMP2 proteins, as well as amino acid sequence-related proteins such as BMP5, BMP6 and its murine homolog, Vgr-1, DPP and 60A (from Drosophila), Vgl (from
Xenopus), and GDF-1 (from mouse) see, for example, U.S. Patent No. 5,011,691 to Oppermann et al., Lee (1991) PNAS 88: 4250-4254, and Wharton et al. (1991) PNAS 88 : 9214-9218. These TGF-β superfamily members comprise a distinct subfamily of proteins different from other members of the TGF-β superfamily in that the family of morphogenic proteins are able to induce the full cascade of events that result in tissue morphogenesis, including stimulating cell proliferation and cell differentiation of progenitor cells, and supporting the growth and maintenance of differentiated cells. The morphogenic proteins apparently can act as endocrine, paracrine or autocrine factors. Specifically, the endogenous morphogens may be synthesized by the cells on which they act, by neighboring cells, or by cells of a distant tissue, the secreted protein being
transported to the cells to be acted on. In addition, the family of morphogenic proteins induce true tissue morphogenesis, rather than inducing formation of fibrotic (scar) tissue as, for example, TGF-β does. The morphogens are synthesized in the cell as a precursor molecule approximately three times larger than the mature protein that is processed to yield mature disulfide-linked dimers comprising the
C-terminal domain of the precursor sequence. The proteins are inactive when reduced e.g., in monomeric form, but are active as oxidized homodimeric species as well as when oxidized in combination with other morphogens under conditions to produce heterodimers. The proteins useful in tissue morphogenesis typically require a suitable environment enabling cells to migrate, proliferate and differentiate in a tissue-specific manner into, e.g., cartilage-producing
chondroblasts, bone-producing osteoblasts, hemopoietic cells, or liver cells, depending on the nature of the local environment. The proliferation and
differentiation of cells induced by the morphogenic proteins requires a suitable local environment, including a suitable substratum on which the cells can anchor. The proliferating and differentiating cells also require the presence of appropriate signals to direct their tissue-specificity, such as cell surface markers. It is an object of this invention to provide a novel purified morphogenic protein, "OP-3", including the amino acid sequence defining it and nucleic acids encoding it, including allelic, species, chimeric, and other amino acid sequence variants thereof, whether naturally occurring or biosynthetically constructed, and methods for utilizing the protein to induce the developmental cascade of tissue morphogenesis for a variety of tissues in mammals. The morphogenic
properties of OP-3 include the ability to induce proliferation and differentiation of progenitor cells, and the ability to support and maintain the
differentiated phenotype through the progression of events that results in the formation of adult tissue. Another object is to provide methods for the expression and isolation of morphogenically active species of OP-3 using recombinant DNA techniques. Yet another object is to provide generic sequences defining useful
morphogens. Still another object is to provide tissue-specific acellular matrices that may be used in
combination with OP-3, and methods for their preparation. Other objects include utilizing OP-3 in a variety of applications including methods for
increasing a progenitor cell population in a mammal; methods for stimulating progenitor cells to
differentiate in vivo or in vitro and to maintain their differentiated phenotype; methods for inducing tissue-specific growth in vivo, and methods for the
replacement of diseased or damaged tissue in vivo.
These and other objects and features of the invention will be apparent from the description, drawings, and claims which follow.
Summary of the Invention
A novel substantially pure genetic sequence
encoding a novel substantially pure protein referred to herein as "OP-3" now has been discovered. This novel protein is a member of the morphogenic protein family previously described by Applicants (see, for example, US92/01968 (W092/15323), and US92/07432 (WO93/05751). Accordingly, the invention provides methods for
utilizing OP-3 to induce the developmental cascade of tissue morphogenesis in a mammal. Specifically, methods are provided for utilizing OP-3 to induce the proliferation of uncommitted progenitor cells, to induce the differentiation of these stimulated
progenitor cells in a tissue-specific manner under appropriate environmental conditions, and to support the growth and maintenance of these differentiated cells. The protein also may be used to stimulate the "redifferentiation" of cells that have strayed from their differentiated phenotypes. Accordingly, OP-3 can be utilized to initiate and maintain the developmental cascade of tissue morphogenesis in an appropriate, morphogenically permissive environment. As used herein, useful OP-3 morphogens include proteins encoded by the DNA sequence provided in Seq. ID No. 1 ("mOP-3") and allelic and species variants thereof, as well as other naturally-occurring and biosynthetic amino acid sequence variants, including chimeric proteins, that are morphogenically active as defined herein. "Morphogenically active fragment" is understood to include all proteins and protein
fragments encoded by part or all of the sequence of Seq. ID No. 1 and which have morphogenic activity as defined herein. Specifically, as defined herein, a morphogen is a dimeric protein comprising a pair of polypeptide chains, wherein each polypeptide chain comprises at least the C-terminal six cysteine skeleton defined by residues 303 to 399 of Seq. ID No. 1 (or residues 335-431 of OP1, Seq. ID no. 3), including functionally equivalent arrangements of these cysteines (e.g., amino acid insertions or deletions which alter the linear arrangement of the cysteines in the sequence but not their relationship in the folded structure), such that, when the polypeptide chains are folded, the dimeric protein species comprising the pair of
polypeptide chains has the appropriate three-dimensional structure, including the appropriate intra- or inter-chain disulfide bonds such that the protein is capable of acting as a morphogen as defined herein. Specifically, the morphogens generally are capable of all of the following biological functions in a
morphogenically permissive environment: stimulating proliferation of progenitor cells; stimulating the differentiation of progenitor cells; stimulating the proliferation of differentiated cells; and supporting the growth and maintenance of differentiated cells.
In one aspect, the morphogens of this invention comprise a morphogenically active dimeric species comprising a pair of polypeptide chains, wherein at least one of the polypeptide chains comprises the amino acid sequence defined by residues 303 to 399 of Seq. ID No. 1 including allelic, species and other amino acid sequence variants thereof. In preferred morphogens, at least one polypeptide chain comprises the sequence defined by residues 298-399, residues 261-399 or residues 264-399 of Seq. ID No. 1. Alternatively, the amino acid sequence of both polypeptide chains may be defined by part or all of the amino acid sequence of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof, including
naturally-occurring sequence or biosynthetically constructed variants, and chimeric constructs as described below. Where only one polypeptide chain is defined by the amino acid sequence of part or all of Seq. ID. No. 1, the other polypeptide chain preferably comprises at least the sequence defining the C-terminal six cysteine skeleton of any of the other known
morphogen family members, including OP-1, OP-2, CBMP2A, CBMP2B, BMP3, BMP5, BMP6, Vgr-1, Vgl, 60A, DPP and GDF-1, described, for example, in US92/07432 (WO93/05751), including allelic, species and other amino acid
sequence variants thereof, including chimeric variants. Other useful sequences include biosynthetic constructs, such as are described in U.S. Pat. No. 5,011,691.
In still another aspect of the invention, generic sequences are provided which accommodate the sequence identity of useful morphogens and incorporate OP-3's novel features. In another aspect of the invention, morphogens of this invention comprise morphogenically active proteins encoded by part or all of the genetic sequence listed in Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof. In still another aspect, the invention comprises morphogens encoded by nucleic acids that hybridize to part or all of the pro region of the OP-3 protein, bases 120 to 848 of Seq ID No. 1, under stringent hybridization conditions. As used herein, "stringent hybridization conditions" are defined as hybridization in 40% formamide, 5 × SSPE, 5 × Denhardt's Solution, and 0.1% SDS at 37°C
overnight, and washing in 0.1 × SSPE, 0.1% SDS at 50°C. In one aspect of the invention, morphogenically active fragments of OP-3 are useful in the replacement of diseased or damaged tissue in a mammal, including, but not limited to, damaged lung tissue resulting from emphysema; cirrhotic tissue, including cirrhotic kidney or liver tissue; damaged heart or blood vessel tissue, as may result from cardiomyopathies and/or
atherothrombotic or cardioembolic strokes; damaged stomach and other mucosal tissues of the
gastrointestinal tract resulting from ulceric
perforations and/or their repair; damaged nerve tissue as may result from physical injury, degenerative diseases such as Alzheimer's disease, multiple
sclerosis, or strokes; damaged cartilage and bone tissue as may result from metabolic bone diseases and other bone remodeling disorders; damaged dentin, periodontal and/or cementum tissue as may result from disease or mechanical injury; and in the replacement of damaged tissue as a result of inflammation and/or chronic inflammatory disease.
As provided herein, morphogenically active
fragments of OP-3 are provided to a tissue-specific locus in vivo, to induce the developmental cascade of tissue morphogenesis at that site. Cells stimulated ex vivo by contact with OP-3 also may be provided to the tissue locus. In these cases the existing tissue provides the necessary matrix requirements, providing a suitable substratum or scaffold for the proliferating and differentiating cells in a morphogenically
permissive environment, as well as providing the necessary signals for directing the tissue-specificity of the developing tissue. The proteins or stimulated cells also may be combined with a formulated matrix and implanted as a device at a locus in vivo. The
formulated matrix should be a biocompatible, preferably biodegradable acellular matrix having the
characteristics described below. Where the necessary signals for directing the tissue-specificity of the developing tissue are not provided endogenously, the matrix preferably also is tissue-specific.
In another aspect, the members of the morphogen protein family also can control the body's cellular and humoral inflammatory response to a foreign object or an initial tissue injury. In many instances, the loss of tissue function results from the tissue destructive effects and the subsequent formation of scar tissue associated with the body's immune/inflammatory response to an initial or repeated injury to the tissue. The degree of scar tissue formation generally depends on the regenerative properties of the injured tissue, and on the degree and type of tissue damage. Thus, in another aspect, morphogenically active fragments of OP-3 may be used to prevent or to substantially inhibit the formation of scar tissue, including alleviating immune response-mediated tissue damage, by providing OP-3 or cells stimulated by exposure to OP-3 protein, to a newly injured tissue locus. The OP-3 protein also may be provided as a prophylactic, provided to a site in anticipation of tissue injury, such as part of a surgical or other clinical procedure likely to produce tissue damage, and to induce an inflammatory/immune response. In a particularly useful embodiment, OP-3 may be used as part of a transplant procedure, to enhance the tissue viability of the organ and/or tissue to be transplanted. The morphogen may be provided to the organ and/or tissue to be transplanted prior to harvest, during its transport, and/or during
transplantation into the recipient host as described below.
OP-3 also may be used to increase or regenerate a mesenchymal progenitor or stem cell population in vitro or in a mammal. For example, progenitor cells may be isolated from an individual's bone marrow, stimulated ex vivo with morphogenic OP-3 for a time and at a concentration sufficient to induce the cells to
proliferate, and returned to the bone marrow. Other sources of progenitor cells that may be suitable include biocompatible cells obtained from a cultured cell line, stimulated in culture, and subsequently provided to the body. Alternatively, OP-3 may be provided by systemic (e.g., oral or parenteral) administration, or it may be injected or otherwise provided to a progenitor cell population in an
individual to induce its mitogenic activity in vivo. For example, a morphogenically active fragment of OP-3 may be provided to the cells in vivo, e.g., by systemic injection, to induce mitogenic activity. Similarly, a particular population of hemopoietic stem cells may be increased by exposure to OP-3, for example by perfusing (plasmaphoresing) an individual's blood to extract the cells of interest, stimulating these cells ex vivo, and returning the stimulated cells to the blood.
It is anticipated that the ability to augment an individual's progenitor cell population will enhance existing methods for treating disorders resulting from a loss or reduction of a renewable cell population significantly. Two particularly significant
applications include the treatment of blood disorders and diseases involving impaired or lost immune
function. The morphogens of this invention also can inhibit proliferation of epithelial cell populations. The ability to inhibit epithelial cell proliferation may be exploited to reduce tissue damage associated with psoriasis and dermatitis, and other inflammatory skin diseases, as well as ulcerative diseases of the
gastrointestinal tract, such as, for example, in the healing of ulcers, including gastric ulcers, and the ulcerations induced in oral mucocitis and inflammatory bowel disease. Morphogens may be used to particular advantage as a cytoprotective agent in clinical
therapies likely to effect proliferating epithelial populations, such as cancer radiotherapies and
chemotherapies that typically induce oral mucositis, hair loss and/or skin disorders.
In another aspect of the invention, morphogenic OP-3 may be used to support the growth and maintenance of differentiated cells, inducing existing
differentiated cells to continue expressing their phenotype. It is anticipated that this activity will be particularly useful in the treatment of tissue disorders where loss of function is caused by reduced or lost metabolic function in which cells become senescent or quiescent, such as may occur in aging cells and/or may be manifested in osteoporosis and a number of nerve degenerative diseases, including
Alzheimer's disease. Application of OP-3 directly to the cells to be treated, or providing it systemically, as by oral or parenteral administration, can stimulate these cells to continue expressing their phenotype, thereby significantly reversing the effects of the dysfunction. In addition, a morphogenically active fragment of OP-3 also may be used in gene therapy protocols to stimulate the growth of quiescent cells, thereby potentially enhancing the ability of these cells to incorporate exogenous DNA.
In yet another aspect of the invention, a
morphogenically active fragment of OP-3 also may be used to induce "redifferentiation" of cells that have strayed from their differentiation pathway, such as can occur during tumorgenesis. It is anticipated that this activity will be particularly useful in treatments to reduce or substantially inhibit the growth of
neoplasms. The method also is anticipated to induce the de- and/or re-differentiation of these cells. As described supra, a morphogenically active OP-3 fragment may be provided to the cells directly or systemically, stimulating these cells to revert back to a morphology and phenotype characteristic of untransformed cells.
In still another aspect of the invention, OP-3 may be used to stimulate cell adhesion molecule (CAM) expression levels in a cell. CAMs are molecules defined as carrying out cell-cell interactions
necessary for tissue formation. CAMs are believed to play a fundamental regulatory role in tissue
development, including tissue boundary formation. embryonic induction and migration, and tissue
stabilization and regeneration. Altered CAM levels have been implicated in a number of tissue disorders, including congenital defects, neoplasias, and
degenerative diseases.
In particular, N-CAM expression is associated with normal neuronal cell development and differentiation, including retinal formation, synaptogenesis, and nervemuscle tissue adhesion. Inhibition of one or more of the N-CAM isoforms is known to prevent proper tissue development. Altered N-CAM expression levels also are associated with neoplasias, including neuroblastomas (see infra), as well as with a number of neuropathies, including normal pressure hydrocephalous and type II schizophrenia. Application of the morphogen directly to the cells to be treated, or providing the morphogen to the mammal systemically, for example, parenterally, or indirectly by oral administration, may be used to induce cellular expression of one or more CAMs, particularly N-CAMs and L1.
CAMs also have been postulated as part of a
morphoregulatory pathway whose activity is induced by a to date unidentified molecule (See, for example,
Edelman, G.M. (1986) Ann. Rev. Cell Biol., 2:81-116). Without being limited to any given theory, the
morphogens described herein may act as inducers of this pathway. The matrices utilized in the methods of the
invention may be derived from organ-specific tissue, or they may be formulated synthetically. In one
embodiment of the invention, when OP-3 (or a collection of progenitor cells stimulated by OP-3) is provided at a tissue-specific locus, e.g., by systemic
administration, implantation or injection at a tissue-specific locus, the existing tissue at that locus, whether diseased or damaged, has the capacity of acting as a suitable matrix or scaffold for the
differentiation and proliferation of migrating
progenitor cells. Alternatively, a formulated matrix may be provided externally together with the stimulated progenitor cells or morphogenically active OP-3
fragment, as may be necessary when the extent of injury sustained by the damaged tissue is large. The matrix should be a biocompatible, suitably modified acellular matrix having dimensions such that it allows the differentiation and proliferation of migratory
progenitor cells, and is capable of providing a
morphogenically permissive environment. The matrix also preferably allows cellular attachment and is biodegradable. Where the necessary tissue-directing signals can not be provided endogenously, the matrix preferably also is tissue-specific.
Formulated matrices may be generated from
dehydrated organ-specific tissue prepared, for example, by treating the tissue with solvents to substantially remove the intracellular, non-structural components from the tissue. Alternatively, the matrix may be formulated synthetically using a biocompatible, preferably in vivo biodegradable, structural molecule, and may be formulated with suitable tissue-specific cell attachment factors. The molecule may be a
naturally occurring one such as collagen, laminin or hyaluronic acid, or a synthetic polymer comprising, for example, polylactic acid, polybutyric acid,
polyglycolic acid, and copolymers thereof. Currently preferred structural polymers comprise tissue-specific collagens. Currently preferred cell attachment factors include glycosaminoglycans and proteoglycans. The matrix further may be treated with an agent or agents to increase the number of pores and micropits on its surfaces, so as to enhance the influx, proliferation and differentiation of migratory progenitor cells from the body of the mammal.
The invention thus relates to compositions and methods for the use of morphogenically active fragments of OP-3, a novel species variant of the generic family of morphogens disclosed in USSN 667,274 and USSN
752,764, as a tissue morphogen. Morphogenically active OP-3 and protein fragments can be isolated from
naturally-occurring sources, or they may be constructed biosynthetically using conventional recombinant DNA technology. Active OP-3 useful in the compositions and methods of this invention may include forms having varying glycosylation patterns, varying N-termini and active truncated forms, e.g., produced by recombinant DNA techniques. Active OP-3 proteins also include chimeric constructs as described below, comprising both an OP-3 active domain and a non-OP-3 sequence as, for example, the pro domain and/or the N-terminal region of the mature protein. OP-3 protein can be expressed from intact or truncated cDNA or from synthetic DNAs in procaryotic or eucaryotic host cells, and purified, cleaved, refolded, and dimerized to form
morphogenically active compositions. Useful host cells include procaryotes, including E. coli, and eucaryotic cells, including mammalian cells, such as CHO, COS, melanoma or BSC cells, or the insect/baculovirus system. Thus recombinant DNA techniques may be
utilized to produce large quantities of OP-3 capable of inducing tissue-specific cell differentiation and tissue morphogenesis in a variety of mammals, including humans.
Brief Description of the Drawings
Figure 1 is a nucleotide sequence comparison of the mouse cDNA sequence of OP-2 and OP-3. Exon boundaries are indicated by bars beneath the sequence; diamonds indicate nucleotide differences within exons 2 and 3; and
Figure 2 is an immunoblot comparing mammalian cell expression of an OP1/OP3 chimeric protein construct (lanes 4-8) with that of authentic, recombinant OP1 ( lane 1). Detailed Description
The invention provides a novel genetic sequence, mOP-3, encoding a novel protein, OP-3, having
morphogenic properties. The genetic sequence
originally was identified in a mouse cDNA library, and the invention provides methods for identifying and isolating the gene from other species. As will be appreciated by those skilled in the art, the methods described herein also may be used to isolate the OP-3 gene from other libraries, including genomic libraries. The invention also provides means for producing the OP-3 genetic sequence and the encoded protein. The invention further provides methods and compositions for inducing the developmental cascade of tissue
morphogenesis in a mammal utilizing morphogenically active fragments of OP-3. The methods and compositions provided herein may be utilized in a range of
applications, including stimulating the proliferation and/or differentiation of progenitor cells and inducing the repair and regeneration of damaged tissue. The morphogenic OP-3 species of the invention are novel members of the family of morphogens disclosed in
US92/01968 (W092/15323) and US92/07432 (WO93/05751). As described herein, OP-3 may be isolated from natural sources or constructed biosynthetically utilizing conventional recombinant DNA technology or constructed synthetically using standard chemical techniques.
Morphogenically active fragments of OP-3 are useful for initiating and maintaining the tissue-specific developmental cascade in a variety of tissues, including, but not limited to, bone, cartilage, dentin, neural tissue, liver, periodontal ligament, cementum, lung, heart, kidney and numerous tissues of the
gastrointestinal tract. When combined with naive mesenchymal progenitor cells as disclosed herein, OP-3 can induce the proliferation and differentiation of these progenitor cells. In the presence of appropriate tissue-specific signals to direct the differentiation of these cells, and a morphogenically permissive environment, OP-3 is capable of reproducing the cascade of cellular and molecular events that occur during embryonic development to yield functional tissue. For example, the protein can induce the de novo formation of cartilage and endochondral bone, including inducing the proliferation and differentiation of progenitor cells into chondrocytes and osteoblasts, inducing appropriate mineralization and bone remodeling,
inducing formation of an appropriate bone tissue vascular supply and inducing formation of
differentiated bone marrow (see Example 7 below.)
Provided below is a detailed description of the nucleic acid and amino acid sequences which describe OP-3 proteins useful in the compositions and methods of this invention, including a description of how to make them, and methods and means for their therapeutic administration. Also provided are numerous,
nonlimiting examples which (1) illustrate the
suitability of these proteins as tissue morphogens and therapeutic agents, and (2) provide assays with which to test the morphogens encompassed by the invention in different tissues. Also provided in Example 9 is a method for screening compounds to identify morphogen stimulating agents capable of stimulating endogenous OP-3 expression and/or secretion. OP-3 stimulating agents then may be used in any of the therapeutic applications described herein in place of, or in addition to, OP-3 protein administration.
I . Useful Morphogens
As defined herein a protein is morphogenic if it is capable of inducing the developmental cascade of cellular and molecular events that culminate in the formation of new, organ-specific tissue and comprises at least the conserved C-terminal six cysteine skeleton or its functional equivalent (see supra).
Specifically, the morphogens generally are capable of all of the following biological functions in a
morphogenically permissive environment: stimulating proliferation of progenitor cells; stimulating the differentiation of progenitor cells; stimulating the proliferation of differentiated cells; and supporting the growth and maintenance of differentiated cells.
Details of how the morphogen family of proteins
described herein first were identified, as well as a description of how to make, use and test them for morphogenic activity are disclosed, for example, in international application US92/01968 (W092/15323). As disclosed therein, the morphogens may be purified from naturally-sourced material or recombinantly produced from procaryotic or eucaryotic host cells, preferably as described therein. Alternatively, novel morphogenic sequences may be identified following the procedures disclosed therein.
Particularly useful morphogens identified to date include OP-1, OP-2, CBMP2A and CBMP2B (the
morphogenically active domains of proteins referred to in the art as BMP2A and BMP2B, or BMP2 and BMP4, respectively), BMP3, BMP5, BMP6, Vgr-1, GDF-1, Vgl, DPP and 60A, including their allelic and species variants, as well as other amino acid sequence variants,
including chimeric morphogens. Morphogenically active biosynthetic constructs such as those disclosed in U.S. Pat. No. 5,011,691, (e.g., COP-1, COP-3, COP-4, COP-5, COP-7, and COP-16) also are envisioned to be useful.
The novel morphogen OP-3 and its genetic sequence now have been identified. The OP-3 proteins useful in the invention include any morphogenically active fragment of the OP-3 amino acid sequence present in Seq. ID No. 1, or allelic, species or other amino acid sequence variants thereof. The morphogenically active fragment of OP-3 also may include any morphogenically active protein encoded by part or all of the nucleic acid sequence presented in Seq. ID No. 1. The
morphogenic protein also may comprise a protein encoded by part or all of a nucleic acid which hybridizes to at least part of the nucleic acid sequence encoding the "pro" region of the OP-3 protein, e.g., bases 120-848 of Seq. ID No. 1, under stringent conditions. The mOP-3 gene encodes a protein ("mOP-3") first expressed as an immature translation product that is 399 amino acids in length. This precursor form, referred to herein as the "prepro" form, (Seq. ID.
No. 1, amino acid residues 1-399) includes an
N-terminal signal peptide sequence, typically less than about 20 residues, followed by a "pro" domain that is cleaved to yield the mature sequence. The "pro" form of the protein includes the pro domain and the mature domain, and forms a soluble species that appears to be the primary form secreted from cultured mammalian cells. The signal peptide, anticipated to include residues 1-17 for mOP3, is cleaved rapidly upon
translation, at a cleavage site that can be predicted in a given sequence using the method of Von Heijne ((1986) Nucleic Acids Research 14:4683-4691). The preferred form of morphogenically active OP-3 protein comprises a processed sequence, including fragments thereof, appropriately dimerized and disulfide bonded. Where a soluble form of the protein is desired, the protein preferably comprises both the mature domain, or an active portion thereof, and part or all of the pro domain. By amino acid sequence homology with other, known morphogens, the pro domain likely is cleaved at
residues 257-260 of Seq. ID No. 1, which represent the canonical Arg-Xaa-Xaa-Arg cleavage site, to yield a mature sequence 139 amino acids in length (Seq. ID No. 1, residues 261-399). Alternatively, the pro domain may be cleaved at residues 260-263 to yield a shorter sequence 135 amino acids in length (Seq. ID No. 1, amino acid residues 264-399). All morphogens, including OP-1, OP-2 and the OP-3 proteins disclosed herein, comprise at least a conserved six cysteine skeleton in the amino acid sequence C-terminal domain and, preferably, a conserved seven cysteine skeleton (see, for example, US92/01968 (W092/15323). The conserved six cysteine skeleton in mOP-3 (Seq. ID No. 1) is defined by amino acid residues 303-399; the conserved seven cysteine skeleton is defined by amino acid residues 298-399. In addition to the conserved six cysteine skeleton found in known morphogen family members including OP-1, OP-2, CBMP2A, CBMP2B, BMP3, BMP5, BM 6, Vgr-1, Vgl, 60A, DPP and GDF-1, described, for example, in PCT/US92/07432 (WO93/05751), the OP-3 proteins, like the OP-2 proteins, also has one
additional cysteine residue (residue 338 of Seq. ID No. 1) in the conserved C-terminal domain. The mature sequence of OP-3 shares significant amino acid sequence homology with the morphogens identified to date. Specifically, the seven cysteine fragment shows greater than 79% amino acid identity with the corresponding mOP-2 and hOP-2 sequences, and greater than 66% identity with the corresponding OP-1 sequences. Like OP-2, OP-3 has an eighth cysteine within the seven cysteine domain (e.g., at position 338 of Seq. ID No. 1). In addition, OP-3 is unique among the morphogens identified to date in that the residue at position 9 in the conserved seven cysteine domain (e.g., residue 315 of Seq. ID No. 1) is a serine, whereas other morphogens typically have a tryptophan at this location (see Table I below, and Table II in
PCT/US92/07358 (WO93/04692), for example.)
As used herein, "amino acid sequence homology" is understood to mean amino acid sequence similarity, and homologous sequences share identical or similar amino acids, where similar amino acids are conserved amino acids as defined by Dayoff et al.. Atlas of Protein Sequence and Structure; vol.5, Suppl.3, pp.345-362 (M.O. Dayoff, ed., Nat'l BioMed. Research Fdn.,
Washington D.C. 1978.) Thus, a candidate sequence sharing 70% amino acid homology with a reference sequence requires that, following alignment of the candidate sequence with the reference sequence, 70% of the amino acids in the candidate sequence are identical to the corresponding amino acid in the reference sequence, or constitute a conserved amino acid change thereto. "Amino acid sequence identity" is understood to require identical amino acids between two aligned sequences. Thus, a candidate sequence sharing 60% amino acid identity with a reference sequence requires that, following alignment of the candidate sequence with the reference sequence , 60% of the amino acids in the candidate sequence are identical to the
corresponding amino acid in the reference sequence. As used herein, all homologies and identities calculated use OP-3 as the reference sequence. Also as used herein, sequences are aligned for homology and identity calculations using the method of Needleman et al. (1970) J.Mol. Biol. 48:443-453 and identities calculated by the Align program (DNAstar, Inc.) In all cases, internal gaps and amino acid insertions in the candidate sequence as aligned are ignored when making the homology/identity calculation.
Thus, useful OP-3 variants include, but are not limited to, amino acid sequences derived from Seq. ID No. 1 and wherein the cysteine at position 338 is replaced with another amino acid, preferably a
tyrosine, histidine, isoleucine or serine and
conservative substitutions thereof, e.g., such as defined by Dayoff et al., Atlas of Protein Sequence and Structure; vol. 5, Suppl. 3, pp.345-362 (M.O. Dayoff, ed., Nat'l BioMed. Research Fdn., Washington D.C.
1979.). Still other useful OP-3 variants include proteins wherein the serine at position 315 is replaced with another amino acid, preferably a tryptophan and conservative substitutions thereof. Generic Sequence 7 (Seq. ID No. 12) and Generic Sequence 8 (Seq. ID No. 13) disclosed below,
accommodate the homologies shared among perferred morphogen protein family members identified to date, including OP-1, OP-2, OP-3, CBMP2A, CBMP2B, BMP3, 60A, DPP, Vgl, BMP5, BMP6, Vrg-1, and GDF-1. The amino acid sequences for these proteins are described herein (see Sequence Listing and Table I below) and/or in the art, as well as in PCT publication US 92/07358, filed August 28, 1992, for example. The generic sequences include both the amino acid identity shared by these sequences in the C-terminal domain, defined by the six and seven cysteine skeletons (Generic Sequences 7 and 8,
respectively), as well as alternative residues for the variable positions within the sequence. The generic sequences allow for an additional cysteine at position 41 (Generic Sequence 7) or position 46 (Generic
Sequence 8), providing an appropriate cysteine skeleton where inter- or intramolecular disulfide bonds can form, and containing certain critical amino acids which influence the tertiary structure of the proteins.
Generic Sequence 7
Leu Xaa Xaa Xaa Phe
1 5
Xaa Xaa Xaa Gly Trp Xaa Xaa Xaa Xaa
10
Xaa Xaa Pro Xaa Xaa Xaa Xaa Ala
15 20
Xaa Tyr Cys Xaa Gly Xaa Cys Xaa
25 30 Xaa Pro Xaa Xaa Xaa Xaa Xaa
35
Xaa Xaa Xaa Asn His Ala Xaa Xaa
40 45
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa
50
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
55 60
Cys Xaa Pro Xaa Xaa Xaa Xaa Xaa
65
Xaa Xaa Xaa Leu Xaa Xaa Xaa
70 75
Xaa Xaa Xaa Xaa Val Xaa Leu Xaa
80
Xaa Xaa Xaa Xaa Met Xaa Val Xaa
85 90
Xaa Cys Xaa Cys Xaa
95
wherein each Xaa is independently selected from a group of one or more specified amino acids defined as
follows: "Res." means "residue" and Xaa at res.2 = (Tyr or Lys); Xaa at res.3 = Val or lie); Xaa at res.4 = (Ser, Asp or Glu); Xaa at res.6 = (Arg, Gln, Ser, Lys or Ala); Xaa at res.7 = (Asp or Glu); Xaa at res .8 = (Leu, Val or Ile); Xaa at res.11 = (Gln, Leu, Asp, His, Asn or Ser); Xaa at res . 12 = (Asp, Arg , Asn or Glu ) ; Xaa at res . 13 = (Trp or Ser); Xaa at res.14 = (Ile or Val); Xaa at res.15 = (Ile or Val); Xaa at res.16 (Ala or Ser); Xaa at res.18 = (Glu, Gln, Leu, Lys, Pro or Arg); Xaa at res.19 = (Gly or Ser); Xaa at res.20 = (Tyr or Phe); Xaa at res.21 = (Ala, Ser, Asp, Met, His, Gln, Leu or Gly); Xaa at res.23 = (Tyr, Asn or Phe); Xaa at res.26 = (Glu, His, Tyr, Asp, Gln, Ala or Ser); Xaa at res.28 = (Glu, Lys, Asp, Gln or Ala); Xaa at res.30 = (Ala, Ser, Pro, Gln, Ile or Asn); Xaa at res.31 = (Phe, Leu or Tyr); Xaa at res.33 = (Leu, Val or Met); Xaa at res.34 = (Asn, Asp, Ala, Thr or Pro); Xaa at res.35 = (Ser, Asp, Glu, Leu, Ala or Lys); Xaa at res.36 = (Tyr, Cys, His, Ser or Ile); Xaa at res.37 = (Met, Phe, Gly or Leu); Xaa at res.38 = (Asn, Ser or Lys); Xaa at res.39 = (Ala, Ser, Gly or Pro); Xaa at res.40 = (Thr, Leu or Ser); Xaa at res.44 = (Ile, Val or Thr); Xaa at res.45 = (Val, Leu, Met or Ile); Xaa at res.46 = (Gln or Arg); Xaa at res.47 = (Thr, Ala or Ser); Xaa at res.48 = (Leu or Ile); Xaa at res.49 = (Val or Met); Xaa at res.50 = (His, Asn or Arg); Xaa at res.51 = (Phe, Leu, Asn, Ser, Ala or Val); Xaa at res.52 = (Ile, Met, Asn, Ala, Val, Gly or Leu); Xaa at res.53 = (Asn, Lys, Ala, Glu, Gly or Phe); Xaa at res.54 = (Pro, Ser or Val); Xaa at res.55 = (Glu, Asp, Asn, Gly, Val, Pro or Lys); Xaa at res.56 = (Thr, Ala, Val, Lys, Asp, Tyr, Ser, Gly, Ile or His); Xaa at res.57 = (Val, Ala or Ile); Xaa at res.58 = (Pro or Asp); Xaa at res.59 = (Lys, Leu or Glu); Xaa at
res.60 = (Pro, Val or Ala); Xaa at res .63 = (Ala or Val); Xaa at res.65 = (Thr, Ala or Glu); Xaa at res.66 = (Gln, Lys, Arg or Glu); Xaa at res.67 = (Leu, Met or Val); Xaa at res.68 = (Asn, Ser, Asp or Gly); Xaa at res.69 = (Ala, Pro or Ser); Xaa at res.70 = (Ile, Thr, Val or Leu); Xaa at res.71 = (Ser, Ala or Pro); Xaa at res.72 = (Val, Leu, Met or Ile); Xaa at res.74 = (Tyr or Phe); Xaa at res.75 = (Phe, Tyr, Leu or His); Xaa at res.76 = (Asp, Asn or Leu); Xaa at res.77 = (Asp, Glu, Asn, Arg or Ser); Xaa at res.78 = (Ser, Gln, Asn, Tyr or Asp); Xaa at res.79 = (Ser, Asn, Asp, Glu or Lys); Xaa at res.80 = (Asn, Thr or Lys); Xaa at res.82 = (Ile, Val or Asn); Xaa at res.84 = (Lys or Arg); Xaa at res.85 = (Lys, Asn, Gln, His, Arg or Val); Xaa at res.86 = (Tyr, Glu or His); Xaa at res.87 = (Arg, Gln, Glu or Pro); Xaa at res.88 = (Asn, Glu, Trp or Asp);
Xaa at res.90 = (Val, Thr, Ala or Ile); Xaa at res.92 = (Arg, Lys, Val, Asp, Gln or Glu); Xaa at res.93 = (Ala, Gly, Glu or Ser); Xaa at res.95 = (Gly or Ala) and Xaa at res.97 = (His or Arg).
As described above. Generic Sequence 8 (Seq. ID No. 13) includes all of Generic Sequence 7 and in addition includes the following sequence at its N-terminus: Cys Xaa Xaa Xaa Xaa
1 5
Accordingly, beginning with residue 7, each "Xaa" in Generic Seq. 8 is a specified amino acid defined as for Generic Seq. 7, with the distinction that each residue number described for Generic Sequence 7 is shifted by five in Generic Seq. 8. Thus, "Xaa at res.2 =(Tyr or Lys)" in Gen. Seq. 7 refers to Xaa at res. 7 in Generic Seq. 8. In Generic Seq. 8, Xaa at res.2 = (Lys, Arg, Ala or Gln); Xaa at res.3 = (Lys, Arg or Met); Xaa at res.4 = (His, Arg or Gln); and Xaa at res.5 = (Glu, Ser, His, Gly, Arg, Pro, Thr, or Tyr).
Table I, set forth below, compares the C-terminal amino acid sequences defining the seven cysteine skeleton of human OP-1, mouse OP-1, human OP-2, mouse OP-2, and mouse OP-3 (mOP-3, Seq. ID No. 1). In the table, the sequences are aligned essentially following the method of Needleman et al. (1970) J. Mol. Biol., 48: 443-453, calculated using the Align Program
(DNAstar, Inc.) In the table, three dots indicates that the amino acid in that position is the same as the amino acid in hOP-1. Three dashes indicate that no amino acid is present in that position, and are
included for purposes of illustrating homologies. As is apparent from the following amino acid sequence comparisons, significant amino acid sequence homology exists between mouse OP-3 and mouse and human OP-1 and OP-2.
TABLE I
hOP-1 Cys Lys Lys His Glu Leu Tyr Val mOP-1 ... ... ... ... ... ... ... ...
hOP-2 ... Arg Arg ... ... ... ... ...
mOP-2 ... Arg Arg ... ... ... ... ...
mOP-3 ... Arg Arg ... ... ... ... ... hOP-1 Ser Phe Arg Asp Leu Gly Trp Gln Asp mOP-1 ... ... ... ... ... ... ... ... ... hOP -2 ... ... Gln ... ... ... ... Leu . . . mOP-2 ... ... ... ... ... ... . . . Leu . . . mOP-3 ... ... ... ... ... ... ... Leu . . .
10 15
hOP-1 Trp Ile Ile Ala Pro Glu Gly Tyr Ala mOP-1 ... ... ... ... ... ... ... ...
hOP-2 ... Val ... ... ... Gln ... ... Ser mOP-2 ... Val . .. ... ... Gln . . . . . . Ser mOP-3 Ser Val . .. ... ... Gln . . . . . . Ser
20 25 hOP-1 Ala Tyr Tyr Cys Glu Gly Glu Cys Ala mOP-1 ... ... ... ... ... ... ... ... ... hOP-2 ... ... ... ... ... ... ... ... Ser mOP-2 ... ... ... ... ... ... ... ... ... mOP-3 ... ... ... ... Ala ... ... ... Ile
30 35 hOP-1 Phe Pro Leu Asn Ser Tyr Met Asn Ala mOP-1 ... ... ... ... ... ... ... ... ... hOP-2 ... ... ... ... ... ... ... ... ... mOP-2 ... ... ... Asp ... Cys ... ... ... mOP-3 Tyr ... ... ... ... Cys ... ... Ser
40
hOP-1 Thr Asn His Ala Ile Val Gln Thr Leu mOP-1 ... ... ... ... ... ... ... ... ... hOP-2 ... ... ... .... ... ... ... ... . . . mOP-2 ... ... ... ... ... ... ... ... . . . mOP-3 ... ... ... ... Thr Met ... Ala ...
45 50 hOP-1 Val His Phe Ile Asn Pro Glu Thr Val mOP-1 ... ... ... ... ... ... Asp ... ... hOP-2 ... ... Leu Met Lys ... Asn Ala ... mOP-2 ... ... Leu Met Lys ... Asp Val ... mOP-3 ... ... Leu Met Lys ... Asp Ile Ile
55 60 hOP-1 Pro Lys Pro Cys Cys Ala Pro Thr Gln mOP-1 ... ... ... ... ... ... ... ... ... hOP-2 ... ... Ala ... ... ... ... ... Lys mOP-2 ... ... Ala ... ... ... ... ... Lys mOP-3 ... ... Val ... ... Val ... ... Glu
65 70 hOP-1 Leu Asn Ala Ile Ser Val Leu Tyr Phe mOP-1 .... ... ... ... ... ... ... ... ... hOP-2 ... Ser ... Thr ... ... ... ... Tyr mOP-2 ... Ser ... Thr ... ... ... ... Tyr mOP-3 ... Ser ... ... ... Leu ... ... Tyr
75 80 hOP-1 Asp Asp Ser Ser Asn Val Ile Leu Lys mOP-1 ... ... ... ... ... ... ... ... ... hOP-2 ... Ser ... Asn ... ... ... ... Arg mOP-2 ... Ser ... Asn ... ... ... ... Arg mOP-3 ... Arg Asn Asn ... ... ... ... Arg
85
hOP-1 Lys Tyr Arg Asn Met Val Val Arg mOP-1 ... ... ... ... ... ... ... ...
hOP-2 ... His ... ... ... ... ... Lys mOP-2 ... His ... ... ... ... ... Lys mOP-3 Arg Glu ... ... ... ... ... Gln
90 95 hOP-1 Ala Cys Gly Cys His
mOP-1 ... ... ... ... ...
hOP-2 ... ... ... ... ...
mOP-2 ... ... ... ... ...
mOP-3 ... ... ... ... ...
100 II. Formulations and Methods for Administering OP-3 Protein as Therapeutic Agents
II.A OP-3 Protein Considerations
The morphogens described herein may be provided to an individual by any suitable means, preferably
directly or systemically, e.g., parenterally or orally. Where the morphogen is to be provided directly (e.g., locally, as by injection, to a desired tissue site), or parenterally, such as by intravenous, subcutaneous, intramuscular, intraorbital, ophthalmic,
intraventricular, intracranial, intracapsular,
intraspinal, intracisternal, intraperitoneal, buccal, rectal, vaginal, intranasal or by aerosol
administration, the morphogen preferably comprises part of an aqueous solution. The solution is
physiologically acceptable so that in addition to delivery of the desired morphogen to the patient, the solution does not otherwise adversely affect the patient's electrolyte and volume balance. The aqueous medium for the morphogen thus may comprise normal physiologic saline (0.9% NaCl, 0.15M), pH 7-7.4. The aqueous solution containing the morphogen can be made, for example, by dissolving the protein in 50% ethanol, or acetonitrile containing 0.1% trifluoroacetic acid (TFA) or 0.1% HCl, or equivalent solvents. One volume of the resultant solution then is added, for example, to ten volumes of phosphate buffered saline (PBS), which further may include 0.1-0.2% human serum albumin (HSA). The resultant solution preferably is vortexed extensively. If desired, a given morphogen may be made more soluble by association with a suitable molecule. For example, association of the mature dimer with the pro domain of the morphogen increases solubility of the protein significantly. For example,the pro form of OP-3 comprises a species that is soluble in
physiologically buffered solutions. In fact, the endogenous protein is thought to be transported (e.g., secreted and circulated) to particular tissues in this form. This soluble form of the protein may be obtained from the culture medium of morphogen-secreting
mammalian cells. Alternatively, a soluble species may be formulated by complexing the mature dimer (or an active fragment thereof) with part or all of a pro domain. Another molecule capable of enhancing
solubility and particularly useful for oral
administrations, is casein. For example, addition of 0.2% casein increases solubility of the mature active form of OP-1 by 80%. Other components found in milk and/or various serum proteins also may be useful.
Useful solutions for oral or parenteral
administration may be prepared by any of the methods well known in the pharmaceutical art, described, for example, in Remington's Pharmaceutical Sciences,
(Gennaro, A., ed.). Mack Pub., 1990. Formulations may include, for example, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin,
hydrogenated naphthalenes, and the like. Formulations for direct administration, in particular, may include glycerol and other compositions of high viscosity. Biocompatible, preferably bioresorbable polymers, including, for example, hyaluronic acid, collagen, tricalcium phosphate, polybutyrate, polylactide, polyglycolide and lactide/glycolide copolymers, may be useful excipients to control the release of the
morphogen in vivo.
Other potentially useful parenteral delivery systems for these morphogens include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation administration may contain as excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally.
Alternatively, the morphogens described herein may be administered orally. Oral administration of
proteins as therapeutics generally is not practiced as most proteins readily are degraded by digestive enzymes and acids in the mammalian digestive system before they can be absorbed into the bloodstream. However, the morphogens described herein typically are acid-stable and protease-resistant (see, for example, U.S. Pat. No. 4,968,590.) In addition, at least one morphogen, OP-1, has been identified in bovine mammary gland extract, colostrum and milk, as well as saliva. Moreover, the OP-1 purified from mammary gland extract is
morphogenically active. For example, this protein induces endochondral bone formation in mammals when implanted subcutaneously in association with a suitable matrix material, using a standard in vivo bone assay, such as is disclosed in U.S. Pat. No. 4,968,590. In addition, endogenous morphogen also is detected in human serum. These findings indicate that oral and parenteral administration are viable means for
administering morphogens to an individual. Moreover, while the mature forms of certain morphogens described herein typically are sparingly soluble, the morphogen form found in milk (and mammary gland extract and colostrum) is readily soluble, probably by association of the mature, morphogenically active form with the pro domain of the intact sequence and/or by association with one or more milk components. Accordingly, the compounds provided herein also may be associated with molecules capable of enhancing their solubility in vitro or in vivo, including, for example, part or all of a morphogen pro domain, as described below, and casein, as described above.
The compounds provided herein also may be
associated with molecules capable of targeting the morphogen to a desired tissue. For example,
tetracycline and diphosphonates (bisphosphonates) are known to bind to bone mineral, particularly at zones of bone remodeling, when they are provided systemically in a mammal. Accordingly, these molecules may be included as useful agents for targeting OP-3 to bone tissue. Alternatively, an antibody or other binding protein that interacts specifically with a surface molecule on the desired target tissue cells also may be used. Such targeting molecules further may be covalently
associated to the morphogen, e.g., by chemical
crosslinking, or by using standard genetic engineering means to create, for example, an acid labile bond such as an Asp-Pro linkage. Useful targeting molecules may be designed, for example, using the single chain binding site technology disclosed, for example, in U.S. Pat. No. 5,091,513.
As described above, the morphogen family members share significant sequence homology in the C-terminal active domains. By contrast, the sequences diverge significantly in the sequences which define the pro domain and the N-terminal 39 amino acids of the mature protein. Accordingly, the pro domain and/or N-terminal sequence may be morphogen-specific. As described above, it also is known that the various morphogens identified to date are differentially expressed in the different tissues. Accordingly, without being limited to any given theory, it is likely that, under natural conditions in the body, selected morphogens typically act on a given tissue. Accordingly, part or all of morphogen-specific sequences may serve as targeting molecules for the morphogens described herein. For example, the pro domains may interact specifically with one or more molecules at the target tissue to direct the morphogen associated with the pro domain to that tissue. Thus, another useful targeting molecule for targeting OP-3 to bone tissue, for example, may include part or all of a morphogen-specific sequence, such as part or all of a pro domain and/or the N-terminus of the mature protein. Particularly useful are the morphogen-specific sequences of OP-1, BMP2 or BMP4, all of which proteins are found naturally associated with bone tissue (see, for example, US Pat. No. 5,011,691). Alternatively, the morphogen-specific sequences of GDF-1 may be used to target morphogenic OP-3 to nerve tissue, particularly brain tissue where GDF-1 appears to be primarily expressed (see, for example, Lee,
(1991) PNAS, 88:4250-4254. As described above, pro forms of the proteins may be obtained from the culture medium of morphogen-secreting mammalian cells.
Alternatively, a suitable species may be formulated by complexing the mature dimer (or an active fragment thereof) with part or all of a pro domain. Chimeric OP-3 proteins comprising, for example, non-OP-3 pro domains and/or non-OP-3 N-termini, may be synthesized using standard recombinant DNA methodology and/or automated chemical nucleic acid synthesis methodology well described in the art and as disclosed below.
Finally, the OP-3 proteins provided herein may be administered alone or in combination with other
molecules known to have a beneficial effect on tissue morphogenesis, including molecules capable of tissue repair and regeneration and/or inhibiting inflammation. Examples of useful cofactois for stimulating bone tissue growth in osteoporotic individuals, for example, include but are not limited to, vitamin D3, calcitonin, prostaglandins, parathyroid hormone, dexamethasone, estrogen and IGF-I or IGF-II. Useful cofactors for nerve tissue repair and regeneration may include nerve growth factors. Other useful cofactors include
symptom-alleviating cofactors, including antiseptics, antibiotics, antiviral and antifungal agents and analgesics and anesthetics.
The compounds provided herein can be formulated into pharmaceutical compositions by admixture with pharmaceutically acceptable nontoxic excipients and carriers. As noted above, such compositions may be prepared for parenteral administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets or capsules; or intranasally, particularly in the form of powders, nasal drops or aerosols. Where adhesion to a tissue surface is desired the composition may include the morphogen dispersed in a fibrinogen-thrombin composition or other bioadhesive such as is disclosed, for example in PCT US91/09275, (WO92/10567). The composition then may be painted, sprayed or otherwise applied to the desired tissue surface.
The compositions can be formulated for parenteral or oral administration to humans or other mammals in therapeutically effective amounts, e.g., amounts which provide appropriate concentrations of OP-3 to target tissue for a time sufficient to induce morphogenesis, including particular steps thereof, as described above. Where OP-3 is to be used as part of a transplant procedure, the morphogen may be provided to the living tissue or organ to be transplanted prior to removal of tissue or organ from the donor. OP-3 may be provided to the donor host directly, as by injection of a formulation comprising OP-3 into the tissue, or
indirectly, e.g., by oral or parenteral administration, using any of the means described above. Alternatively or, in addition, once removed from the donor, the organ or living tissue may be placed in a preservation solution containing OP-3. In addition, the recipient also preferably is provided with the morphogen just prior to, or concommitant with,
transplantation. In all cases, OP-3 may be
administered directly to the tissue at risk, as by injection to the tissue, or it may be provided
systemically, either by oral or parenteral
administration, using any of the methods and
formulations described herein and/or known in the art.
Where OP-3 comprises part of a tissue or organ preservation solution, any commercially available preservation solution may be used to advantage. For example, useful solutions known in the art include
Collins solution, Wisconsin solution, Belzer solution, Eurocollins solution and lactated Ringer's solution. Generally, an organ preservation solution usually possesses one or more of the following properties: (a) an osmotic pressure substantially equal to that of the inside of a mammalian cell, (solutions typically are hyperosmolar and have K+ and/or Mg++ ions present in an amount sufficient to produce an osmotic pressure slightly higher than the inside of a mammalian cell); (b) the solution typically is capable of maintaining substantially normal ATP levels in the cells; and (c) the solution usually allows optimum maintenance of glucose metabolism in the cells. Organ preservation solutions also may contain anticoagulants, energy sources such as glucose, fructose and other sugars, metabolites, heavy metal chelators, glycerol and other materials of high viscosity to enhance survival at low temperatures, free oxygen radical inhibiting and/or scavenging agents and a pH indicator. A detailed description of preservation solutions and useful components may be found, for example, in US Patent No. 5,002,965.
OP-3 is envisioned to be useful in enhancing viability of any organ or living tissue to be
transplanted. The morphogens may be used to particular advantage in lung, heart, liver, kidney or pancreas transplants, as well as in the transplantation and/or grafting of bone marrow, skin, gastrointestinal mucosa, and other living tissues.
As will be appreciated by those skilled in the art, the concentration of the compounds described in a therapeutic composition will vary depending upon a number of factors, including the dosage of the drug to be administered, the chemical characteristics (e.g., hydrophobicity) of the compounds employed, and the route of administration. The preferred dosage of drug to be administered also is likely to depend on such variables as the type and extent of tissue loss or defect, the overall health status of the particular patient, the relative biological efficacy of the compound selected, the formulation of the compound, the presence and types of excipients in the formulation, and the route of administration. In general terms, the compounds of this invention may be provided in an aqueous physiological buffer solution containing about 0.001 to 10% w/v compound for parenteral
administration. Typical dose ranges are from about 10 ng/kg to about 1 g/kg of body weight per day; a
preferred dose range is from about 0.1 μg/kg to
100 mg/kg of body weight. No obvious morphogen-induced pathological lesions are induced when mature morphogen (e.g., OP-1, 20 μg ) is administered daily to normal growing rats for 21 consecutive days. Moreover, 10 μg systemic injections of morphogen (e.g., OP-1) injected daily for 10 days into normal newborn mice does not produce any gross abnormalities.
II.B Matrix Preparation
A morphogenically active fragment of OP-3 may be implanted surgically, dispersed in a biocompatible, preferably in vivo biodegradable matrix appropriately modified to provide a structure or scaffold in which the OP-3 may be dispersed and which allows the
differentiation and proliferation of migrating progenitor cells. The matrix also may provide signals capable of directing the tissue specificity of the differentiating cells, as well as providing a
morphogenically permissive environment, being
essentially free of growth inhibiting signals.
The formulated matrix may be shaped as desired in anticipation of surgery or may be shaped by the
physician or technician during surgery. Thus, the material may be used in topical, subcutaneous,
intraperitoneal, or intramuscular implants to repair tissue or to induce its growth de novo. The matrix preferably is biodegradable in vivo, being slowly absorbed by the body and replaced by new tissue growth, in the shape or very nearly in the shape of the
implant. The matrix also may be particulate in nature.
Details of how to make and how to use the matrices useful in this invention are disclosed below.
II.B(i) Tissue-Derived Matrices
Suitable biocompatible, in vivo biodegradable acellular matrices may be prepared from naturally-occurring tissue. The tissue is treated with suitable agents to substantially extract the cellular,
nonstructural components of the tissue. The agents also should be capable of extracting any morphogenesis inhibiting components associated with the tissue. The resulting material is a porous, acellular matrix, substantially depleted in nonstructurally-associated components. The matrix also may be further treated with agents that modify the matrix, increasing the number of pores and micropits on its surfaces. Those skilled in the art will know how to determine which agents are best suited to the extraction of nonstructural components for different tissues. For example, soft tissues such as liver and lung may be thin-sectioned and exposed to a nonpolar solvent such as, for example, 100% ethanol, to destroy the cellular structure of the tissue and extract nonstructural components. The material then may be dried and pulverized to yield nonadherent porous particles or it may be maintained as a gel-like
solution. Structural tissues such as cartilage and dentin where collagen is a primary proteinaceous component may be demineralized and extracted with guanidinium hydrochloride, essentially following the method of Sampath et al. (1983) PNAS 80: 6591-6595. For example, pulverized and demineralized dentin is
extracted with five volumes of 4M guanidinium-HCl, 50mM Tris-HCl, pH 7.0 for 16 hours at 4°C. The suspension then is filtered. The insoluble material that remains is collected and used to fabricate the matrix. The material is mostly collagenous in matter. It is devoid of morphogenic activity. The matrix particles may further be treated with a collagen fibril-modifying agent that extracts potentially unwanted components from the matrix, and alters the surface structure of the matrix material. Useful agents include acids, organic solvents or heated aqueous media. A detailed description of these matrix treatments are disclosed, for example, in U.S. Patent No. 4,975,526 and PCT publication US90/00912, published September 7, 1990 (WO90/10018). The currently most preferred agent is a heated aqueous fibril-modifying medium such as water, to increase the matrix particle surface area and porosity. The currently most preferred aqueous medium is an acidic aqueous medium having a pH of less than about 4.5, e.g., within the range of about pH 2 - pH 4 which may help to "swell" the collagen before heating. 0.1% acetic acid, which has a pH of about 3, currently is most preferred. 0.1 M acetic acid also may be used.
Various amounts of delipidated, demineralized guanidine-extracted bone collagen are heated in the aqueous medium ( lg matrix/30ml aqueous medium) under constant stirring in a water jacketed glass flask, and maintained at a given temperature for a predetermined period of time. Preferred treatment times are about one hour, although exposure times of between about 0.5 to two hours appear acceptable. The temperature employed is held constant at a temperature within the range of about 37°C to 65°C. The currently preferred heat treatment temperature is within the range of about 45°C to 60°C.
After the heat treatment, the matrix is filtered, washed, lyophilized and used for implant. Where an acidic aqueous medium is used, the matrix also is preferably neutralized prior to washing and
lyophilization. A currently preferred neutralization buffer is a 200mM sodium phosphate buffer, pH 7.0. To neutralize the matrix, the matrix preferably first is allowed to cool following thermal treatment, the acidic aqueous medium (e.g., 0.1% acetic acid) then is removed and replaced with the neutralization buffer and the matrix agitated for about 30 minutes. The
neutralization buffer then may be removed and the matrix washed and lyophilized.
Other useful fibril-modifying treatments include acid treatments (e.g., trifluoroacetic acid and
hydrogen fluoride) and solvent treatments such as dichloromethane, acetonitrile, isopropanol and
chloroform, as well as particular acid/solvent
combinations. After contact with the fibril-modifying agent, the treated matrix may be washed to remove any extracted components, following a form of the procedure set forth below: 1. Suspend matrix preparation in TBS (Tris-buffered saline) 1g/200 ml and stir at 4°C for 2 hrs; or in 6 M urea, 50 mM Tris-HCl, 500 mM NaCl, pH 7.0 (UTBS) or water and stir at room temperature (RT) for 30 minutes (sufficient time to neutralize the pH);
2. Centrifuge and repeat wash step; and
3. Centrifuge; discard supernatant; water wash residue; and then lyophilize. Alternatively, suitable matrix materials may be obtained commercially. For example, an extracellular matrix extract such as Matrigel™, (Collaborative
Research, Inc., Bedford) derived from mouse sarcoma cells, may be used to advantage.
II.B(ii) Synthetic Matrices
In addition to the naturally-derived tissue-specific matrices described above, useful tissue-specific matrices may be formulated synthetically.
These porous biocompatible, in vivo biodegradable synthetic matrices are disclosed in PCT publication US91/03603, published December 12, 1991 (W091/18558). Briefly, the matrix comprises a porous crosslinked structural polymer of biocompatible, biodegradable collagen and appropriate, tissue-specific
glycosammoglycans as tissue-specific cell attachment factors. Collagen derived from a number of sources may be suitable for use in these synthetic matrices, including insoluble collagen, acid-soluble collagen, collagen soluble in neutral or basic aqueous solutions, as well as those collagens which are commercially available.
Glycosammoglycans (GAGs) or mucopolysaccharides are hexosamine-containing polysaccharides of animal origin that have a tissue specific distribution, and therefore may be used to help determine the tissue specificity of the morphogen-stimulated differentiating cells. Reaction with the GAGs also provides collagen with another valuable property, i.e., inability to provoke an immune reaction (foreign body reaction) from an animal host.
Chemically, GAGs are made up of residues of
hexosamines glycosidically bound and alternating in a more-or-less regular manner with either hexouronic acid or hexose moieties (see, e.g., Dodgson et al. in
Carbohydrate Metabolism and its Disorders (Dickens et al., eds.) Vol. 1, Academic Press (1968)). Useful GAGs include hyaluronic acid, heparin, heparin sulfate, chondroitin 6-sulfate, chondroitin 4-sulfate, dermatan sulfate, and keratin sulfate. Other GAGs are suitable for forming the matrix described herein, and those skilled in the art will either know or be able to ascertain other suitable GAGs using no more than routine experimentation. For a more detailed
description of mucopolysaccharides, see Aspinall,
Polysaccharides, Pergamon Press, Oxford (1970). For example, as disclosed in U.S. Application Serial
No. 529,852, chondroitin-6-sulfate can be used where endochondral bone formation is desired. Heparin sulfate, on the other hand, may be used to formulate synthetic matrices for use in lung tissue repair.
Collagen can be reacted with a GAG in aqueous acidic solutions, preferably in diluted acetic acid solutions. By adding the GAG dropwise into the aqueous collagen dispersion, coprecipitates of tangled collagen fibrils coated with GAG results. This tangled mass of fibers then can be homogenized to form a homogeneous dispersion of fine fibers and then filtered and dried.
Insolubility of the collagen-GAG products can be raised to the desired degree by covalently cross-linking these materials, which also serves to raise the resistance to resorption of these materials. In general, any covalent cross-linking method suitable for cross-linking collagen also is suitable for cross-linking these composite materials, although
crosslinking by a dehydrothermal process is preferred. When dry, the crosslinked particles are essentially spherical, with diameters of about 500 μm. Scanning electron miscroscopy shows pores of about 20 μm on the surface and 40 μm on the interior. The interior is made up of both fibrous and sheet-like structures, providing surfaces for cell attachment. The voids interconnect, providing access to the cells throughout the interior of the particle. The material appears to be roughly 99.5% void volume, making the material very efficient in terms of the potential cell mass that can be grown per gram of microcarrier.
Another useful synthetic matrix is one formulated from biocompatible, in vivo biodegradable synthetic polymers, such as those composed of glycolic acid, lactic acid and/or butyric acid, including copolymers and derivatives thereof. These polymers are well described in the art and are available commercially. For example, polymers composed of polyactic acid (e.g., MW 100 kDa), 80% polylactide/20% glycoside or poly 3-hydroxybutyric acid (e.g., MW 30 kDa) all may be purchased from PolySciences, Inc. The polymer
compositions generally are obtained in particulate form. In addition, one can alter the morphology of the polymer compositions, for example to increase porosity, using any of a number of particular solvent treatments known in the art. Where the morphogen is adsorbed to the matrix surface, the steps preferably are performed under conditions which avoid hydrolysis of the polymers (e.g., non-aqueous conditions such as in an ethanol-trifluoro-acetic acid solution).
The OP-3 proteins described herein can be combined and dispersed in a suitable matrix using any of the methods described below:
1. Ethanol Precipitation
Matrix is added to the morphogen dissolved in guanidine-HCl. Samples are vortexed and incubated at a low temperature. Samples are then further vortexed. Cold absolute ethanol is added to the mixture which is then stirred and incubated. After centrifugation
(microfuge, high speed) the supernatant is discarded. The matrix is washed with cold concentrated ethanol in water and then lyophilized. 2. Acetonitrile Trifluoroacetic Acid Lyophilization
In this procedure, a morphogenically active
fragment of OP-3 in an acetonitrile trifluroacetic acid (ACN/TFA) solution is added to the carrier material. Samples are vigorously vortexed many times and then lyophilized.
3. Buffered Saline Lyophilization
A preparation of a morphogenically active fragment of OP-3 in physiological saline also may be vortexed with the matrix and lyophilized to produce
morphogenically active material.
Tissue morphogenesis requires a morphogenically permissive environment. Clearly, in fully-functioning healthy tissue that is not composed of a permanently renewing cell population, there must exist signals to prevent continued tissue growth. Thus, it is
postulated that there exists a control mechanism, such as a feedback control mechanism, which regulates the control of cell growth and differentiation. In fact, it is known that both TGF-β, and MIS are capable of inhibiting cell growth when present at appropriate concentrations. In addition, using the bone model system it can be shown that osteogenic devices
comprising a bone-derived carrier (matrix) that has been demineralized and guanidine-extracted to
substantially remove the noncollagenous proteins does allow endochondral bone formation when implanted in association with an osteoinductive morphogen. If, however, the bone-derived carrier is not demineralized but rather is washed only in low salt, for example, induction of endochondral bone formation is inhibited, suggesting the presence of one or more inhibiting factors within the carrier.
III. Examples
Example 1. Recombinant Production of OP-3
OP-3 proteins useful in the methods and
compositions of this invention may be purified from natural sources or produced using standard recombinant methodology. General considerations for the
recombinant production of OP3 morphogens are described below. A. Identification of Novel mOP-3 Sequences
A genetic sequence encoding the morphogenic OP-3 protein was identified using a 0.3 kb EcoRI-BamHI OP-2 fragment from a mouse OP-2 cDNA as a hybridization probe, specific to the mid-pro region of OP-2
(corresponding to amino acid residues 125 to 225 of the pre-pro protein) essentially as described in
USSN 667,274. The 32P-labeled probe was prepared using the random hexanucleotide priming method, and the hybridizations were performed using the following conditions: 40% formamide, 5 X SSPE, 5 X Denhardt's Solution, 0.1% SDS, at 37°C overnight, and washing in 0.1 X SSPE, 0.1% SDS at 50°C. Approximately 1 X 106 phages from a mouse cDNA (carried in lambda zapll) library made from the teratocarcinoma cell line PCC4 (Stratagene, Inc., La Jolla, CA, cat # 936301) were screened. This screening yielded four individual clones which were purified over three rounds of
screening. The plasmid DNA containing the cDNAs was obtained using the lambda zapll excision process following manufacturer's directions. Three of the four clones were shown by DNA sequencing to encode OP-3. The DNA sequence, referred to herein as mOP-3 and described in Seq. ID No. 1, was identified by this procedure.
The isolated mOP-3 DNA sequence, in accordance with other known morphogens, encodes a protein comprising a "pro" region (defined essentially by residues 20-260 or 20-263 of Seq. ID No. 1) and a mature region (defined essentially by residues 261-399 or 264-399 of Seq. ID No. 1), including a functional domain comprising the conserved cysteine skeleton.
Like OP-2, OP-3 is marked by an eighth cysteine within the seven cysteine domain (e.g., at position 338 of Seq. ID No.l). The extra cysteine likely helps stabilize the folded structure, possibly by providing inter-molecular disulfide bonding. The extra cysteine also allows for heterodimer formation between OP-3 and another morphogen comprising the "eighth" cysteine, like OP-2 for example, or a modified OP-1, wherein an extra cysteine has been inserted at the appropriate location. The extra cysteine also may allow tetramer formation. The extra cysteine does not inhibit
synthesis or reduce the stability of the translated sequence significantly as expressed proteins comprising the extra cysteine are readily detected by SDS gel electrophoresis. A primary glycosylation site occurs just C terminal to the extra cysteine in both OP-2 and OP-3, which may provide a protective effect.
The cDNA sequences for both human and mouse OP-2 are provided in Seq. ID Nos. 7 and 9, and the genomic sequence for human OP-2 is provided in Seq. ID No. 11, wherein the exons defining the coding region of these proteins are indicated. The exon boundaries also are indicated in Fig. 1, described below. The human OP-2 locus was isolated from a genomic library (Clontech, EMBL-3 #HL1067J) on three overlapping phage clones, using standard cloning procedures. The OP-2 coding information was spread over 27 kb and, like OP-1, contains 7 exons. A comparison of exon-intron
boundaries in the 7 cysteine domain showed matching locations with those of OP-1. The first OP-2 exon contains 334 bp of coding sequence (111 amino acids), including the signal peptide, and is followed by the largest intron (14.6 kb). The second exon (190 bp, 64 amino acids) is separated by a short intron (0.4 kb) from exon 3 (149 bp, 49 amino acids). It follows a large third intron of 9.5 kb. The fourth exon (195 bp, 65 amino acids) encodes the maturation site
("OP-2-Ala") and is followed by a 0.8 kb intron. The 7 cysteine domains resides on exons 5 to 7: exon 5 (80 bp, 27 amino acids) encodes the first cysteine of mature OP-2 and is followed by intron 5 (0.5 kb in length), exon 6 (111 bp, 37 amino acids) is separated by a 2.5 kb intron from the seventh, last exon with 147 bp (49 amino acids) of coding sequence. As stated above, the exon-intron boundaries are conserved between human OP-1 and OP-2, two different members of the morphogen family of proteins. By analogy, the exonintron boundaries between human and mouse OP-2, two species variants of a morphogen, are anticipated to be conserved as well.
Figure 1 shows the alignment of the murine OP-2 and murine OP-3 coding regions of the cDNA. The exon boundaries are indicated by bars beneath the sequence. Both sequences have the same number of nucleotides.
The nucleotide sequence is about 80% conserved in the N-terminal and C-terminal regions. In the figure, nucleotide identity between the sequences is indicated by stippling. In addition, the central region of the sequence is highly conserved and this conserved region falls into the boundaries of exon 2 and 3. There are only three nucleotide differences in this region, indicated in the figure by diamonds. The high degree of conservation in the nucleotide sequences indicates that OP-2 and OP-3 likely share the nucleotide sequence of exon 2 and 3. The different proteins may result from alternatively spliced
transcripts, or they may arise from independent genes which share part of their coding sequence. Intron 1, which lies upstream of exon 2 in OP-2 (see Seq. ID No.11) is large (14.6kb) and could include the start of the OP-3 gene and/or its first exon sequence.
Certainly, as has been found for other mammalian genes, one or more of the introns of these morphogens may include sequences having a transcription regulatory function. Using the screening procedure described herein and in USSN 752,764, and the labelled OP-2 fragment, or preferably a labelled OP-3 fragment, OP-3 genetic sequences from other species and other libraries may be isolated. Alternatively, or in addition, a probe to the N-terminal region of the mature protein, or the 3' noncoding region flanking and immediately following the stop codon, also may be used to screen for other OP-3 species variants. These sequences vary substantially among the morphogens and represent morphogen-specific sequences. Mammalian cell expression of OP-3 readily can be achieved using COS (simian kidney ATCC, CRL-1650) or CHO (Chinese hamster ovary) cells (e.g., CHO-DXBII, from Lawrence Chasin, Columbia University, NY). An exemplary protocol for mammalian cell expression is provided below. Other useful eukaryotic cell systems include the insect/baculovirus system or the mammalian complement system. B. Expression of Novel OP-3 Sequences
To express the OP-3 protein, the OP-3 DNA is subcloned into an insertion site of a suitable, commercially available pUC-type vector (e.g., pUC-19, ATCC #37254, Rockville, MD), along with a suitable promoter/enhancer sequences and 3' termination
sequences . Currently preferred promoter/enhancer sequences are the CMV-MIE promoter (human
cytomegalovirus major intermediate-early promoter, preferably the intron-free or "short" form of the promoter) and the mouse mammary tumor virus promoter (mMTV) boosted by the rous sarcoma virus LTR enhancer sequence (e.g., from Clontech, Inc., Palo Alto).
Expression also may be further enhanced using
transactivating enhancer sequences. The plasmid also preferably contains a selectable marker, most
preferably an amplifiable marker such as DHFR, e.g., under SV40 early promoter control (ATCC #37148).
Transfection, cell culturing, gene amplification and protein expression conditions are standard conditions, well known in the art, such as are described, for example in Ausubel et al., ed.. Current Protocols in Molecular Biology, John Wiley & Sons, NY (1989).
Briefly, transfected cells are cultured in medium containing 0.1-0.5% dialyzed fetal calf serum (FCS), stably transfected high expression cell lines obtained by subcloning and evaluated by standard Northern blot. Southern blots also are used to assess the state of integrated OP-3 sequences and the extent of their copy number amplification . Chimeric OP-3 morphogens, e.g., comprising an OP-3 active domain and, for example, part or all of a pro domain from another, different morphogen may be
constructed using standard recombinant DNA technology and/or an automated DNA synthesizer to construct the desired sequence. Useful chimeras include those wherein the non-OP-3 sequence is joined to the OP-3 sequence encoding the mature OP-3 protein, and the non-OP-3 sequence encodes part or all of the sequence between the signal peptide processing site and the
"Arg-Xaa-Xaa-Arg" processing sequence from at least one morphogen. Alternatively, the non-OP-3 sequence may be joined to an OP-3 sequence encoding, for example, the 6 or 7 cysteine skeletons, wherein the non-OP-3 sequence includes the sequence encoding the N-terminus of the mature protein. As will be appreciated by persons skilled in the art, the non-OP-3 sequences may be composed of sequences from one or morphogens and/or may comprise novel biosynthetic sequences.
Mammalian expression of a biosynthetic gene
construct encoding a chimeric OP1-OP3 polypeptide chain is demonstrated in the immunoblot presented in Fig. 2. A vector carrying the construct under CMV promoter control was transfected into CHO cells (CHO-DXB11) using standard procedures and as described herein.
A chimeric gene was constructed by replacing the conserved seven cysteine domain of OP-1 with that of OP-3. The resulting chimeric gene contains the entire pre-pro-domain of human OP-1 and the region of mature OP-1 between the maturation site and the first cysteine of the conserved C-terminal seven cysteine domain, fused to the conserved seven cysteine domain of mouse OP-3, but with two arginine residues in place of the native lysine residues found in OP-3 at the start of the seven cysteine domain.
The gene fusion was accomplished by splicing the Sad site of OP-3 (near the first cysteine of the seven cysteine domain) with a newly created Sad site in OP-1, created at the matching residues by silent mutagenesis. The Sad site encodes the Glu-Leu
dipeptide in the sequence Cys-Arg-Arg-His-Glu-Leu of OP-1 and Cys-Lys-Lys-His-Glu-Leu of OP-3, respectively.
The chimeric gene was placed downstream of the CMV (Cytomegalovirus) MIE "short" (intron-free) promoter and upstream of the SV40 transcriptional terminator in a pUC vector. This plasmid was cotransfected with DNA encoding the DHFR marker and viral trans-activating elements (e.g., VA1, E1A) into a CHO dhfr(-) host and subjected to Methotrexate selection and one round of amplification at 1 mM Methotrexate including
subcloning. 10 μl of "spent" culture supernatant
(3 days old) was analysed by "Western blot"
( immunoblot), as follows.
The 10 μl harvested medium was briefly heated with concentrated SDS sample buffer, containing β-mercapto ethanol (5%) and directly analysed by electrophoresis on a 15% SDS- polyacrylamide gel (in the buffer system of Laemmli) along with a set of prestained molecular weight standards (Bio-rad, Richmond, CA). Proteins were transferred from the gel to Immobilon membrane by the "Western blot" procedure. The chimeric OP-1/OP-3 protein was detected by reaction with rabbit serum raised against a synthetic peptide representing the first 17 amino acids of mature OP-1, starting with serine-threonine-glycine-serine-. Authentic
recombinant OP-1, expressed in CHO cells was included for comparison. In the figure sample lanes were as follows: lane 1: OP-1; lanes 4, 5, 6, 7, and 8:
chimeric OP-1/OP-3; lanes 9 and 10: prestained
molecular weight standards. The apparent mobility of the recombinant proteins, at approximately 20 kDa on this gel, is due to glycosylation of the OP-1 and OP-3 proteins which may also be the cause of the multiple species observed.
The expressed protein then can be purified as follows. For a typical 2L preparation of transfected mammalian cells conditioned in 0.5% FCS, for example, the total protein is typically about 700 mg. The amount of OP-3 in the media, estimated by Western blot, is between about 0.1-5.0 mg. OP-3 media then is diluted in a low salt, physiologically buffered 6M urea solution, and loaded onto an S-Sepharose column, which acts as a strong cation exchanger. OP-3 binds to the column in low salt, and serum proteins are removed. The column subsequently is developed with an NaCl gradient, e.g., 0.1M NaCl-1.0M NaCl, in 6M urea, 20mM HEPES, pH 7.0. Most contaminants are removed at the start of the gradient, and OP-3 is eluted primarily at a higher salt concentration. The sample then is loaded onto a phenyl-Sepharose column (hydrophobic interaction chromatography). OP-3 binds phenyl-Sepharose in the presence of high
concentrations of a weak chaotropic salt (e.g., IM (NH4)2SO4 in a physiologically buffered 6M urea
solution). Once OP-3 is bound, the column is developed with a decreasing ammonium sulfate gradient, e.g., 0.6M-0.0M (NH4)2SO4 gradient in a physiologically buffered, 6M urea solution. Again, most contaminants are removed at the start of the gradient, and OP-3 elutes primarily at low or no ammonium sulfate
concentrations.
The 0P-3 eluted from the phenyl-Sepharose column then is dialyzed against water, and prepared for loading onto a reverse phase chromatography column (e.g., C-18 HPLC), for example, by dialyzing against 30% acetonitrile, 0.1% TFA. An alternative chromatography protocol is to perform the S-Sepharose chromatography in the absence of 6 M urea. The bound proteins then are eluted with salt step elutions (e.g., 0.1-0.6M NaCl). Remaining OP-3 then can be eluted in the presence of 6M urea. The 6M urea elution also may be used in place of the non-urea elution to achieve maximum recovery in one step. In addition, OP-3 may be eluted from the phenylSepharose column in 38% ethanol-0.01% TFA, thereby eliminating the need to dialyze the eluent before applying it to the C-18 column. Finally, multiple C-18 columns may be used (e.g., three), to further enhance purification and concentration of the protein. OP-3 also will bind hydroxyapatite efficiently, typically in the absence of 6 M urea and at low
phosphate concentrations (less than 5 mM phosphate). Bound OP-3 can be removed from the column with an elution gradient of about .001-0.5M step elution of phosphate in a physiologically buffered solution.
Additionally, urea (6M) may be added during the elution step. Other related chromatography methods also may be useful in purifying OP-3 from eucaryotic cell culture systems. For example, heparin-Sepharose may be used in combination with the S-Sepharose column.
Alternatively, immobilized metal-ion affinity
chromatography (IMAC) (e.g., Cu2+ or Zn+ ) and a
physiologically buffered phosphate solution may be used to advantage.
C. Soluble OP3 Complexes
A currently preferred form of the OP-3 morphogen useful in therapeutic formulations, having improved solubility in aqueous solutions and consisting
essentially of amino acids, is a dimeric morphogenic protein comprising at least the 100 amino acid peptide sequence having the pattern of seven or more cysteine residues characteristic of the morphogen family
complexed with a peptide comprising part or all of a pro region of a member of the morphogen family, or an allelic, species or other sequence variant thereof. Preferably, the dimeric morphogenic protein is
complexed with two peptides. Also, the dimeric
morphogenic protein preferably is noncovalently
complexed with the pro region peptide or peptides. The pro region peptides also preferably comprise at least the N-terminal eighteen amino acids that define the OP-3 morphogen pro region (e.g., residues 18-35 of Seq. ID No. 1). In a most preferred embodiment, peptides defining substantially the full length pro region are used.
Other soluble forms of morphogens include dimers of the uncleaved pro forms of these proteins, as well as "hemi-dimers" wherein one subunit of the dimer is an uncleaved pro form of the protein, and the other subunit comprises the mature form of the protein, including truncated forms thereof, preferably
noncovalently associated with a cleaved pro domain peptide.
As described above, useful pro domains include the full length pro regions, as well as various truncated forms hereof, particularly truncated forms cleaved at proteolytic Arg-Xaa-Xaa-Arg cleavage sites. In OP-3, possible pro sequences cleaved at Arg-Xaa-Xaa-Arg sites include sequences defined by residues 18-260 of Seq. ID No. 1 (anticipated full length form); or by residues 18-263. Accordingly, currently preferred pro sequences are those encoding the full length form of the pro region for OP-3 or another, known morphogen. Other pro sequences contemplated to have utility include
biosynthetic pro sequences, particularly those that incorporate a sequence derived from the N-terminal portion of one or more morphogen pro sequences.
As will be appreciated by those having ordinary skill in the art, useful sequences encoding the pro region may be obtained from genetic sequences encoding known morphogens. Alternatively, chimeric pro regions can be constructed from the sequences of one or more known morphogens. Still another option is to create a synthetic sequence variant of one or more known pro region sequences. In another preferred aspect, useful pro region peptides include polypeptide chains comprising an amino acid sequence encoded by a nucleic acid that hybridizes under stringent conditions with a DNA or RNA sequence encoding at least the N-terminal eighteen amino acids of the pro region sequence for OP-3 e.g., nucleotides 120-173 of Seq. ID No. 1.
In yet another preferred aspect, useful pro region peptides include polypeptide chains comprising an amino acid sequence encoded by a nucleic acid that hybridizes under stringent conditions with a DNA or RNA sequence encoding at least the N-terminal eighteen amino acids of the pro region sequence for OP1 or OP2, e.g., nucleotides 136-192 and 152-211 of Seq. ID No. 3 and 7, respectively. C.1. Isolation of Soluble morphogen complex from conditioned media or body fluid
Morphogens are expressed from mammalian cells as soluble complexes. Typically, however the complex is disassociated during purification, generally by
exposure to denaturants often added to the purification solutions, such as detergents, alcohols, organic solvents, chaotropic agents and compounds added to reduce the pH of the solution. Provided below is a currently preferred protocol for purifying the soluble proteins from conditioned media (or, optionally, a body fluid such as serum, cerebro-spinal or peritoneal fluid), under non-denaturing conditions. The method is rapid, reproducible and yields isolated soluble
morphogen complexes in substantially pure form.
Soluble OP-3 morphogen complexes can be isolated from conditioned media using a simple, three step chromatographic protocol performed in the absence of denaturants. The protocol involves running the media (or body fluid) over an affinity column, followed by ion exchange and gel filtration chromatographies. The affinity column described below is a Zn-IMAC column. The present protocol has general applicability to the purification of a variety of morphogens, all of which are anticipated to be isolatable using only minor modifications of the protocol described below. An alternative protocol also envisioned to have utility an immunoaffinity column, created using standard
procedures and, for example, using antibody specific for a the OP-3 pro domain (complexed, for example, to a protein A-conjugated Sepharose column.) Protocols for developing immunoaffinity columns are well described in the art, (see, for example. Guide to Protein
Purification, M. Deutscher, ed.. Academic Press, San Diego, 1990, particularly sections VII and XI.)
In this experiment OP-1 was expressed in mammalian CHO (Chinese hamster ovary) cells as described in the art (see, for example, international application
US90/05903 (WO91/05802).) The CHO cell conditioned media containing 0.5% FBS was initially purified using Immobilized Metal-Ion Affinity Chromatography (IMAC). The soluble OP-1 complex from conditioned media binds very selectively to the Zn-IMAC resin and a high concentration of imidazole (50 mM imidazole, pH 8.0) is required for the effective elution of the bound
complex. The Zn-IMAC step separates the soluble OP-1 from the bulk of the contaminating serum proteins that elute in the flow through and 35 mM imidazole wash fractions. The Zn-IMAC purified soluble OP-1 is next applied to an S-Sepharose cation-exchange column equilibrated in 20 mM NaPO4 (pH 7.0) with 50 mM NaCl. This S-Sepharose step serves to further purify and concentrate the soluble OP-1 complex in preparation for the following gel filtration step. The protein was applied to a Sephacryl S-200HR column equilibrated in TBS. Using substantially the same protocol, soluble morphogens also may be isolated from one or more body fluids, including serum, cerebro-spinal fluid or peritoneal fluid. IMAC was performed using Chelating-Sepharose
(Pharmacia) that had been charged with three column volumes of 0.2 M ZnSO4. The conditioned media was titrated to pH 7.0 and applied directly to the ZN-IMAC resin equilibrated in 20 mM HEPES (pH 7.0) with 500 mM NaCl. The Zn-IMAC resin was loaded with 80 mL of starting conditioned media per mL of resin. After loading, the column was washed with equilibration buffer and most of the contaminating proteins were eluted with 35 mM imidazole (pH 7.0) in equilibration buffer. The soluble OP-1 complex then is eluted with 50 mM imidazole (pH 8.0) in 20 mM HEPES and 500 mM NaCl. The 50 mM imidazole eluate containing the soluble OP-1 complex was diluted with nine volumes of 20 mM NaPO. (pH 7.0) and applied to an S-Sepharose
(Pharmacia) column equilibrated in 20 mM NaPO4 (pH 7.0) with 50 mM NaCl. The S-Sepharose resin was loaded with an equivalent of 800 mL of starting conditioned media per mL of resin. After loading the S-Sepharose column was washed with equilibration buffer and eluted with 100 mM NaCl followed by 300 mM and 500 mM NaCl in 20 mM NaPO4 (pH 7.0). The 300 mM NaCl pool was further purified using gel filtration chromatography. Fifty mis of the 300 mm NaCl eluate was applied to a 5.0 X 90 cm Sephacryl S-200HR (Pharmacia) equilibrated in Tris buffered saline (TBS), 50 mM Tris, 150 mM NaCl
(pH 7.4). The column was eluted at a flow rate of 5 mL/minute collecting 10 mL fractions. The apparent molecular of the soluble OP-1 was determined by
comparison to protein molecular weight standards (alcohol dehydrogenase (ADH, 150 kDa), bovine serum albumin (BSA, 68 kDa), carbonic anhydrase (CA, 30 kDa) and cytochrome C (cyt C, 12.5 kDa). The purity of the S-200 column fractions was determined by separation on standard 15% polyacrylamide SDS gels stained with coomassie blue. The identity of the mature OP-1 and the pro-domain was determined by N-terminal sequence analysis after separation of the mature OP-1 from the pro-domain using standard reverse phase C18 HPLC.
The soluble OP-1 complex elutes with an apparent molecular weight of 110 kDa. This agrees well with the predicted composition of the soluble OP-1 complex with one mature OP-1 dimer (35-36 kDa) associated with two pro-domains (39 kDa each). Purity of the final complex can be verified by running the appropriate fraction in a reduced 15% polyacrylamide gel.
The complex components can be verified by running the complex-containing fraction from the S-200 or S- 200HR columns over a reverse phase C18 HPLC column and eluting in an acetonitrile gradient (in 0.1% TFA), using standard procedures. The complex is dissociated by this step, and the pro domain and mature species elute as separate species. These separate species then can be subjected to N-terminal sequencing using
standard procedures (see, for example. Guide to
Protein Purification, M. Deutscher, ed.. Academic
Press, San Diego, 1990, particularly pp. 602-613), and the identity of the isolated 36kD, 39kDa proteins confirmed as mature morphogen and isolated, cleaved pro domain, respectively. N-terminal sequencing of the isolated pro domain from mammalian cell produced OP-1 revealed 2 forms of the pro region, the intact form (beginning at residue 30 of Seq. ID No. 16) and a truncated form, (beginning at residue 48 of Seq. ID No. 16.) N-terminal sequencing of the polypeptide subunit of the isolated mature species reveals a range of
N-termini for the mature sequence, beginning at
residues 293, 300, 313, 315, 316, and 318, of Seq. ID No. 16, all of which are active as demonstrated by the standard bone induction assay.
C.2. In Vitro Soluble Morphogen Complex Formation
As an alternative to purifying soluble complexes from culture media or a body fluid, soluble complexes may be formulated from purified pro domains and mature dimeric species. Successful complex formation
apparently requires association of the components under denaturing conditions sufficient to relax the folded structure of these molecules, without affecting
disulfide bonds. Preferably, the denaturing conditions mimic the environment of an intracelluiar vesicle sufficiently such that the cleaved pro domain has an opportunity to associate with the mature dimeric species under relaxed folding conditions. The
concentration of denaturant in the solution then is decreased in a controlled, preferably step-wise manner, so as to allow proper refolding of the dimer and pro regions while maintaining the association of the pro domain with the dimer. Useful denaturants include 4-6M urea or guanidine hydrochloride (GuHCl), in buffered solutions of pH 4-10, preferably pH 6-8. The soluble complex then is formed by controlled dialysis or dilution into a solution having a final denaturant concentration of less than 0.1-2M urea or GuHCl, preferably 1-2 M urea of GuHCl, which then preferably can be diluted into a physiological buffer. Protein purification/renaturing procedures and considerations are well described in the art, and details for
developing a suitable renaturing protocol readily can be determined by one having ordinary skill in the art. One useful text one the subject is Guide to Protein Purification, M. Deutscher, ed.. Academic Press, San Diego, 1990, particularly section V. Complex formation also may be aided by addition of one or more chaperone proteins.
C3. Stability of Soluble Morphogen Complexes
The stability of the highly purified soluble morphogen complex in a physiological buffer, e.g., tris-buffered saline (TBS) and phosphate-buffered saline (PBS), can be enhanced by any of a number of means. Currently preferred is by means of a pro region that comprises at least the first 18 amino acids of the pro sequence (e.g., residues 18-35 of Seq. ID NO. 1 for OP-3), and preferably is the full length pro region. Residues 18-35 show sequence homology to the N-terminal portion of other morphogens and are believed to have particular utility in enhancing complex stability for all morphogens. Other useful means for enhancing the stability of soluble morphogen complexes include three classes of additives. These additives include basic amino acids (e.g., L-arginine, lysine and betaine); nonionic detergents (e.g., Tween 80 or Nonidet P-120); and carrier proteins (e.g., serum albumin and casein). Useful concentrations of these additives include 1-100 mM, preferably 10-70 mM, including 50 mM, basic amino acid;, 0.01-1.0%, preferably 0.05-0.2%, including 0.1% (v/v) nonionic detergent;, and 0.01-1.0%, preferably 0.05-0.2%, including 0.1% (w/v) carrier protein.
Example 2. Mitogenic Effect of OP-3
2.1 Mitogenic Effect of Morphogen on Rat and
Human Osteoblasts
The following example can be used to demonstrate the ability of OP-3 to induce proliferation of
osteoblasts in vitro using the following assay. In this and all examples involving osteoblast cultures, rat osteoblast-enriched primary cultures preferably are used. Although these cultures are heterogeneous in that the individual cells are at different stages of differentiation, the culture is believed to more accurately reflect the metabolism and function of osteoblasts in vivo than osteoblast cultures obtained from established cell lines. Unless otherwise
indicated, all chemicals referenced are standard, commercially available reagents, readily available from a number of sources, including Sigma Chemical, Co., St. Louis; Calbiochem, Corp., San Diego and Aldrich
Chemical Co., Milwaukee.
Rat osteoblast-enriched primary cultures are prepared by sequential collagenase digestion of newborn suture-free rat calvaria (e.g., from 1-2 day-old animals, Long-Evans strain, Charles River Laboratories, Wilmington, MA), following standard procedures, such as are described, for example, in Wong et al., (1975) PNAS 72:3167-3171. Rat osteoblast single cell suspensions then are plated onto a multi-well plate (e.g., a
24 well plate) at a concentration of 50,000 osteoblasts per well in alpha MEM (modified Eagle's medium, Gibco, Inc., Long Island) containing 10% FBS (fetal bovine serum), L-glutamine and penicillin/streptomycin. The cells are incubated for 24 hours at 37°C, at which time the growth medium is replaced with alpha MEM containing 1% FBS and the cells incubated for an additional
24 hours so that cells are in serum-deprived growth medium at the time of the experiment.
The cultured cells are divided into three groups: (1) wells which receive, for example, 0.1, 1.0, 10.0, 40 and 80.0 ng of OP-3; (2) wells which receive 0.1, 1.0, 10.0 and 40 ng of a local-acting growth factor (e.g., TGF-β); and (3) the control group, which receive no growth factors. The cells then are incubated for an additional 18 hours after which the wells are pulsed with 2μCi/well of 3 H-thymidine and incubated for six more hours. The excess label then is washed off with a cold solution of 0.15 M NaCl and then 250 μl of 10% tricholoracetic acid is added to each well and the wells incubated at room temperature for 30 minutes. The cells then are washed three times with cold
distilled water, and lysed by the addition of 250 μl of 1% sodium dodecyl sulfate (SDS) for a period of
30 minutes at 37°C The resulting cell lysates are harvested using standard means well known in the art, and the incorporation of 3 H-thymidine into cellular DNA determined by liquid scintillation as an indication of mitogenic activity of the cells. In the experiment, OP-3 is anticipated to stimulate 3 H-thymidine
incorporation into DNA, and thus promote osteoblast cell proliferation. By contrast, the effect of TGF-β is transient and biphasic. At high concentrations, TGF-β has no significant effect on osteoblast cell proliferation.
The in vitro effect of OP-3 on osteoblast
proliferation also may be evaluated using human primary osteoblasts (obtained from bone tissue of a normal adult patient and prepared as described above) and on human osteosarcoma-derived cell lines. In all cases OP-3 is anticipated to induce cell proliferation in accordance with the morphogen's ability to induce endochondral bone formation (see Example 7, below). 2.2 Progenitor Cell Stimulation
The following example demonstrates the ability of OP-3 to stimulate the proliferation of mesenchymal progenitor cells. Useful naive stem cells include pluripotential stem cells, which may be isolated from bone marrow or umbilical cord blood using conventional methodologies, (see, for example. Faradji et al.,
(1988) Vox Sang., 55 (3): 133-138 or Broxmeyer et al.,
(1989) PNAS 86 : 3828-3832), as well as naive stem cells obtained from blood. Alternatively, embryonic cells
(e.g., from a cultured mesodermal cell line) may be useful.
Another method for obtaining progenitor cells and for determining the ability of OP-3 fragments to stimulate cell proliferation is to capture progenitor cells from an in vivo source. For example, a
biocompatible matrix material able to allow the influx of migratory progenitor cells may be implanted at an in vivo site long enough to allow the influx of migratory progenitor cells. For example, a bone-derived, guanidine-extracted matrix, formulated as disclosed for example in Sampath et al. ((1983) PNAS 80:6591-6595), or U.S. Patent No. 4,975,526, may be implanted into a rat at a subcutaneous site, essentially following the method of Sampath et al. After three days the implant is removed, and the progenitor cells associated with the matrix dispersed and cultured.
Progenitor cells, however obtained, then are incubated in vitro with OP-3 under standard cell culture conditions well described in the art and described hereinabove. In the absence of external stimuli, the progenitor cells do not, or only
minimally, proliferate on their own in culture.
However, progenitor cells cultured in the presence of a morphogenically active fragment of OP-3 are anticipated to proliferate. Cell growth can be determined visually or spectrophotometrically using standard methods well known in the art.
Example 3. Morphogen-Induced Cell Differentiation 3.1 Embryonic Mesenchyme Differentiation
Morphogenically active fragments of OP-3 can be utilized to induce cell differentiation. The ability of OP-3 to induce cell differentiation can be
demonstrated by culturing early mesenchymal cells in the presence of OP-3 and then studying the histology of the cultured cells by staining with toluidine blue using standard cell culturing and cell staining methodologies well described in the art. For example, it is known that rat mesenchymal cells destined to become mandibular bone, when separated from the overlying epithelial cells at stage 11 and cultured in vitro under standard tissue culture conditions, e.g., in a chemically defined, serum-free medium, containing for example, 67% DMEM (Dulbecco's modified Eagle's medium), 22% F-12 medium, 10mM Hepes pH 7, 2mM
glutamine, 50 μg/ml transferrin, 25 μg/ml insulin, trace elements, 2mg/ml bovine serum albumin coupled to oleic acid, with HAT (0.1 mM hypoxanthine, lOμM
aminopterin, 12 μM thymidine, will not continue to differentiate. However, if these same cells are left in contact with the overlying endoderm for an
additional day, at which time they become stage
12 cells, they will continue to differentiate on their own in vitro to form chondrocytes. Further
differentiation into osteoblasts and, ultimately, mandibular bone, requires an appropriate local
environment, e.g., a vascularized environment.
Stage 11 mesenchymal cells, cultured in vitro in the presence of OP-3, e.g., 10-100 ng/ml, are
anticipated to continue to differentiate in vitro to form chondrocytes just as they continue to
differentiate in vitro if they are cultured with the cell products harvested from the overlying endodermal cells. This experiment may be performed with different mesenchymal cells to demonstrate the cell
differentiation capability of OP-3 in different
tissues. As another example of morphogen-induced cell differentiation, the ability of OP-3 to induce
osteoblast differentiation may be demonstrated in vitro using primary osteoblast cultures, or osteoblast-like cells lines, and assaying for a variety of bone cell markers that are specific markers for the
differentiated osteoblast phenotype, e.g., alkaline phosphatase activity, parathyroid hormone-mediated cyclic AMP (cAMP) production, osteocalcin synthesis, and enhanced mineralization rates.
3.2 Alkaline Phosphatase Induction of
Osteoblasts by OP-3 The cultured cells in serum-free medium are
incubated with, a range of OP-3 concentrations, for example, 0.1, 1.0, 10.0, 40.0 or 80.0 ng OP-3/ml medium; or with a similar range of TGF-β
concentrations. 72 hours after the incubation period the cell layer is extracted with 0.5 ml of 1% Triton X-100. The resultant cell extract then, is
centrifuged, and 100 μl of the extract is added to 90 μl of paranitrosophenylphospate (PNPP)/glycerine mixture and incubated for 30 minutes in a 37°C water bath and the reaction stopped with 100 μl NaOH. The samples then are run through a plate reader (e.g., Dynatech MR700 plate reader, and absorbance measured at 400 nm, using p-nitrophenol as a standard) to determine the presence and amount of alkaline phosphate activity. Protein concentrations are determined by the Biorad method. Alkaline phosphatase activity is calculated in units/μg protein, where 1 unit=1 nmol p-nitrophenol liberated/30 minutes at 37°C OP-3 alone stimulates the production of alkaline phosphatase in osteoblasts, and thus promotes the growth and expression of the osteoblast differentiated phenotype.
The long term effect of OP-3 morphogen on the production of alkaline phosphatase by rat osteoblasts also may be demonstrated as follows. Rat osteoblasts are prepared and cultured in multiwell plates as described above. In this example six sets of 24 well plates are plated with 50,000 rat osteoblasts per well. The wells in each plate,
prepared as described above, then are divided into three groups: (1) those which receive, for example, 1 ng of OP-3 per ml of medium; (2) those which receive 40 ng of OP-3 per ml of medium; and (3) those which received 80 ng of OP-3 per ml of medium. Each plate then is incubated for different lengths of time:
0 hours (control time), 24 hours, 48 hours, 96 hours, 120 hours and 144 hours. After each incubation period, the cell layer is extracted with 0.5 ml of 1% Triton X-100. The resultant cell extract is centrifuged, and alkaline phosphatase activity determined as for
Example 3.1, using paranitroso-phenylphosphate (PNPP). OP-3 stimulates the production of alkaline phosphatase in osteoblasts in dose-dependent manner so that
increasing doses of OP-3 further increase the level of alkaline phosphatase production, and moreover, the OP-3-stimulated elevated levels of alkaline phosphatase in the treated osteoblasts is anticipated to last for an extended period of time. 3.3 OP-3 Protein Induction of PTH-Mediated cAMP.
The effect of a OP-3 on parathyroid hormone-mediated cAMP production in rat osteoblasts in vitro may be demonstrated as follows.
Rat osteoblasts are prepared and cultured in a multiwell plate as described above. The cultured cells then are divided into three groups: (1) wells which receive, for example, 1.0, 10.0 and 40.0 ng OP-3/ml medium); (2) wells which receive for example, TGF-β, at similar concentration ranges; and (3) a control group which receives no growth factors. The plate is then incubated for another 72 hours. At the end of the 72 hours the cells are treated with medium containing 0.5% bovine serum albumin (BSA) and ImM 3-isobutyl-1-methylxanthine for 20 minutes followed by the addition into half of the wells of human recombinant parathyroid hormone (hPTH, Sigma, St. Louis) at a concentration of 200 ng/ml for 10 minutes. The cell layer then is extracted from each well with 0.5 ml of 1% Triton
X-100. The cAMP levels then are determined using a radioimmunoassay kit (e.g., Amersham, Arlington
Heights, Illinois). OP-3 alone stimulates an increase in the PTH-mediated cAMP response, and thus promotes the growth and expression of the osteoblast
differentiated phenotype. 3.4 OP-3 Protein Induction of Osteocalcin
Production
Osteocalcin is a bone-specific protein synthesized by osteoblasts which plays an integral role in the rate of bone mineralization in vivo. Circulating levels of osteocalcin in serum are used as a marker for
osteoblast activity and bone formation in vivo.
Induction of osteocalcin synthesis in osteoblastenriched cultures can be used to demonstrate
OP-3 morphogenic efficacy in vitro.
Rat osteoblasts are prepared and cultured in a multi-well plate as above. In this experiment the medium is supplemented with 10%FBS, and on day 2, cells are fed with fresh medium supplemented with fresh 10 mM β-glycerophosphate (Sigma, Inc.). Beginning on day 5 and twice weekly thereafter, cells are fed with a complete mineralization medium containing all of the above components plus fresh L(+)-ascorbate, at a final concentration of 50μg/ml medium. OP-3 then is added to the wells directly, e.g., in 50% acetonitrile (or 50% ethanol) containing 0.1% trifluoroacetic acid (TFA), at no more than 5μl morphogen/ml medium. Control wells receive solvent vehicle only. The cells then are re-fed and the conditioned medium sample diluted 1:1 in standard radioimmunoassay buffer containing standard protease inhibitors and stored at -20° C until assayed for osteocalcin. Osteocalcin synthesis is measured by standard radioimmunoassay using a commercially
available osteocalcin-specific antibody. Mineralization is determined on long term cultures (13 day) using a modified von Kossa staining technique on fixed cell layers: cells are fixed in fresh 4% paraformaldehyde at 23° C for 10 min, following rinsing cold 0.9% NaCl. Fixed cells then are stained for endogenous alkaline phosphatase at pH 9.5 for 10 min, using a commercially available kit (Sigma, Inc.)
Purple stained cells then are dehydrated with methanol and air dried, after 30 min incubation in 3% AgNO3 in the dark, H2O-rinsed samples are exposed for 30 sec to 254 nm UV light to develop the black silver-stained phosphate nodules. Individual mineralized foci (at least 20 μm in size) are counted under a dissecting microscope and expressed as nodules/culture.
OP-3 stimulates osteocalcin synthesis in osteoblast cultures. The increased osteocalcin synthesis in response to OP-3 is dose dependent and shows a
significant increase over the basal level after 13 days of incubation. The enhanced osteocalcin synthesis also can be confirmed by detecting the elevated osteocalcin mRNA message (20-fold increase) using a rat
osteocalcin-specific probe. In addition, the increase in osteoclacin synthesis correlates with increased mineralization in long term osteoblast cultures as determined by the appearance of mineral nodules. OP-3 increases the initial mineralization rate significantly compared to untreated cultures. 3.5 Morphogen-Induced CAM Expression
The morphogens described herein induce CAM
expression, particularly N-CAM expression, as part of their induction of morphogenesis (see copending
USSN 922,813). CAMs are morphoregulatory molecules identified in all tissues as an essential step in tissue development. N-CAMs, which comprise at least 3 isoforms (N-CAM-180, N-CAM-140 and N-CAM-120, where "180", "140" and "120" indicate the apparent molecular weights of the isoforms as measured by SDS
polyacrylamide gel electrophoresis) are expressed at least transiently in developing tissues, and
permanently in nerve tissue. Both the N-CAM-180 and N-CAM-140 isoforms are expressed in both developing and adult tissue. The N-CAM-120 isoform is found only in adult tissue. Another neural CAM is L1.
The ability of OP-3 to stimulate CAM expression can be demonstrated using the following protocol, using
NG108-15 cells. NG108-15 is a transformed hybrid cell line (neuroblastoma × glioma, America Type Tissue
Culture (ATCC), Rockville, MD), exhibiting a morphology characteristic of transformed embryonic neurons. As described in Example 4, below, untreated NG108-15 cells exhibit a fibroblastic, or minimally differentiated, morphology and express only the 180 and 140 isoforms of N-CAM normally associated with a developing cell.
Following morphogen treatment these cells exhibit a morphology characteristic of adult neurons and express enhanced levels of all three N-CAM isoforms. In this example, NG108-15 cells are cultured for 4 days in the presence of increasing concentrations of OP-3 using standard culturing procedures, and standard Western blots performed on whole cell extracts. N-CAM isoforms are detected with an antibody which
crossreacts with all three isoforms, mAb H28.123, obtained from Sigma Chemical Co., St. Louis, the different isoforms being distinguishable by their different mobilities on an electrophoresis gel.
Control NG108-15 cells (untreated) express both the 140 kDa and the 180 kDa isoforms, but not the 120 kDa, as determined by Western blot analyses using up to 100 μg of protein. Treatment of NG108-15 cells with OP-3 results in a dose-dependent increase in the expression of the 180 kDa and 140 kDa isoforms, as well as the induction of the 120 kDa isoform induced. In addition, OP-3-induced CAM expression correlates with cell aggregation, as determined by histology. Example 4. OP-3 Protein-Induced Redifferentiation of
Transformed Phenotype
The OP-3 morphogens described herein also can induce redifferentiation of transformed cells to a morphology characteristic of untransformed cells. The examples provided below detail morphogen-induced redifferentiation of a transformed human cell line of neuronal origin (NG108-15); as well as mouse
neuroblastoma cells (N1E-115), and human embryo
carcinoma cells, to a morphology characteristic of untransformed cells. As described above, NG108-15 is a transformed hybrid cell line produced by fusing neuroblastoma × glioma cells (obtained from ATTC, Rockville, MD), and exhibiting a morphology characteristic of transformed embryonic neurons, e.g., having a fibroblastic
morphology. Specifically, the cells have polygonal cell bodies, short, spike-like processes and make few contacts with neighboring cells (see copending
USSN 922,813). Incubation of NG108-15 cells, cultured in a chemically defined, serum-free medium, with 0.1 to 300 ng/ml of morphogen (e.g; OP-3) for four hours is anticipated to induce an orderly, dose-dependent change in cell morphology. In the example, NG108-15 cells are subcultured on poly-L-lysine coated 6 well plates. Each well contains 40-50,000 cells in 2.5 ml of chemically defined medium. On the third day, 2.5 μl of morphogen (e.g., OP-3) in 60% ethanol containing 0.025% trifluoroacetic is added to each well. Morphogenic OP-3 of varying
concentrations are tested (typically, concentration ranges of 0-300 ng/ml are tested). The media is changed daily with new aliquots of morphogen. OP-3 is anticipated to induce a dose-dependent
redifferentiation of the transformed cells, including a rounding of the soma, an increase in phase brightness, extension of the short neurite processes, and other significant changes in the cellular ultrastructure.
After several days treated cells should begin to form epithelioid sheets that then become highly packed, multi-layered aggregates, as determined visually by microscopic examination. Moreover, morphogen-induced redifferentiation occurs without any associated changes in DNA synthesis, cell division, or cell viability, making it unlikely that the morphologic changes are secondary to cell differentiation or a toxic effect of the morphogen. In addition, the morphogen-induced redifferentiation does not inhibit cell division, as determined by
3H-thymidine uptake, unlike other molecules which have been shown to stimulate differentiation of transformed cells, such as butyrate, DMSO, retanoic acid or
Forskolin in analogous experiments. Thus, OP-3
maintains cell stability and viability after inducing redifferentiation. The OP-3 morphogens described herein accordingly provide useful therapeutic agents for the treatment of neoplasias and neoplastic lesions of the nervous system, particularly in the treatment of
neuroblastomas, including retinoblastomas, and gliomas.
As yet another, related example, the ability of OP-3 to induce the "redifferentiation" of transformed human cells may be demonstrated using the following assay. Specifically, the effect of OP-3 on human EC cells (embryo carcinoma cells, e.g., NTERA-Z CL.D1,
ATCC, Rockville, MD) may be determined. In the absence of an external stimulant, these cells can be maintained as undifferentiated stem cells, and can be induced to grow in serum free media (SFM). In the absence of treatment with a morphogen, the cells proliferate rampantly and are anchorage-independent. In the presence of morphogen, EC cells grow as flattened cells, becoming anchorage dependent and forming
aggregates. In addition, growth rate is reduced approximately 10 fold. Ultimately, the cells are induced to differentiate. In the example, varying concentrations of OP-3 (e.g., 0-300 ng/ml) are added daily to cultured cells (e.g., 40-50,000 cells in
2.5 ml chemically defined medium), and the effects of treatment determined by visual examination. OP-3 is anticipated to stimulate redifferentiation of these cells to a morphology characteristic of untransformed embryo cells. Example 5. Maintenance of Phenotype
Morphogenically active fragments of OP-3 also may be used to maintain a cell's differentiated phenotype. This application is particularly useful for inducing the continued expression of phenotype in senescent or quiescent cells.
5.1 In Vitro Model for Phenotypic Maintenance The phenotypic maintenance capability of morphogens is determined readily. A number of differentiated cells become senescent or quiescent after multiple passages in vitro under standard tissue culture
conditions well described in the art (e.g., Culture of Animal Cells: A Manual of Basic Techniques, C.R. Freshney, ed., Wiley, 1987). However, if these cells are cultivated in vitro in association with a morphogen such as OP-3, cells are stimulated to maintain
expression of their phenotype through multiple
passages. For example, the alkaline phosphatase activity of cultured osteoblasts, such as. cultured osteosarcoma cells and calvaria cells, is significantly reduced after multiple passages in vitro. However, if the cells are cultivated in the presence of OP-3, alkaline phosphatase activity should be maintained over extended periods of time. Similarly, phenotypic expression of myocytes also is maintained in the presence of a morphogen. In the experiment,
osteoblasts are cultured as described in Example 2. The cells are divided into groups, incubated with varying concentrations of OP-3 (e.g., 0-300 ng/ml) and passaged multiple times (e.g., 3-5 times) using
standard methodology. Passaged cells then are tested for alkaline phosphatase activity, as described in Example 3 as an indication of differentiated cell metabolic function. Osteoblasts cultured in the absence of OP-3 should have reduced alkaline
phosphatase activity, as compared to OP-3-treated cells.
5.2 In Vivo Model for Phenotypic Maintenance
Phenotypic maintenance capability also may be demonstrated in vivo, using a rat model for
osteoporosis, as disclosed in international application PCT/US92/07432 (WO93/05751). As described therein. Long Evans female rats (Charles River Laboratories, Wilmington, MA) are Sham-operated (control animals) or ovariectomized using standard surgical techniques, to produce an osteoporotic condition resulting from decreased estrogen production. Shortly following surgery, e.g., 200 days after ovariectomy, rats are systemically provided with phosphate buffered saline (PBS) or morphogen, (e.g., OP-3, 1-100 μg) for 21 days (e.g., by daily tail vein injection.) The rats then are sacrificed and serum alkaline phosphatase levels, serum calcium levels, and serum osteocalcin levels are determined, using standard methodologies as described therein and above. Elevated levels of osteocalcin and alkaline phosphatase should be observed in the rats treated with an effective amount of OP-3. Moreover, histomorphometric analysis on the tibial diasypheal bone is anticipated to show improved bone mass in
OP-3-treated animals as compared with untreated, ovariectomized rats. In fact, the bone mass of OP-3-animals is anticipated to be comparable to (e.g., approaches) that of the sham-operated (e.g.,
nonovarectomized) rats.
Example 6. Proliferation of Progenitor Cell Populations
Progenitor cells may be stimulated to proliferate in vivo or ex vivo. The cells may be stimulated in vivo by injecting or otherwise providing a sterile preparation containing the morphogenically active fragment of OP-3 into the individual. For example, the hemopoietic pluripotential stem cell population of an individual may be stimulated to proliferate by
injecting or otherwise providing an appropriate
concentration of OP-3 to the individual's bone marrow.
Progenitor cells may be stimulated ex vivo by contacting progenitor cells of the population to be enhanced with a morphogenically active fragment of OP-3 under sterile conditions at a concentration and for a time sufficient to stimulate proliferation of the cells. Suitable concentrations and stimulation times may be determined empirically, essentially following the procedure described in Example 2, above. A
morphogen concentration of between about 0.1-100 ng/ml and a stimulation period of from about 10 minutes to about 72 hours, or, more generally, about 24 hours, typically should be sufficient to stimulate a cell population of about 104 to 106 cells. The stimulated cells then are provided to the individual as, for example, by injecting the cells to an appropriate in vivo locus. Suitable biocompatible progenitor cells may be obtained by any of the methods known in the art or described hereinabove. Example 7. Regeneration of Damaged or Diseased Tissue
OP-3 may be used to repair diseased or damaged mammalian tissue. The tissue to be repaired preferably is assessed first, and excess necrotic or interfering scar tissue removed as needed, e.g., by ablation or by surgical, chemical, or other methods known in the medical arts. OP-3 then may be provided directly to the tissue locus as part of a sterile, biocompatible composition, either by surgical implantation or injection. The morphogen also may be provided systemically, as by oral or parenteral administration. Alternatively, a
sterile, biocompatible composition containing
progenitor cells stimulated by a morphogenically active fragment of OP-3 may be provided to the tissue locus. The existing tissue at the locus, whether diseased or damaged, provides the appropriate matrix to allow the proliferation and tissue-specific differentiation of progenitor cells. In addition, a damaged or diseased tissue locus, particularly one that has been further assaulted by surgical means, provides a morphogenically permissive environment. Systemic provision of OP-3 should be sufficient for certain applications (e.g., in the treatment of osteoporosis and other disorders of the bone remodeling cycle, as an example). In some circumstances, particularly where tissue damage is extensive, the tissue may not be capable of providing a sufficient matrix for cell influx and proliferation. In these instances, it may be necessary to provide OP-3 or progenitor cells stimulated by OP-3 to the tissue locus in association with a suitable, biocompatible, formulated matrix, prepared by any of the means described below. The matrix preferably is in vivo biodegradable. The matrix also may be
tissue-specific and/or may comprise porous particles having dimensions within the range of 70-850μm, most preferably 150-420μm. OP-3 also may be used to prevent or substantially inhibit immune/inflammatory response-mediated tissue damage and scar tissue formation following an injury. OP-3 is provided to a newly injured tissue locus, to induce tissue morphogenesis at the locus, preventing the aggregation of migrating fibroblasts into nondifferentiated connective tissue. OP-3 preferably is provided as a sterile pharmaceutical preparation injected into the tissue locus within five hours of the injury. Where an immune/inflammatory response is unavoidably or deliberately induced, as part of, for example, a surgical or other aggressive clinical therapy, OP-3 preferably is provided prophylactically to the patient, prior to, or concomitant with, the therapy.
Below are several examples, describing protocols for demonstrating OP-3-induced tissue morphogenesis in bone, liver, nerve, dentin, cementum and periodontal tissue.
7.1 OP-3-Induced Bone Morphogenesis
A particularly useful mammalian tissue model system for demonstrating and evaluating the morphogenic activity of a protein is the endochondral bone tissue morphogenesis model known in the art and described, for example, in U.S. Pat. No. 4,968,590. The ability to induce endochondral bone formation includes the ability to induce the proliferation of progenitor cells into chondroblasts and osteoblasts, the ability to induce cartilage matrix formation, cartilage calcification, and bone remodeling, and the ability to induce
formation of an appropriate vascular supply and
hematopoeitic bone marrow differentiation.
The local environment in which the morphogenic material is placed is important for tissue
morphogenesis. As used herein, "local environment" is understood to include the tissue structural matrix and the environment surrounding the tissue. For example, in addition to needing an appropriate anchoring
substratum for their proliferation, the cells
stimulated by morphogens need signals to direct the tissue-specificity of their differentiation. These signals vary for the different tissues and may include cell surface markers. In addition, vascularization of new tissue requires a local environment which supports vascularization.
The following sets forth various procedures for evaluating the in vivo morphogenic utility of OP-3 and OP-3-containing compositions. The compositions may be injected or surgically implanted in a mammal, following any of a number of procedures well known in the art. For example, surgical implant bioassays may be
performed essentially following the procedure of
Sampath et al. (1983) PNAS 80: 6591-6595 and U.S. Pat No. 4,968,590. Histological sectioning and staining is preferred to determine the extent of morphogenesis in vivo, particularly in tissue repair procedures. Excised implants are fixed in Bouins Solution, embedded in paraffin, and cut into 6-8 μm sections. Staining with toluidine blue or hemotoxylin/eosin demonstrates clearly the ultimate development of the new tissue. Twelve day implants are usually sufficient to determine whether the implants contain newly induced tissue.
Successful implants exhibit a controlled
progression through the stages of induced tissue development allowing one to identify and follow the tissue-specific events that occur. For example, in endochondral bone formation the stages include:
(1) leukocytes on day one; (2) mesenchymal cell
migration and proliferation on days two and three;
(3) chondrocyte appearance on days five and six;
(4) cartilage matrix formation on day seven;
(5) cartilage calcification on day eight; (6) vascular invasion, appearance of osteoblasts, and formation of new bone on days nine and ten; (7) appearance of osteoclastic cells, and the commencement of bone remodeling and dissolution of the implanted matrix on days twelve to eighteen; and (8) hematopoietic bone marrow differentiation in the resulting ossicles on day twenty-one.
In addition to histological evaluation, biological markers may be used as markers for tissue
morphogenesis. Useful markers include tissue-specific enzymes whose activities may be assayed (e.g.,
spectrophotometrically) after homogenization of the implant. These assays may be useful for quantitation and for rapidly obtaining an estimate of tissue formation after the implants are removed from the animal. For example, alkaline phosphatase activity may be used as a marker for osteogenesis.
Incorporation of systemically provided OP-3 may be followed using tagged fragments (e.g., radioactively labelled) and determining their localization in the new tissue, and/or by monitoring their disappearance from the circulatory system using a standard labeling protocol and pulse-chase procedure. OP-3 also may be provided with a tissue-specific molecular tag, whose uptake may be monitored and correlated with the
concentration of OP-3 provided. As an example, ovary removal in female rats results in reduced bone alkaline phosphatase activity, and renders the rats predisposed to osteoporosis (as described in Example 5). If the female rats now are provided with OP-3, a reduction in the systemic concentration of calcium should be seen, which correlates with the presence of the provided OP-3 and which is anticipated to correspond with increased alkaline phosphatase activity.
7.2 Morphogen-Induced Liver Regeneration
As another example, a method for inducing
morphogenesis of substantially injured liver tissue following a partial hepatectomy utilizing OP-3 is presented. Variations on this general protocol may be used to test morphogen activity of OP-3 in other different tissues. The general method involves
excising an essentially nonregenerating portion of a tissue and providing OP-3, preferably as a soluble pharmaceutical preparation to the excised tissue locus, closing the wound, and examining the site at a future date. Like bone, liver has a potential to regenerate upon injury during post-fetal life.
OP-3, e.g., 1 mg/ml, in a biocompatible solution, for example, (e.g., a purified recombinant mature form of OP-3, is solubilized in 50% ethanol, or compatible solvent, containing 0.1% trifluoroacetic acid, or compatible acid. Alternatively, the mature protein may be solubilized by association with a pro domain. The injectable OP-3 solution is prepared, e.g., by diluting one volume of OP-3 solvent-acid stock solution with 9 volumes of 0.2% rat serum albumin in sterile PBS (phosphate-buffered saline).
In the experiment, growing rats or aged rats (e.g., Long Evans, Charles River Laboratories, Wilmington) are anesthetized by using ketamine. Two of the liver lobes (left and right) are cut out (approximately 1/3 of the lobe) and the OP-3 is injected locally at multiple sites along the cut ends. The amount of OP-3 injected may be, e.g., 100 μg in 1000 μl of PBS/RSA (phosphate buffered saline/rat serum albumin) injection buffer. Placebo samples are injection buffer only. In
experimental essays, five rats in each group preferably are used. The wound is closed and the rats are allowed to eat normal food and drink tap water. After 12 days, the rats are sacrificed and liver regeneration is observed visually, to evaluate the effects of the OP-3 on liver regeneration most
effectively. The OP-3 fragment-injected group is anticipated to show, e.g., complete liver tissue regeneration with no sign remaining of any cut in the liver. By contrast, the control group into which only PBS is injected, show only minimal regeneration with the incision remaining in the sample. Previous experiments with other morphogens (e.g., OP-1) show these morphogens alone induce liver tissue
regeneration.
7.3 Morphogen-Induced Dentin, Cementum and
Periodontal Ligament Regeneration
As still another example, the ability of OP-3 to induce dentinogenesis also may be demonstrated. To date, the unpredictable response of dental pulp tissue to injury is a basic clinical problem in dentistry. Cynomolgus monkeys are chosen as primate models as monkeys are presumed to be more indicative of human dental biology than models based on lower non-primate mammals.
Using standard dental surgical procedures, small areas (e.g., 2mm) of dental pulps are surgically exposed by removing the enamel and dentin immediately above the pulp (by drilling) of sample teeth,
performing a partial amputation of the coronal pulp tissue, inducing hemostasis, application of the pulp treatment, and sealing and filling the cavity by standard procedures. Pulp treatments used may include: a
morphogenically active fragment of OP-3 dispersed in a carrier matrix; carrier matrix alone, and no treatment. Twelve teeth per animal (four for each treatment) are prepared, and two animals are used. At four weeks, teeth are extracted and processed histologically for analysis of dentin formation, and/or ground to analyze dentin mineralization. The effect of OP-3 on
osteodentin reparation may be observed visually by comparing control samples treatment (PBS) with OP-3. OP-3 plus a carrier matrix induces formation of
reparative or osteodentin bridges on surgically exposed healthy dental pulps. By contrast, pulps treated with carrier matrix alone, do not form reparative dentin.
Similarly, implanting demineralized teeth and OP-3 into surgically prepared canine tooth sockets is anticipated to stimulate new periodontal tissue
formation, including new cementum and periodontal ligament, as well as new alveolar bone and dentin tissue, as described for OP-1 in international
application PCT/US92/08742, filed 9/15/93. By
contrast, untreated teeth or teeth treated with carrier vehicle alone do not induce periodontal tissue growth.
7.4 Morphogen-Induced Nerve Tissue Repair
As yet another example, the induction of
regenerative effects on central nervous system (CNS) repair, by a morphogenically active fragment of OP-3, may be demonstrated using a rat brain stab model. In the experiment, male Long Evans rats are anesthetized and the head area prepared for surgery. The calvariae is exposed using standard surgical procedures and a hole drilled toward the center of each lobe using a 0.035K wire, just piercing the calvariae. 25μl
solutions containing either morphogen (e.g., OP-3, 25μg) or PBS then is provided to each of the holes by Hamilton syringe. Solutions are delivered to a depth approximately 3 mm below the surface, into the
underlying cortex, corpus callosum and hippocampus. The skin then is sutured and the animal allowed to recover.
Three days post surgery, rats are sacrificed by decapitation and their brains processed for sectioning. Scar tissue formation is evaluated by immunofluoresence staining for glial fibrillary acidic protein, a marker protein for glial scarring, to qualitatively determine the degree of scar formation. Sections also are probed with OP-3-specific antibody to determine the presence of the protein. Reduced levels of glial fibrillary acidic protein are anticipated to be observed in the tissue sections of animals treated with OP-3,
evidencing the ability of the morphogen to inhibit glial scar formation, thereby stimulating nerve
regeneration.
The ability of OP-3 to stimulate peripheral nervous system axonal growth over extended distances may be demonstrated using the following model. Neurons of the peripheral nervous system can sprout new processes on their own following injury, but without guidance these sproutings typically fail to connect appropriately and die. Where the break is extensive, e.g., greater than 5 or 10 mm, regeneration is poor or nonexistent.
Previous experiments with other morphogens, e.g., OP-1, show that morphogens stimulate peripheral nervous system axonal growth over extended distances, allowing repair and regeneration of damaged peripheral neural pathways.
In this example OP-3 stimulation of nerve
regeneration is demonstrated using the rat sciatic nerve model. The rat sciatic nerve can regenerate spontaneously across a 5 mm gap, and occasionally across a 10 mm gap, provided that the severed ends are inserted in a saline-filled nerve guidance channel. In this experiment, nerve regeneration across at least a 12mm gap is tested.
Adult female Sprague-Dawley rats (Charles River Laboratories, Inc.) weighing 230-250 g are anesthetized with intraperitoneal injections of sodium pentobarbital (35 mg/kg body weight). A skin incision is made parallel and just posterior to the femur. The
avascular intermuscular plane between vastus lateralis and hamstring muscles are entered and followed to the loose fibroareolar tissue surrounding the sciatic nerve. The loose tissue is divided longitudinally thereby freeing the sciatic nerve over its full extent without devascularizing any portion. Under a surgical microscope the sciatic nerves are transected with microscissors at mid-thigh and grafted with a OP-3 gel graft that separates the nerve stumps by 12 mm. The graft region is encased in a silicone tube 20 mm in length with a 1.5 mm inner diameter, the interior of which is filled with the morphogen solution.
Specifically, the central 12 mm of the tube consists of an OP-3 gel prepared by mixing 1 to 5 μg of
substantially pure recombinantly produced OP-3 protein with approximately 100 μl of MATRIGEL™ (from
Collaborative Research, Inc., Bedford, MA), an
extracellular matrix extract derived from mouse sarcoma tissue, and containing solubilized tissue basement membrane, including laminin, type IV collagen, heparin sulfate, proteoglycan and entactin, in phosphatebuffered saline. The morphogen-filled tube then is implanted directly into the defect site, allowing 4 mm on each end to insert the nerve stumps . Each stump is abutted against the morphogen gel and is secured in the silicone tube by three stitches of commercially
available surgical 10-0 nylon through the epineurium, the fascicle protective sheath.
In addition to OP-3 gel grafts, control grafts of empty silicone tubes, silicone tubes filled with gel only and "reverse" autografts, wherein 12 mm transected segments of the animal's sciatic nerve are rotated 180° prior to suturing, preferably also are grafted. All experiments preferably are performed in quadruplicate. All wounds preferably are closed by wound clips that are removed after 10 days. Rats can be grafted on both legs. At 3 weeks the animals are sacrificed, and the grafted segments removed and frozen on dry ice
immediately. Frozen sections then are cut throughout the graft site, and examined for axonal regeneration by immunofluorescent staining using anti-neurofilament antibodies labeled with flurocein (obtained, for example, from Sigma Chemical Co., St. Louis).
Regeneration of the sciatic nerve is anticipated to occur across the entire 12 mm distance in all graft sites wherein the gap is filled with the OP-3 gel. By contrast, empty silicone tubes, gel alone and reverse autografts do not show nerve regeneration. Example 8. Identification of Morphogen-Expressing
Tissue
Determining the tissue distribution of morphogens may be used to identify different morphogens expressed in a given tissue, as well as to identify new, related morphogens. Tissue distribution also may be used to identify useful morphogen-producing tissue for use in screening and identifying candidate morphogen-stimulating agents. The morphogens (or their mRNA transcripts) readily are identified in different tissues using standard methodologies and minor
modifications thereof in tissues where expression may be low. For example, protein distribution may be determined using standard Western blot analysis or immunofluorescent techniques, and antibodies specific to the morphogen or morphogens of interest. Similarly, the distribution of morphogen transcripts may be determined using standard Northern hybridization protocols and transcript-specific probes.
Any probe capable of hybridizing specifically to a transcript, and distinguishing the transcript of interest from other, related transcripts may be used. Because the morphogens described herein share such high sequence homology in their active, C-terminal domains, the tissue distribution of a specific morphogen
transcript may best be determined using a probe
specific for the pro region of the immature protein and/or the N-terminal region of the mature protein.
Another useful sequence is the 3' non-coding region flanking and immediately following the stop codon.
These portions of the sequence vary substantially among the morphogens of this invention, and accordingly, are specific for each protein. For example, a particularly useful OP-3-specific probe sequence is one derived from a portion of the 3' untranslated sequence, e.g., nucleotides 1310-1674 of Seq. ID No. 1, which shares little or no homology with other morphogen sequences, including OP-2. The chosen fragment then is labelled using standard means well known and described in the art.
Using these morphogen-specific probes, which may be synthetically engineered or obtained from cloned sequences, morphogen transcripts can be identified in mammalian tissue, using standard methodologies well known to those having ordinary skill in the art. A detailed description of a suitable hybridization protocol is described in Ozkaynak, et al., (1991)
Biochem. Biophys. Res. Commn. 179: 116-123, and
Ozkaynak, et al. (1992) J. Biol. Chemistry
267:25220-25227. Briefly, total RNA is prepared from various tissues (e.g., murine embryo and developing and adult liver, kidney, testis, heart, brain, thymus, stomach) by a standard methodology such as by the method of Chomczyaski et al. ((1987) Anal. Biochem
162:156-159) and described below. Poly (A)+ RNA is prepared by using oligo (dT)-cellulose chromatography (e.g.. Type 7, from Pharmacia LKB Biotechnology, Inc.). Poly (A)+ RNA (generally 15 μg) from each tissue is fractionated on a 1% agarose/formaldehyde gel and transferred onto a Nytran membrane (Schleicher &
Schuell). Following the transfer, the membrane is baked at 80°C and the RNA is cross-linked under UV light (generally 30 seconds at 1 mW/cm2 ) . Prior to hybridization, the appropriate probe is denatured by heating. The hybridization is carried out in a lucite cylinder rotating in a roller bottle apparatus at approximately 1 rev/min for approximately 15 hours at 37°C using a hybridization mix of 40% formamide,
5 × Denhardts, 5 × SSPE, and 0.1% SDS. Following hybridization, the non-specific counts are washed off the filters in 0.1 × SSPE, 0.1% SDS at 50°C
An OP-3-specific 0.5 kb probe was made from a StuI-BglII fragment of OP-3 cDNA. The fragment contains the 3' untranslated sequence from nucleotides 1310-1674, plus an additional 140 bases. The fragment was labelled using standard techniques and the
hybridization performed as described. To date, OP-3, like OP-2, appears to be expressed primarily in early embryonic tissue. Specifically, Northern blots of murine embryos show abundant OP-3 expression in 8-day embryos, demonstrated by a strong band at 2.9 kb and a weaker band at 2.3 kb.
Example 9. Screening Assay for Candidate Compounds which Alter Endogenous Morphogen Levels
Candidate compound(s) which may be administered to affect the level of endogenous OP-3 morphogen may be found using the following screening assay, in which the level of OP-3 production by a cell type which produces measurable levels of the morphogen is determined with and without incubating the cell in culture with the compound, in order to assess the effects of the
compound on the cell. This can be accomplished by detection of the morphogen either at the protein or RNA level. A detailed description also may be found in international application PCT/US92/07359, (WO93/05172).
9.1 Growth of Cells in Culture
Cell cultures of kidney, adrenals, urinary bladder, brain, or other organs, may be prepared as described widely in the literature. For example, kidneys may be explanted from neonatal or new born or young or adult rodents (mouse or rat) and used in organ culture as whole or sliced (1-4 mm) tissues. Primary tissue cultures and established cell lines, also derived from kidney, adrenals, urinary, bladder, brain, mammary, or other tissues may be established in multiwell plates (6 well or 24 well) according to conventional cell culture techniques, and are cultured in the absence or presence of serum for a period of time (1-7 days). Cells may be cultured, for example, in Dulbecco's Modified Eagle medium (Gibco, Long Island, NY) containing serum (e.g., fetal calf serum at 1%-10%, Gibco) or in serum-deprived medium, as desired, or in defined medium (e.g.,
containing insulin, transferrin, glucose, albumin, or other growth factors).
Samples for testing the level of morphogen
production includes culture supernatants or cell lysates, collected periodically and evaluated for morphogen production by immunoblot analysis (Sambrook et al., eds., 1989, Molecular Cloning, Cold Spring Harbor Press, Cold Spring Harbor, NY), or a portion of the cell culture itself, collected periodically and used to prepare polyA+ RNA for RNA analysis. To monitor de novo morphogen synthesis, some cultures are labeled according to conventional procedures with an
S-methionine/ S-cysteine mixture for 6-24 hours and then evaluated for morphogenic protein synthesis by conventional immunoprecipitation methods.
9.2 Determination of Level of Morphogenic Protein In order to quantitate the production of a
morphogenic protein, e.g., OP-3, by a cell type, an immunoassay may be performed to detect the morphogen using a polyclonal or monoclonal antibody specific for that protein. For example, OP-3 may be detected using a polyclonal antibody specific for OP-3 in an ELISA, as follows.
1 μg/100 μl of affinity-purified polyclonal rabbit IgG specific for OP-3 is added to each well of a
96-well plate and incubated at 37 °C for an hour. The wells are washed four times with 0.167M sodium borate buffer with 0.15 M NaCl (BSB), pH 8.2, containing 0.1% Tween 20. To minimize non-specific binding, the wells are blocked by filling completely with 1% bovine serum albumin (BSA) in BSB and incubating for 1 hour at 37°C The wells are then washed four times with BSB
containing 0.1% Tween 20. A 100 μl aliquot of an appropriate dilution of each of the test samples of cell culture supernatant is added to each well in triplicate and incubated at 37°C for 30 min. After incubation, 100 μl biotinylated rabbit anti-OP-3 serum (stock solution is about 1 mg/ml and diluted 1:400 in BSB containing 1% BSA before use) is added to each well and incubated at 37°C for 30 min. The wells are then washed four times with BSB containing 0.1% Tween 20. 100 μl strepavidin-alkaline (Southern Biotechnology Associates, Inc. Birmingham, Alabama, diluted 1:2000 in BSB containing 0.1% Tween 20 before use) is added to each well and incubated at 37°C for 30 min. The plates are washed four times with 0.5M Tris buffered Saline (TBS), pH 7.2. 50μl substrate (ELISA Amplification System Kit, Life Technologies, Inc., Bethesda, MD) is added to each well incubated at room temperature for 15 min. Then, 50 μl amplifier (from the same
amplification system kit) is added and incubated for another 15 min at room temperature. The reaction is stopped by the addition of 50 μl 0.3 M sulphuric acid. The OD at 490 nm of the solution in each well is recorded. To quantitate OP-3 in culture media, an OP-3 standard curve is performed in parallel with the test samples. Polyclonal antibody may be prepared as follows. Each rabbit is given a primary immunization of 100 ug/500 μl recombinantly-produced OP-3 protein or protein fragment in 0.1% SDS mixed with 500 μl
Complete Freund's Adjuvant. The antigen is injected subcutaneously at multiple sites on the back and flanks of the animal. The rabbit is boosted after a month in the same manner using incomplete Freund's Adjuvant. Test bleeds are taken from the ear vein seven days later. Two additional boosts and test bleeds are performed at monthly intervals until antibody against OP-3 is detected in the serum using an ELISA assay. Then, the rabbit is boosted monthly with 100 μg of antigen and bled (15 ml per bleed) at days seven and ten after boosting.
Monoclonal antibody specific for a given morphogen may be prepared as follows. A mouse is given two injections of OP-3 protein or a protein fragment specific for OP-3. The protein preferably is
recombinantly produced. The first injection contains lOOμg of OP-3 in complete Freund's adjuvant and is given subcutaneously. The second injection contains 50 μg of OP-3 in incomplete adjuvant and is given
intraperitoneally. The mouse then receives a total of 230 μg of OP-3 in four intraperitoneal injections at various times over an eight month period. One week prior to fusion, the mouse is boosted intraperitoneally with OP-3 (e.g., 100 μg) and may be additionally boosted with an OP-3-specific peptide (e.g.,
corresponding to the N-terminus of the mature protein) conjugated to bovine serum albumin with a suitable crosslinking agent. This boost can be repeated five days (IP), four days (IP), three days (IP) and one day (IV) prior to fusion. The mouse spleen cells then are fused to commercially available myeloma cells at a ratio of 1:1 using PEG 1500 (Boeringer Mannheim,
Germany), and the fused cells plated and screened for OP-3-specific antibodies using OP-3 as antigen. The cell fusion and monoclonal screening steps readily are performed according to standard procedures well
described in standard texts widely available in the art. Other Embodiments
The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The present embodiments are therefore to be considered in all respects as
illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein. SEQUENCE LISTING (1 ) ) GENERAL INFORMATION:
(i) APPLICANT:
(A) NAME: CREATIVE BIOMOLECULES, INC.
(B) STREET: 45 SOUTH STREET
(C) CITY: HOPKINTON
(D) STATE: MA
(E) COUNTRY: USA
(F) POSTAL CODE (ZIP): 01748
(G) TELEPHONE: 1-508-435-9001
(H) TELEFAX: 1-508-435-0454
(I) TELEX:
(ii) TITLE OF INVENTION: OP3-INDUCED MORPHOGENESIS
(iii) NUMBER OF SEQUENCES: 13
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: CREATIVE BIOMOLECULES, INC.
(B) STREET: 45 SOUTH STREET
(C) CITY: HOPKINTON
(D) STATE: MA
(E) COUNTRY: USA
(F) ZIP: 01748
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.25 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/667,274
(B) FILING DATE: ll-MAR-1991
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/752,764
(B) FILING DATE: 30-AUG-1991
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/753,059
(B) FILING DATE: 30-AUG-1991 (vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/752,857
(B) FILING DATE: 30-AUG-1991
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/923,780
(B) FILING DATE: 31-JUL-1992
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 07/922,813
(B) FILING DATE: 31-JUL-1992
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: PITCHER ESQ, EDMUND R
(B) REGISTRATION NUMBER: 27,829
(C) REFERENCE/DOCKET NUMBER: CRP-076PC
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (508)435-9001
(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1674 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 69..1268
(D) OTHER INFORMATION: /note= "mOP3-PP"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
GGATCCGCGG CGCTGTCCCA TCCTTGTCGT CGAGGCGTCG CTGGATGCGA GTCCGCTAAA 60
CGTCCGAG ATG GCT GCG CGT CCG GGA CTC CTA TGG CTA CTG GGC CTG GCT 110
Met Ala Ala Arg Pro Gly Leu Leu Trp Leu Leu Gly Leu Ala 1 5 10
CTG TGC GTG TTG GGC GGC GGT CAC CTC TCG CAT CCC CCG CAC GTC TTT 158 Leu Cys Val Leu Gly Gly Gly His Leu Ser His Pro Pro His Val Phe
15 20 25 30 CCC CAG CGT CGA CTA GGA GTA CGC GAG CCC CGC GAC ATG CAG CGC GAG 206 Pro Gln Arg Arg Leu Gly Val Arg Glu Pro Arg Asp Met Gln Arg Glu
35 40 45
ATT CGG GAG GTG CTG GGG CTA GCC GGG CGG CCC CGA TCC CGA GCA CCG 254 Ile Arg Glu Val Leu Gly Leu Ala Gly Arg Pro Arg Ser Arg Ala Pro
50 55 60
GTC GGG GCT GCC CAG CAG CCA GCG TCT GCG CCC CTC TTT ATG TTG GAC 302 Val Gly Ala Ala Gln Gln Pro Ala Ser Ala Pro Leu Phe Met Leu Asp
65 70 75
CTG TAC CGT GCC ATG ACG GAT GAC AGT GGC GGT GGG ACC CCG CAG CCT 350 Leu Tyr Arg Ala Met Thr Asp Asp Ser Gly Gly Gly Thr Pro Gln Pro
80 85 90
CAC TTG GAC CGT GCT GAC CTG ATT ATG AGC TTT GTC AAC ATA GTG GAA 398
His Leu Asp Arg Ala Asp Leu Ile Met Ser Phe Val Asn Ile Val Glu
95 100 105 110
CGC GAC CGT ACC CTG GGC TAC CAG GAG CCA CAC TGG AAG GAA TTC CAC 446 Arg Asp Arg Thr Leu Gly Tyr Gln Glu Pro His Trp Lys Glu Phe His
115 120 125
TTT GAC CTA ACC CAG ATC CCT GCT GGG GAG GCT GTC ACA GCT GCT GAG 494 Phe Asp Leu Thr Gln Ile Pro Ala Gly Glu Ala Val Thr Ala Ala Glu
130 135 140
TTC CGG ATC TAC AAA GAA CCC AGT ACC CAC CCG CTC AAC ACA ACC CTC 542 Phe Arg Ile Tyr Lys Glu Pro Ser Thr His Pro Leu Asn Thr Thr Leu
145 150 155
CAC ATC AGC ATG TTC GAA GTG GTC CAA GAG CAC TCC AAC AGG GAG TCT 590 His Ile Ser Met Phe Glu Val Val Gln Glu His Ser Asn Arg Glu Ser
160 165 170
GAC TTG TTC TTT TTG GAT CTT CAG ACG CTC CGA TCT GGG GAC GAG GGC 638 Asp Leu Phe Phe Leu Asp Leu Gln Thr Leu Arg Ser Gly Asp Glu Gly
175 180 185 190
TGG CTG GTG CTG GAC ATC ACA GCA GCC AGT GAC CGA TGG CTG CTG AAC 686 Trp Leu Val Leu Asp Ile Thr Ala Ala Ser Asp Arg Trp Leu Leu Asn
195 200 205 CAT CAC AAG GAC CTA GGA CTC CGC CTC TAT GTG GAA ACC GAG GAT GGG 734 His His Lys Asp Leu Gly Leu Arg Leu Tyr Val Glu Thr Glu Asp Gly
210 215 220 CAC AGC ATA GAT CCT GGC CTA GCT GGT CTG CTT GGA CGA CAA GCA CCA 782 His Ser Ile Asp Pro Gly Leu Ala Gly Leu Leu Gly Arg Gln Ala Pro
225 230 235
CGC TCC AGA CAG CCT TTC ATG GTT GGT TTC TTC AGG GCC AAC CAG AGT 830 Arg Ser Arg Gln Pro Phe Met Val Gly Phe Phe Arg Ala Asn Gln Ser
240 245 250
CCT GTG CGG GCC CCT CGA ACA GCA AGA CCA CTG AAG AAG AAG CAG CTA 878 Pro Val Arg Ala Pro Arg Thr Ala Arg Pro Leu Lys Lys Lys Gln Leu
255 260 265 270
AAT CAA ATC AAC CAG CTG CCG CAC TCC AAC AAA CAC CTA GGA ATC CTT 926 Asn Gln Ile Asn Gln Leu Pro His Ser Asn Lys His Leu Gly Ile Leu
275 280 285
GAT GAT GGC CAC GGT TCT CAC GGC AGA GAA GTT TGC CGC AGG CAT GAG 974 Asp Asp Gly His Gly Ser His Gly Arg Glu Val Cys Arg Arg His Glu
290 295 300
CTC TAT GTC AGC TTC CGT GAC CTT GGC TGG CTG GAC TCT GTC ATT GCC 1022 Leu Tyr Val Ser Phe Arg Asp Leu Gly Trp Leu Asp Ser Val Ile Ala
305 310 315
CCC CAG GGC TAC TCC GCC TAT TAC TGT GCT GGG GAG TGC ATC TAC CCA 1070 Pro Gln Gly Tyr Ser Ala Tyr Tyr Cys Ala Gly Glu Cys Ile Tyr Pro
320 325 330
CTG AAC TCC TGT ATG AAC TCC ACC AAC CAC GCC ACT ATG CAG GCC CTG 1118 Leu Asn Ser Cys Met Asn Ser Thr Asn His Ala Thr Met Gln Ala Leu
335 340 345 350
GTA CAT CTG ATG AAG CCA GAT ATC ATC CCC AAG GTG TGC TGT GTG CCT 1166 Val His Leu Met Lys Pro Asp Ile Ile Pro Lys Val Cys Cys Val Pro
355 360 365
ACT GAG CTG AGT GCC ATT TCT CTG CTC TAC TAT GAT AGA AAC AAT AAT 1214
Thr Glu Leu Ser Ala Ile Ser Leu Leu Tyr Tyr Asp Arg Asn Asn Asn
370 375 380
GTC ATC CTG CGC AGG GAG CGC AAC ATG GTA GTC CAG GCC TGT GGC TGC 1262
Val Ile Leu Arg Arg Glu Arg Asn Met Val Val Gln Ala Cys Gly Cys
385 390 395
CAC TGAGTCCCTG CCCAACAGCC TGCTGCCATC CCATCTATCT AGTCAGGCCT 1315
His
400
CTCTTCCAAG GCAGGAAACC AACAAAGAGG GAAGGCAGTG CTTTCAACTC CATGTCCACA 1375 TTCACAGTCT TGGCCCTCTC TGTTCTTTTT GCCAAGGCTG AGAAGATGGT CCTAGTTATA 1435
ACCCTGGTGA CCTCAGTAGC CCGATCTCTC ATCTCCCCAA ACTCCCCAAT GCAGCCAGGG 1495 GCATCTATGT CCTTTGGGAT TGGGCACAGA AGTCCAATTT ACCAACTTAT TCATGAGTCA 1555
CTACTGGCCC AGCCTGGACT TGAACCTGGA ACACAGGGTA GAGCTCAGGC TCTTCAGTAT 1615
CCATCAGAAG ATTTAGGTGT GTGCAGACAT GACCACACTC CCCCTAGCAC TCCATAGCC 1674
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 399 amino acids
(B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:
Met Ala Ala Arg Pro Gly Leu Leu Trp Leu Leu Gly Leu Ala Leu Cys
1 5 10 15
Val Leu Gly Gly Gly His Leu Ser His Pro Pro His Val Phe Pro Gln
20 25 30
Arg Arg Leu Gly Val Arg Glu Pro Arg Asp Met Gln Arg Glu Ile Arg
35 40 45
Glu Val Leu Gly Leu Ala Gly Arg Pro Arg Ser Arg Ala Pro Val Gly
50 55 60
Ala Ala Gln Gln Pro Ala Ser Ala Pro Leu Phe Met Leu Asp Leu Tyr
65 70 75 80
Arg Ala Met Thr Asp Asp Ser Gly Gly Gly Thr Pro Gln Pro His Leu
85 90 95
Asp Arg Ala Asp Leu Ile Met Ser Phe Val Asn Ile Val Glu Arg Asp
100 105 110
Arg Thr Leu Gly Tyr Gln Glu Pro His Trp Lys Glu Phe His Phe Asp
115 120 125
Leu Thr Gln Ile Pro Ala Gly Glu Ala Val Thr Ala Ala Glu Phe Arg
130 135 140 Ile Tyr Lys Glu Pro Ser Thr His Pro Leu Asn Thr Thr Leu His Ile 145 150 155 160
Ser Met Phe Glu Val Val Gln Glu His Ser Asn Arg Glu Ser Asp Leu
165 170 175
Phe Phe Leu Asp Leu Gln Thr Leu Arg Ser Gly Asp Glu Gly Trp Leu
180 185 190
Val Leu Asp Ile Thr Ala Ala Ser Asp Arg Trp Leu Leu Asn His His
195 200 205
Lys Asp Leu Gly Leu Arg Leu Tyr Val Glu Thr Glu Asp Gly His Ser 210 215 220
Ile Asp Pro Gly Leu Ala Gly Leu Leu Gly Arg Gln Ala Pro Arg Ser 225 230 235 240
Arg Gln Pro Phe Met Val Gly Phe Phe Arg Ala Asn Gln Ser Pro Val
245 250 255
Arg Ala Pro Arg Thr Ala Arg Pro Leu Lys Lys Lys Gln Leu Asn Gln
260 265 270
Ile Asn Gln Leu Pro His Ser Asn Lys His Leu Gly Ile Leu Asp Asp
275 280 285
Gly His Gly Ser His Gly Arg Glu Val Cys Arg Arg His Glu Leu Tyr 290 295 300
Val Ser Phe Arg Asp Leu Gly Trp Leu Asp Ser Val Ile Ala Pro Gln
305 310 315 320
Gly Tyr Ser Ala Tyr Tyr Cys Ala Gly Glu Cys Ile Tyr Pro Leu Asn
325 330 335
Ser Cys Met Asn Ser Thr Asn His Ala Thr Met Gln Ala Leu Val His
340 345 350
Leu Met Lys Pro Asp Ile Ile Pro Lys Val Cys Cys Val Pro Thr Glu
355 360 365
Leu Ser Ala Ile Ser Leu Leu Tyr Tyr Asp Arg Asn Asn Asn Val Ile 370 375 380
Leu Arg Arg Glu Arg Asn Met Val Val Gln Ala Cys Gly Cys His 385 390 395 (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1822 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(F) TISSUE TYPE: HIPPOCAMPUS
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 49..1341
(C) IDENTIFICATION METHOD: experimental
(D) OTHER INFORMATION: /function= "OSTEOGENIC PROTEIN"
/product= "hOP1-PP"
/note= "hOP1 cDNA"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GGTGCGGGCC CGGAGCCCGG AGCCCGGGTA GCGCGTAGAG CCGGCGCG ATG CAC GTG 57
Met His Val
1
CGC TCA CTG CGA GCT GCG GCG CCG CAC AGC TTC GTG GCG CTC TGG GCA 105 Arg Ser Leu Arg Ala Ala Ala Pro His Ser Phe Val Ala Leu Trp Ala
5 10 15
CCC CTG TTC CTG CTG CGC TCC GCC CTG GCC GAC TTC AGC CTG GAC AAC 153 Pro Leu Phe Leu Leu Arg Ser Ala Leu Ala Asp Phe Ser Leu Asp Asn
20 25 30 35
GAG GTG CAC TCG AGC TTC ATC CAC CGG CGC CTC CGC AGC CAG GAG CGG 201 Glu Val His Ser Ser Phe Ile His Arg Arg Leu Arg Ser Gln Glu Arg
40 45 50
CGG GAG ATG CAG CGC GAG ATC CTC TCC ATT TTG GGC TTG CCC CAC CGC 249 Arg Glu Met Gln Arg Glu Ile Leu Ser Ile Leu Gly Leu Pro His Arg
55 60 65 CCG CGC CCG CAC CTC CAG GGC AAG CAC AAC TCG GCA CCC ATG TTC ATG 297 Pro Arg Pro His Leu Gln Gly Lys His Asn Ser Ala Pro Met Phe Met
70 75 80
CTG GAC CTG TAC AAC GCC ATG GCG GTG GAG GAG GGC GGC GGG CCC GGC 345 Leu Asp Leu Tyr Asn Ala Met Ala Val Glu Glu Gly Gly Gly Pro Gly
85 90 95
GGC CAG GGC TTC TCC TAC CCC TAC AAG GCC GTC TTC AGT ACC CAG GGC 393 Gly Gln Gly Phe Ser Tyr Pro Tyr Lys Ala Val Phe Ser Thr Gln Gly
100 105 110 115
CCC CCT CTG GCC AGC CTG CAA GAT AGC CAT TTC CTC ACC GAC GCC GAC 441 Pro Pro Leu Ala Ser Leu Gln Asp Ser His Phe Leu Thr Asp Ala Asp
120 125 130
ATG GTC ATG AGC TTC GTC AAC CTC GTG GAA CAT GAC AAG GAA TTC TTC 489 Met Val Met Ser Phe Val Asn Leu Val Glu His Asp Lys Glu Phe Phe
135 140 145
CAC CCA CGC TAC CAC CAT CGA GAG TTC CGG TTT GAT CTT TCC AAG ATC 537 His Pro Arg Tyr His His Arg Glu Phe Arg Phe Asp Leu Ser Lys Ile
150 155 160
CCA GAA GGG GAA GCT GTC ACG GCA GCC GAA TTC CGG ATC TAC AAG GAC 585 Pro Glu Gly Glu Ala Val Thr Ala Ala Glu Phe Arg Ile Tyr Lys Asp
165 170 175
TAC ATC CGG GAA CGC TTC GAC AAT GAG ACG TTC CGG ATC AGC GTT TAT 633 Tyr Ile Arg Glu Arg Phe Asp Asn Glu Thr Phe Arg Ile Ser Val Tyr
180 185 190 195
CAG GTG CTC CAG GAG CAC TTG GGC AGG GAA TCG GAT CTC TTC CTG CTC 681 Gln Val Leu Gln Glu His Leu Gly Arg Glu Ser Asp Leu Phe Leu Leu
200 205 210
GAC AGC CGT ACC CTC TGG GCC TCG GAG GAG GGC TGG CTG GTG TTT GAC 729 Asp Ser Arg Thr Leu Trp Ala Ser Glu Glu Gly Trp Leu Val Phe Asp
215 220 225
ATC ACA GCC ACC AGC AAC CAC TGG GTG GTC AAT CCG CGG CAC AAC CTG 777 Ile Thr Ala Thr Ser Asn His Trp Val Val Asn Pro Arg His Asn Leu
230 235 240
GGC CTG CAG CTC TCG GTG GAG ACG CTG GAT GGG CAG AGC ATC AAC CCC 825 Gly Leu Gln Leu Ser Val Glu Thr Leu Asp Gly Gln Ser Ile Asn Pro
245 250 255 AAG TTG GCG GGC CTG ATT GGG CGG CAC GGG CCC CAG AAC AAG CAG CCC 873 Lys Leu Ala Gly Leu Ile Gly Arg His Gly Pro Gln Asn Lys Gln Pro
260 265 270 275
TTC ATG GTG GCT TTC TTC AAG GCC ACG GAG GTC CAC TTC CGC AGC ATC 921 Phe Met Val Ala Phe Phe Lys Ala Thr Glu Val His Phe Arg Ser Ile
280 285 290
CGG TCC ACG GGG AGC AAA CAG CGC AGC CAG AAC CGC TCC AAG ACG CCC 969 Arg Ser Thr Gly Ser Lys Gln Arg Ser Gln Asn Arg Ser Lys Thr Pro
295 300 305
AAG AAC CAG GAA GCC CTG CGG ATG GCC AAC GTG GCA GAG AAC AGC AGC 1017 Lys Asn Gln Glu Ala Leu Arg Met Ala Asn Val Ala Glu Asn Ser Ser
310 315 320
AGC GAC CAG AGG CAG GCC TGT AAG AAG CAC GAG CTG TAT GTC AGC TTC 1065 Ser Asp Gln Arg Gln Ala Cys Lys Lys His Glu Leu Tyr Val Ser Phe
325 330 335
CGA GAC CTG GGC TGG CAG GAC TGG ATC ATC GCG CCT GAA GGC TAC GCC 1113 Arg Asp Leu Gly Trp Gln Asp Trp Ile Ile Ala Pro Glu Gly Tyr Ala
340 345 350 355
GCC TAC TAC TGT GAG GGG GAG TGT GCC TTC CCT CTG AAC TCC TAC ATG 1161 Ala Tyr Tyr Cys Glu Gly Glu Cys Ala Phe Pro Leu Asn Ser Tyr Met
360 365 370
AAC GCC ACC AAC CAC GCC ATC GTG CAG ACG CTG GTC CAC TTC ATC AAC 1209 Asn Ala Thr Asn His Ala Ile Val Gln Thr Leu Val His Phe Ile Asn
375 380 385
CCG GAA ACG GTG CCC AAG CCC TGC TGT GCG CCC ACG CAG CTC AAT GCC 1257 Pro Glu Thr Val Pro Lys Pro Cys Cys Ala Pro Thr Gln Leu Asn Ala
390 395 400
ATC TCC GTC CTC TAC TTC GAT GAC AGC TCC AAC GTC ATC CTG AAG AAA 1305 Ile Ser Val Leu Tyr Phe Asp Asp Ser Ser Asn Val Ile Leu Lys Lys
405 410 415
TAC AGA AAC ATG GTG GTC CGG GCC TGT GGC TGC CAC TAGCTCCTCC 1351
Tyr Arg Asn Met Val Val Arg Ala Cys Gly Cys His
420 425 430
GAGAATTCAG ACCCTTTGGG GCCAAGTTTT TCTGGATCCT CCATTGCTCG CCTTGGCCAG 1411
GAACCAGCAG ACCAACTGCC TTTTGTGAGA CCTTCCCCTC CCTATCCCCA ACTTTAAAGG 1471 TGTGAGAGTA TTAGGAAACA TGAGCAGCAT ATGGCTTTTG ATCAGTTTTT CAGTGGCAGC 1531
ATCCAATGAA CAAGATCCTA CAAGCTGTGC AGGCAAAACC TAGCAGGAAA AAAAAACAAC 1591 GCATAAAGAA AAATGGCCGG GCCAGGTCAT TGGCTGGGAA GTCTCAGCCA TGCACGGACT 1651
CGTTTCCAGA GGTAATTATG AGCGCCTACC AGCCAGGCCA CCCAGCCGTG GGAGGAAGGG 1711
GGCGTGGCAA GGGGTGGGCA CATTGGTGTC TGTGCGAAAG GAAAATTGAC CCGGAAGTTC 1771
CTGTAATAAA TGTCACAATA AAACGAATGA ATGAAAAAAA AAAAAAAAAA A 1822
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 431 amino acids
(B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
Met His Val Arg Ser Leu Arg Ala Ala Ala Pro His Ser Phe Val Ala
1 5 10 15
Leu Trp Ala Pro Leu Phe Leu Leu Arg Ser Ala Leu Ala Asp Phe Ser
20 25 30
Leu Asp Asn Glu Val His Ser Ser Phe Ile His Arg Arg Leu Arg Ser
35 40 45
Gln Glu Arg Arg Glu Met Gln Arg Glu Ile Leu Ser Ile Leu Gly Leu
50 55 60
Pro His Arg Pro Arg Pro His Leu Gln Gly Lys His Asn Ser Ala Pro
65 70 75 80
Met Phe Met Leu Asp Leu Tyr Asn Ala Met Ala Val Glu Glu Gly Gly
85 90 95
Gly Pro Gly Gly Gln Gly Phe Ser Tyr Pro Tyr Lys Ala Val Phe Ser
100 105 110
Thr Gln Gly Pro Pro Leu Ala Ser Leu Gln Asp Ser His Phe Leu Thr
115 120 125 Asp Ala Asp Met Val Met Ser Phe Val Asn Leu Val Glu His Asp Lys 130 135 140
Glu Phe Phe His Pro Arg Tyr His His Arg Glu Phe Arg Phe Asp Leu 145 150 155 160
Ser Lys Ile Pro Glu Gly Glu Ala Val Thr Ala Ala Glu Phe Arg Ile
165 170 175 Tyr Lys Asp Tyr Ile Arg Glu Arg Phe Asp Asn Glu Thr Phe Arg Ile
180 185 190
Ser Val Tyr Gln Val Leu Gln Glu His Leu Gly Arg Glu Ser Asp Leu
195 200 205
Phe Leu Leu Asp Ser Arg Thr Leu Trp Ala Ser Glu Glu Gly Trp Leu 210 215 220
Val Phe Asp Ile Thr Ala Thr Ser Asn His Trp Val Val Asn Pro Arg 225 230 235 240
His Asn Leu Gly Leu Gln Leu Ser Val Glu Thr Leu Asp Gly Gln Ser
245 250 255 Ile Asn Pro Lys Leu Ala Gly Leu Ile Gly Arg His Gly Pro Gln Asn
260 265 270
Lys Gln Pro Phe Met Val Ala Phe Phe Lys Ala Thr Glu Val His Phe
275 280 285
Arg Ser Ile Arg Ser Thr Gly Ser Lys Gln Arg Ser Gln Asn Arg Ser 290 295 300
Lys Thr Pro Lys Asn Gln Glu Ala Leu Arg Met Ala Asn Val Ala Glu 305 310 315 320
Asn Ser Ser Ser Asp Gln Arg Gln Ala Cys Lys Lys His Glu Leu Tyr
325 330 335 Val Ser Phe Arg Asp Leu Gly Trp Gln Asp Trp Ile Ile Ala Pro Glu
340 345 350
Gly Tyr Ala Ala Tyr Tyr Cys Glu Gly Glu Cys Ala Phe Pro Leu Asn
355 360 365
Ser Tyr Met Asn Ala Thr Asn His Ala Ile Val Gln Thr Leu Val His
370 375 380
Phe Ile Asn Pro Glu Thr Val Pro Lys Pro Cys Cys Ala Pro Thr Gln 385 390 395 400 Leu Asn Ala Ile Ser Val Leu Tyr Phe Asp Asp Ser Ser Asn Val Ile
405 410 415
Leu Lys Lys Tyr Arg Asn Met Val Val Arg Ala Cys Gly Cys His
420 425 430
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1873 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: MURIDAE
(F) TISSUE TYPE: EMBRYO
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 104..1393
(D) OTHER INFORMATION: /function= "OSTEOGENIC PROTEIN" /product= "MOP1-PP"
/note= "M0P1 (cDNA)"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5:
CTGCAGCAAG TGACCTCGGG TCGTGGACCG CTGCCCTGCC CCCTCCGCTG CCACCTGGGG 60
CGGCGCGGGC CCGGTGCCCC GGATCGCGCG TAGAGCCGGC GCG ATG CAC GTG CGC 115
Met His Val Arg
1
TCG CTG CGC GCT GCG GCG CCA CAC AGC TTC GTG GCG CTC TGG GCG CCT 163
Ser Leu Arg Ala Ala Ala Pro His Ser Phe Val Ala Leu Trp Ala Pro
5 10 15 20
CTG TTC TTG CTG CGC TCC GCC CTG GCC GAT TTC AGC CTG GAC AAC GAG 211 Leu Phe Leu Leu Arg Ser Ala Leu Ala Asp Phe Ser Leu Asp Asn Glu
25 30 35 GTG CAC TCC AGC TTC ATC CAC CGG CGC CTC CGC AGC CAG GAG CGG CGG 259 Val His Ser Ser Phe Ile His Arg Arg Leu Arg Ser Gln Glu Arg Arg
40 45 50
GAG ATG CAG CGG GAG ATC CTG TCC ATC TTA GGG TTG CCC CAT CGC CCG 307 Glu Met Gln Arg Glu Ile Leu Ser Ile Leu Gly Leu Pro His Arg Pro
55 60 65
CGC CCG CAC CTC CAG GGA AAG CAT AAT TCG GCG CCC ATG TTC ATG TTG 355 Arg Pro His Leu Gln Gly Lys His Asn Ser Ala Pro Met Phe Met Leu
70 75 80
GAC CTG TAC AAC GCC ATG GCG GTG GAG GAG AGC GGG CCG GAC GGA CAG 403
Asp Leu Tyr Asn Ala Met Ala Val Glu Glu Ser Gly Pro Asp Gly Gln
85 90 95 100
GGC TTC TCC TAC CCC TAC AAG GCC GTC TTC AGT ACC CAG GGC CCC CCT 451 Gly Phe Ser Tyr Pro Tyr Lys Ala Val Phe Ser Thr Gln Gly Pro Pro
105 110 115
TTA GCC AGC CTG CAG GAC AGC CAT TTC CTC ACT GAC GCC GAC ATG GTC 499 Leu Ala Ser Leu Gln Asp Ser His Phe Leu Thr Asp Ala Asp Met Val
120 125 130
ATG AGC TTC GTC AAC CTA GTG GAA CAT GAC AAA GAA TTC TTC CAC CCT 547 Met Ser Phe Val Asn Leu Val Glu His Asp Lys Glu Phe Phe His Pro
135 140 145
CGA TAC CAC CAT CGG GAG TTC CGG TTT GAT CTT TCC AAG ATC CCC GAG 595 Arg Tyr His His Arg Glu Phe Arg Phe Asp Leu Ser Lys Ile Pro Glu
150 155 160
GGC GAA CGG GTG ACC GCA GCC GAA TTC AGG ATC TAT AAG GAC TAC ATC 643
Gly Glu Arg Val Thr Ala Ala Glu Phe Arg Ile Tyr Lys Asp Tyr Ile
165 170 175 180
CGG GAG CGA TTT GAC AAC GAG ACC TTC CAG ATC ACA GTC TAT CAG GTG 691 Arg Glu Arg Phe Asp Asn Glu Thr Phe Gln Ile Thr Val Tyr Gln Val
185 190 195
CTC CAG GAG CAC TCA GGC AGG GAG TCG GAC CTC TTC TTG CTG GAC AGC 739 Leu Gln Glu His Ser Gly Arg Glu Ser Asp Leu Phe Leu Leu Asp Ser
200 205 210
CGC ACC ATC TGG GCT TCT GAG GAG GGC TGG TTG GTG TTT GAT ATC ACA 787 Arg Thr Ile Trp Ala Ser Glu Glu Gly Trp Leu Val Phe Asp Ile Thr
215 220 225 GCC ACC AGC AAC CAC TGG GTG GTC AAC CCT CGG CAC AAC CTG GGC TTA 835 Ala Thr Ser Asn His Trp Val Val Asn Pro Arg His Asn Leu Gly Leu
230 235 240
CAG CTC TCT GTG GAG ACC CTG GAT GGG CAG AGC ATC AAC CCC AAG TTG 883 Gln Leu Ser Val Glu Thr Leu Asp Gly Gln Ser Ile Asn Pro Lys Leu
245 250 255 260
GCA GGC CTG ATT GGA CGG CAT GGA CCC CAG AAC AAG CAA CCC TTC ATG 931 Ala Gly Leu Ile Gly Arg His Gly Pro Gln Asn Lys Gln Pro Phe Met
265 270 275
GTG GCC TTC TTC AAG GCC ACG GAA GTC CAT CTC CGT AGT ATC CGG TCC 979 Val Ala Phe Phe Lys Ala Thr Glu Val His Leu Arg Ser Ile Arg Ser
280 285 290
ACG GGG GGC AAG CAG CGC AGC CAG AAT CGC TCC AAG ACG CCA AAG AAC 1027 Thr Gly Gly Lys Gln Arg Ser Gln Asn Arg Ser Lys Thr Pro Lys Asn
295 300 305
CAA GAG GCC CTG AGG ATG GCC AGT GTG GCA GAA AAC AGC AGC AGT GAC 1075 Gln Glu Ala Leu Arg Met Ala Ser Val Ala Glu Asn Ser Ser Ser Asp
310 315 320
CAG AGG CAG GCC TGC AAG AAA CAT GAG CTG TAC GTC AGC TTC CGA GAC 1123 Gln Arg Gln Ala Cys Lys Lys His Glu Leu Tyr Val Ser Phe Arg Asp
325 330 335 340
CTT GGC TGG CAG GAC TGG ATC ATT GCA CCT GAA GGC TAT GCT GCC TAC 1171 Leu Gly Trp Gln Asp Trp Ile Ile Ala Pro Glu Gly Tyr Ala Ala Tyr
345 350 355
TAC TGT GAG GGA GAG TGC GCC TTC CCT CTG AAC TCC TAC ATG AAC GCC 1219 Tyr Cys Glu Gly Glu Cys Ala Phe Pro Leu Asn Ser Tyr Met Asn Ala
360 365 370
ACC AAC CAC GCC ATC GTC CAG ACA CTG GTT CAC TTC ATC AAC CCA GAC 1267 Thr Asn His Ala Ile Val Gln Thr Leu Val His Phe Ile Asn Pro Asp
375 380 385
ACA GTA CCC AAG CCC TGC TGT GCG CCC ACC CAG CTC AAC GCC ATC TCT 1315 Thr Val Pro Lys Pro Cys Cys Ala Pro Thr Gln Leu Asn Ala Ile Ser
390 395 400
GTC CTC TAC TTC GAC GAC AGC TCT AAT GTC GAC CTG AAG AAG TAC AGA 1363 Val Leu Tyr Phe Asp Asp Ser Ser Asn Val Asp Leu Lys Lys Tyr Arg
405 410 415 420 AAC ATG GTG GTC CGG GCC TGT GGC TGC CAC TAGCTCTTCC TGAGACCCTG 1413 Asn Met Val Val Arg Ala Cys Gly Cys His
425 430
ACCTTTGCGG GGCCACACCT TTCCAAATCT TCGATGTCTC ACCATCTAAG TCTCTCACTG 1473
CCCACCTTGG CGAGGAGAAC AGACCAACCT CTCCTGAGCC TTCCCTCACC TCCCAACCGG 1533
AAGCATGTAA GGGTTCCAGA AACCTGAGCG TGCAGCAGCT GATGAGCGCC CTTTCCTTCT 1593
GGCACGTGAC GGACAAGATC CTACCAGCTA CCACAGCAAA .CGCCTAAGAG CAGGAAAAAT 1653
GTCTGCCAGG AAAGTGTCCA GTGTCCACAT GGCCCCTGGC GCTCTGAGTC TTTGAGGAGT 1713 AATCGCAAGC CTCGTTCAGC TGCAGCAGAA GGAAGGGCTT AGCCAGGGTG GGCGCTGGCG 1773
TCTGTGTTGA AGGGAAACCA AGCAGAAGCC ACTGTAATGA TATGTCACAA TAAAACCCAT 1833
GAATGAAAAA AAAAAAAAAA AAAAAAAAAA AAAAGAATTC 1873
(2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 430 amino acids
(B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:
Met His Val Arg Ser Leu Arg Ala Ala Ala Pro His Ser Phe Val Ala
1 5 10 15
Leu Trp Ala Pro Leu Phe Leu Leu Arg Ser Ala Leu Ala Asp Phe Ser
20 25 30
Leu Asp Asn Glu Val His Ser Ser Phe Ile His Arg Arg Leu Arg Ser
35 40 45
Gln Glu Arg Arg Glu Met Gln Arg Glu Ile Leu Ser Ile Leu Gly Leu
50 55 60
Pro His Arg Pro Arg Pro His Leu Gln Gly Lys His Asn Ser Ala Pro
65 70 75 80 Met Phe Met Leu Asp Leu Tyr Asn Ala Met Ala Val Glu Glu Ser Gly 85 90 95
Pro Asp Gly Gln Gly Phe Ser Tyr Pro Tyr Lys Ala Val Phe Ser Thr
100 105 110
Gln Gly Pro Pro Leu Ala Ser Leu Gln Asp Ser His Phe Leu Thr Asp
115 120 125
Ala Asp Met Val Met Ser Phe Val Asn Leu Val Glu His Asp Lys Glu 130 135 140
Phe Phe His Pro Arg Tyr His His Arg Glu Phe Arg Phe Asp Leu Ser 145 150 155 160
Lys Ile Pro Glu Gly Glu Arg Val Thr Ala Ala Glu Phe Arg Ile Tyr
165 170 175
Lys Asp Tyr Ile Arg Glu Arg Phe Asp Asn Glu Thr Phe Gln Ile Thr
180 185 190
Val Tyr Gln Val Leu Gln Glu His Ser Gly Arg Glu Ser Asp Leu Phe
195 200 205
Leu Leu Asp Ser Arg Thr Ile Trp Ala Ser Glu Glu Gly Trp Leu Val 210 215 220
Phe Asp Ile Thr Ala Thr Ser Asn His Trp Val Val Asn Pro Arg His 225 230 235 240
Asn Leu Gly Leu Gln Leu Ser Val Glu Thr Leu Asp Gly Gln Ser Ile
245 250 255
Asn Pro Lys Leu Ala Gly Leu Ile Gly Arg His Gly Pro Gln Asn Lys
260 265 270
Gln Pro Phe Met Val Ala Phe Phe Lys Ala Thr Glu Val His Leu Arg
275 280 285
Ser Ile Arg Ser Thr Gly Gly Lys Gln Arg Ser Gln Asn Arg Ser Lys 290 295 300
Thr Pro Lys Asn Gln Glu Ala Leu Arg Met Ala Ser Val Ala Glu Asn 305 310 315 320
Ser Ser Ser Asp Gln Arg Gln Ala Cys Lys Lys His Glu Leu Tyr Val
325 330 335 Ser Phe Arg Asp Leu Gly Trp Gln Asp Trp Ile Ile Ala Pro Glu Gly
340 345 350
Tyr Ala Ala Tyr Tyr Cys Glu Gly Glu Cys Ala Phe Pro Leu Asn Ser
355 360 365
Tyr Met Asn Ala Thr Asn His Ala Ile Val Gln Thr Leu Val His Phe
370 375 380
Ile Asn Pro Asp Thr Val Pro Lys Pro Cys Cys Ala Pro Thr Gln Leu
385 390 395 400
Asn Ala Ile Ser Val Leu Tyr Phe Asp Asp Ser Ser Asn Val Asp Leu
405 410 415
Lys Lys Tyr Arg Asn Met Val Val Arg Ala Cys Gly Cys His
420 425 430
(2) INFORMATION FOR SEQ ID NO: 7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1723 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens
(F) TISSUE TYPE: HIPPOCAMPUS
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 490..1696
(D) OTHER INFORMATION: /function= "OSTEOGENIC PROTEIN"
/product= "hOP2-PP"
/note= "hOP2 (cDNA)"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
GGCGCCGGCA GAGCAGGAGT GGCTGGAGGA GCTGTGGTTG GAGCAGGAGG TGGCACGGCA 60 GGGCTGGAGG GCTCCCTATG AGTGGCGGAG ACGGCCCAGG AGGCGCTGGA GCAACAGCTC 120
CCACACCGCA CCAAGCGGTG GCTGCAGGAG CTCGCCCATC GCCCCTGCGC TGCTCGGACC 180 GCGGCCACAG CCGGACTGGC GGGTACGGCG GCGACAGAGG CATTGGCCGA GAGTCCCAGT 240
CCGCAGAGTA GCCCCGGCCT CGAGGCGGTG GCGTCCCGGT CCTCTCCGTC CAGGAGCCAG 300
GACAGGTGTC GCGCGGCGGG GCTCCAGGGA CCGCGCCTGA GGCCGGCTGC CCGCCCGTCC 360
CGCCCCGCCC CGCCGCCCGC CGCCCGCCGA GCCCAGCCTC CTTGCCGTCG GGGCGTCCCC 420
AGGCCCTGGG TCGGCCGCGG AGCCGATGCG CGCCCGCTGA GCGCCCCAGC TGAGCGCCCC 480
CGGCCTGCC ATG ACC GCG CTC CCC GGC CCG CTC TGG CTC CTG GGC CTG 528
Met Thr Ala Leu Pro Gly Pro Leu Trp Leu Leu Gly Leu
1 5 10
GCG CTA TGC GCG CTG GGC GGG GGC GGC CCC GGC CTG CGA CCC CCG CCC 576 Ala Leu Cys Ala Leu Gly Gly Gly Gly Pro Gly Leu Arg Pro Pro Pro
15 20 25
GGC TGT CCC CAG CGA CGT CTG GGC GCG CGC GAG CGC CGG GAC GTG CAG 624 Gly Cys Pro Gln Arg Arg Leu Gly Ala Arg Glu Arg Arg Asp Val Gln
30 35 40 45
CGC GAG ATC CTG GCG GTG CTC GGG CTG CCT GGG CGG CCC CGG CCC CGC 672 Arg Glu Ile Leu Ala Val Leu Gly Leu Pro Gly Arg Pro Arg Pro Arg
50 55 60
GCG CCA CCC GCC GCC TCC CGG CTG CCC GCG TCC GCG CCG CTC TTC ATG 720 Ala Pro Pro Ala Ala Ser Arg Leu Pro Ala Ser Ala Pro Leu Phe Met
65 70 75
CTG GAC CTG TAC CAC GCC ATG GCC GGC GAC GAC GAC GAG GAC GGC GCG 768 Leu Asp Leu Tyr His Ala Met Ala Gly Asp Asp Asp Glu Asp Gly Ala
80 85 90
CCC GCG GAG CGG CGC CTG GGC CGC GCC GAC CTG GTC ATG AGC TTC GTT 816 Pro Ala Glu Arg Arg Leu Gly Arg Ala Asp Leu Val Met Ser Phe Val
95 100 105
AAC ATG GTG GAG CGA GAC CGT GCC CTG GGC CAC CAG GAG CCC CAT TGG 864 Asn Met Val Glu Arg Asp Arg Ala Leu Gly His Gln Glu Pro His Trp
110 115 120 125
AAG GAG TTC CGC TTT GAC CTG ACC CAG ATC CCG GCT GGG GAG GCG GTC 912 Lys Glu Phe Arg Phe Asp Leu Thr Gln Ile Pro Ala Gly Glu Ala Val
130 135 140
ACA GCT GCG GAG TTC CGG ATT TAC AAG GTG CCC AGC ATC CAC CTG CTC 960 Thr Ala Ala Glu Phe Arg Ile Tyr Lys Val Pro Ser Ile His Leu Leu
145 150 155 AAC AGG ACC CTC CAC GTC AGC ATG TTC CAG GTG GTC CAG GAG CAG TCC 1008 Asn Arg Thr Leu His Val Ser Met Phe Gln Val Val Gln Glu Gln Ser
160 165 170
AAC AGG GAG TCT GAC TTG TTC TTT TTG GAT CTT CAG ACG CTC CGA GCT 1056 Asn Arg Glu Ser Asp Leu Phe Phe Leu Asp Leu Gln Thr Leu Arg Ala
175 180 185
GGA GAC GAG GGC TGG CTG GTG CTG GAT GTC ACA GCA GCC AGT GAC TGC 1104 Gly Asp Glu Gly Trp Leu Val Leu Asp Val Thr Ala Ala Ser Asp Cys
190 195 200 205
TGG TTG CTG AAG CGT CAC AAG GAC CTG GGA CTC CGC CTC TAT GTG GAG 1152 Trp Leu Leu Lys Arg His Lys Asp Leu Gly Leu Arg Leu Tyr Val Glu
210 215 220
ACT GAG GAC GGG CAC AGC GTG GAT CCT GGC CTG GCC GGC CTG CTG GGT 1200
Thr Glu Asp Gly His Ser Val Asp Pro Gly Leu Ala Gly Leu Leu Gly
225 230 235
CAA CGG GCC CCA CGC TCC CAA CAG CCT TTC GTG GTC ACT TTC TTC AGG 1248 Gln Arg Ala Pro Arg Ser Gln Gln Pro Phe Val Val Thr Phe Phe Arg
240 245 250
GCC AGT CCG AGT CCC ATC CGC ACC CCT CGG GCA GTG AGG CCA CTG AGG 1296 Ala Ser Pro Ser Pro Ile Arg Thr Pro Arg Ala Val Arg Pro Leu Arg
255 260 265
AGG AGG CAG CCG AAG AAA AGC AAC GAG CTG CCG CAG GCC AAC CGA CTC 1344 Arg Arg Gln Pro Lys Lys Ser Asn Glu Leu Pro Gln Ala Asn Arg Leu
270 275 280 285
CCA GGG ATC TTT GAT GAC GTC CAC GGC TCC CAC GGC CGG CAG GTC TGC 1392 Pro Gly Ile Phe Asp Asp Val His Gly Ser His Gly Arg Gln Val Cys
290 295 300
CGT CGG CAC GAG CTC TAC GTC AGC TTC CAG GAC CTC GGC TGG CTG GAC 1440 Arg Arg His Glu Leu Tyr Val Ser Phe Gln Asp Leu Gly Trp Leu Asp
305 310 315
TGG GTC ATC GCT CCC CAA GGC TAC TCG GCC TAT TAC TGT GAG GGG GAG 1488 Trp Val Ile Ala Pro Gln Gly Tyr Ser Ala Tyr Tyr Cys Glu Gly Glu
320 325 330
TGC TCC TTC CCA CTG GAC TCC TGC ATG AAT GCC ACC AAC CAC GCC ATC 1536 Cys Ser Phe Pro Leu Asp Ser Cys Met Asn Ala Thr Asn His Ala Ile
335 340 345 CTG CAG TCC CTG GTG CAC CTG ATG AAG CCA AAC GCA GTC CCC AAG GCG 1584 Leu Gln Ser Leu Val His Leu Met Lys Pro Asn Ala Val Pro Lys Ala
350 355 360 365
TGC TGT GCA CCC ACC AAG CTG AGC GCC ACC TCT GTG CTC TAC TAT GAC 1632 Cys Cys Ala Pro Thr Lys Leu Ser Ala Thr Ser Val Leu Tyr Tyr Asp
370 375 380
AGC AGC AAC AAC GTC ATC CTG CGC AAA GCC CGC AAC ATG GTG GTC AAG 1680 Ser Ser Asn Asn Val Ile Leu Arg Lys Ala Arg Asn Met Val Val Lys
385 390 395
GCC TGC GGC TGC CAC T GAGTCAGCCC GCCCAGCCCT ACTGCAG 1723
Ala Cys Gly Cys His
400
(2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 402 amino acids
(B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
Met Thr Ala Leu Pro Gly Pro Leu Trp Leu Leu Gly Leu Ala Leu Cys
1 5 10 15
Ala Leu Gly Gly Gly Gly Pro Gly Leu Arg Pro Pro Pro Gly Cys Pro
20 25 30
Gln Arg Arg Leu Gly Ala Arg Glu Arg Arg Asp Val Gln Arg Glu Ile
35 40 45
Leu Ala Val Leu Gly Leu Pro Gly Arg Pro Arg Pro Arg Ala Pro Pro
50 55 60
Ala Ala Ser Arg Leu Pro Ala Ser Ala Pro Leu Phe Met Leu Asp Leu
65 70 75 80
Tyr His Ala Met Ala Gly Asp Asp Asp Glu Asp Gly Ala Pro Ala Glu
85 90 95
Arg Arg Leu Gly Arg Ala Asp Leu Val Met Ser Phe Val Asn Met Val
100 105 110 Glu Arg Asp Arg Ala Leu Gly His Gln Glu Pro His Trp Lys Glu Phe 115 120 125
Arg Phe Asp Leu Thr Gln Ile Pro Ala Gly Glu Ala Val Thr Ala Ala 130 135 140
Glu Phe Arg Ile Tyr Lys Val Pro Ser Ile His Leu Leu Asn Arg Thr 145 150 155 160 Leu His Val Ser Met Phe Gln Val Val Gln Glu Gln Ser Asn Arg Glu
165 170 175
Ser Asp Leu Phe Phe Leu Asp Leu Gln Thr Leu Arg Ala Gly Asp Glu
180 185 190
Gly Trp Leu Val Leu Asp Val Thr Ala Ala Ser Asp Cys Trp Leu Leu
195 200 205
Lys Arg His Lys Asp Leu Gly Leu Arg Leu Tyr Val Glu Thr Glu Asp 210 215 220
Gly His Ser Val Asp Pro Gly Leu Ala Gly Leu Leu Gly Gln Arg Ala 225 230 235 240 Pro Arg Ser Gln Gln Pro Phe Val Val Thr Phe Phe Arg Ala Ser Pro
245 250 255
Ser Pro Ile Arg Thr Pro Arg Ala Val Arg Pro Leu Arg Arg Arg Gln
260 265 270
Pro Lys Lys Ser Asn Glu Leu Pro Gln Ala Asn Arg Leu Pro Gly Ile
275 280 285
Phe Asp Asp Val His Gly Ser His Gly Arg Gln Val Cys Arg Arg His 290 295 300
Glu Leu Tyr Val Ser Phe Gln Asp Leu Gly Trp Leu Asp Trp Val Ile 305 310 315 320 Ala Pro Gln Gly Tyr Ser Ala Tyr Tyr Cys Glu Gly Glu Cys Ser Phe
325 330 335
Pro Leu Asp Ser Cys Met Asn Ala Thr Asn His Ala Ile Leu Gln Ser
340 345 350
Leu Val His Leu Met Lys Pro Asn Ala Val Pro Lys Ala Cys Cys Ala
355 360 365 Pro Thr Lys Leu Ser Ala Thr Ser Val Leu Tyr Tyr Asp Ser Ser Asn
370 375 380
Asn Val Ile Leu Arg Lys Ala Arg Asn Met Val Val Lys Ala Cys Gly
385 390 395 400
Cys His (2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1926 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(vi) ORIGINAL SOURCE:
(A) ORGANISM: MURIDAE
(F) TISSUE TYPE: EMBRYO
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 93..1289
(D) OTHER INFORMATION: /function= "OSTEOGENIC PROTEIN"
/product= "mOP2-PP"
/note= "mOP2 cDNA" (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
GCCAGGCACA GGTGCGCCGT CTGGTCCTCC CCGTCTGGCG TCAGCCGAGC CCGACCAGCT 60
ACCAGTGGAT GCGCGCCGGC TGAAAGTCCG AG ATG GCT ATG CGT CCC GGG CCA 113
Met Ala Met Arg Pro Gly Pro
1 5
CTC TGG CTA TTG GGC CTT GCT CTG TGC GCG CTG GGA GGC GGC CAC GGT 161 Leu Trp Leu Leu Gly Leu Ala Leu Cys Ala Leu Gly Gly Gly His Gly
10 15 20
CCG CGT CCC CCG CAC ACC TGT CCC CAG CGT CGC CTG GGA GCG CGC GAG 209 Pro Arg Pro Pro His Thr Cys Pro Gln Arg Arg Leu Gly Ala Arg Glu
25 30 35
CGC CGC GAC ATG CAG CGT GAA ATC CTG GCG GTG CTC GGG CTA CCG GGA 257
Arg Arg Asp Met Gln Arg Glu Ile Leu Ala Val Leu Gly Leu Pro Gly
40 45 50 55 CGG CCC CGA CCC CGT GCA CAA CCC GCC GCT GCC CGG CAG CCA GCG TCC 305 Arg Pro Arg Pro Arg Ala Gln Pro Ala Ala Ala Arg Gln Pro Ala Ser
60 65 70
GCG CCC CTC TTC ATG TTG GAC CTA TAC CAC GCC ATG ACC GAT GAC GAC 353 Ala Pro Leu Phe Met Leu Asp Leu Tyr His Ala Met Thr Asp Asp Asp
75 80 85
GAC GGC GGG CCA CCA CAG GCT CAC TTA GGC CGT GCC GAC CTG GTC ATG 401 Asp Gly Gly Pro Pro Gln Ala His Leu Gly Arg Ala Asp Leu Val Met
90 95 100
AGC TTC GTC AAC ATG GTG GAA CGC GAC CGT ACC CTG GGC TAC CAG GAG 449 Ser Phe Val Asn Met Val Glu Arg Asp Arg Thr Leu Gly Tyr Gln Glu
105 110 115
CCA CAC TGG AAG GAA TTC CAC TTT GAC CTA ACC CAG ATC CCT GCT GGG 497 Pro His Trp Lys Glu Phe His Phe Asp Leu Thr Gln Ile Pro Ala Gly
120 125 130 135
GAG GCT GTC ACA GCT GCT GAG TTC CGG ATC TAC AAA GAA CCC AGC ACC 545 Glu Ala Val Thr Ala Ala Glu Phe Arg Ile Tyr Lys Glu Pro Ser Thr
140 145 150
CAC CCG CTC AAC ACA ACC CTC CAC ATC AGC ATG TTC GAA GTG GTC CAA 593 His Pro Leu Asn Thr Thr Leu His Ile Ser Met Phe Glu Val Val Gln
155 160 165
GAG CAC TCC AAC AGG GAG TCT GAC TTG TTC TTT TTG GAT CTT CAG ACG 641 Glu His Ser Asn Arg Glu Ser Asp Leu Phe Phe Leu Asp Leu Gln Thr
170 175 180
CTC CGA TCT GGG GAC GAG GGC TGG CTG GTG CTG GAC ATC ACA GCA GCC 689 Leu Arg Ser Gly Asp Glu Gly Trp Leu Val Leu Asp Ile Thr Ala Ala
185 190 195
AGT GAC CGA TGG CTG CTG AAC CAT CAC AAG GAC CTG GGA CTC CGC CTC 737 Ser Asp Arg Trp Leu Leu Asn His His Lys Asp Leu Gly Leu Arg Leu
200 205 210 215
TAT GTG GAA ACC GCG GAT GGG CAC AGC ATG GAT CCT GGC CTG GCT GGT 785 Tyr Val Glu Thr Ala Asp Gly His Ser Met Asp Pro Gly Leu Ala Gly
220 225 230 CTG CTT GGA CGA CAA GCA CCA CGC TCC AGA CAG CCT TTC ATG GTA ACC 833 Leu Leu Gly Arg Gln Ala Pro Arg Ser Arg Gln Pro Phe Met Val Thr
235 240 245 TTC TTC AGG GCC AGC CAG AGT CCT GTG CGG GCC CCT CGG GCA GCG AGA 881 Phe Phe Arg Ala Ser Gln Ser Pro Val Arg Ala Pro Arg Ala Ala Arg
250 255 260
CCA CTG AAG AGG AGG CAG CCA AAG AAA ACG AAC GAG CTT CCG CAC CCC 929 Pro Leu Lys Arg Arg Gln Pro Lys Lys Thr Asn Glu Leu Pro His Pro
265 270 275
AAC AAA CTC CCA GGG ATC TTT GAT GAT GGC CAC GGT TCC CGC GGC AGA 977 Asn Lys Leu Pro Gly Ile Phe Asp Asp Gly His Gly Ser Arg Gly Arg
280 285 290 295
GAG GTT TGC CGC AGG CAT GAG CTC TAC GTC AGC TTC CGT GAC CTT GGC 1025 Glu Val Cys Arg Arg His Glu Leu Tyr Val Ser Phe Arg Asp Leu Gly
300 305 310
TGG CTG GAC TGG GTC ATC GCC CCC CAG GGC TAC TCT GCC TAT TAC TGT 1073 Trp Leu Asp Trp Val Ile Ala Pro Gln Gly Tyr Ser Ala Tyr Tyr Cys
315 320 325
GAG GGG GAG TGT GCT TTC CCA CTG GAC TCC TGT ATG AAC GCC ACC AAC 1121 Glu Gly Glu Cys Ala Phe Pro Leu Asp Ser Cys Met Asn Ala Thr Asn
330 335 340
CAT GCC ATC TTG CAG TCT CTG GTG CAC CTG ATG AAG CCA GAT GTT GTC 1169 His Ala Ile Leu Gln Ser Leu Val His Leu Met Lys Pro Asp Val Val
345 350 355
CCC AAG GCA TGC TGT GCA CCC ACC AAA CTG AGT GCC ACC TCT GTG CTG 1217 Pro Lys Ala Cys Cys Ala Pro Thr Lys Leu Ser Ala Thr Ser Val Leu
360 365 370 375
TAC TAT GAC AGC AGC AAC AAT GTC ATC CTG CGT AAA CAC CGT AAC ATG 1265 Tyr Tyr Asp Ser Ser Asn Asn Val Ile Leu Arg Lys His Arg Asn Met
380 385 390
GTG GTC AAG GCC TGT GGC TGC CAC TGAGGCCCCG CCCAGCATCC TGCTTCTACT 1319 Val Val Lys Ala Cys Gly Cys His
395
ACCTTACCAT CTGGCCGGGC CCCTCTCCAG AGGCAGAAAC CCTTCTATGT TATCATAGCT 1379
CAGACAGGGG CAATGGGAGG CCCTTCACTT CCCCTGGCCA CTTCCTGCTA AAATTCTGGT 1439 CTTTCCCAGT TCCTCTGTCC TTCATGGGGT TTCGGGGCTA TCACCCCGCC CTCTCCATCC 1499
TCCTACCCCA AGCATAGACT GAATGCACAC AGCATCCCAG AGCTATGCTA ACTGAGAGGT 1559 CTGGGGTCAG CACTGAAGGC CCACATGAGG AAGACTGATC CTTGGCCATC CTCAGCCCAC 1619
AATGGCAAAT TCTGGATGGT CTAAGAAGGC CCTGGAATTC TAAACTAGAT GATCTGGGCT 1679
CTCTGCACCA TTCATTGTGG CAGTTGGGAC ATTTTTAGGT ATAACAGACA CATACACTTA 1739
GATCAATGCA TCGCTGTACT CCTTGAAATC AGAGCTAGCT TGTTAGAAAA AGAATCAGAG 1799
CCAGGTATAG CGGTGCATGT CATTAATCCC AGCGCTAAAG AGACAGAGAC AGGAGAATCT 1859
CTGTGAGTTC AAGGCCACAT AGAAAGAGCC TGTCTCGGGA GCAGGAAAAA AAAAAAAAAC 1919
GGAATTC 1926
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 399 amino acids
(B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
Met Ala Met Arg Pro Gly Pro Leu Trp Leu Leu Gly Leu Ala Leu Cys
1 5 10 15
Ala Leu Gly Gly Gly His Gly Pro Arg Pro Pro His Thr Cys Pro Gln
20 25 30
Arg Arg Leu Gly Ala Arg Glu Arg Arg Asp Met Gln Arg Glu Ile Leu
35 40 45
Ala Val Leu Gly Leu Pro Gly Arg Pro Arg Pro Arg Ala Gln Pro Ala
50 55 60
Ala Ala Arg Gln Pro Ala Ser Ala Pro Leu Phe Met Leu Asp Leu Tyr
65 70 75 80
His Ala Met Thr Asp Asp Asp Asp Gly Gly Pro Pro Gln Ala His Leu
85 90 95
Gly Arg Ala Asp Leu Val Met Ser Phe Val Asn Met Val Glu Arg Asp
100 105 110 Arg Thr Leu Gly Tyr Gln Glu Pro His Trp Lys Glu Phe His Phe Asp 115 120 125
Leu Thr Gln Ile Pro Ala Gly Glu Ala Val Thr Ala Ala Glu Phe Arg 130 135 140
Ile Tyr Lys Glu Pro Ser Thr His Pro Leu Asn Thr Thr Leu His Ile 145 150 155 160 Ser Met Phe Glu Val Val Gln Glu His Ser Asn Arg Glu Ser Asp Leu
165 170 175
Phe Phe Leu Asp Leu Gln Thr Leu Arg Ser Gly Asp Glu Gly Trp Leu
180 185 190
Val Leu Asp Ile Thr Ala Ala Ser Asp Arg Trp Leu Leu Asn His His
195 200 205
Lys Asp Leu Gly Leu Arg Leu Tyr Val Glu Thr Ala Asp Gly His Ser 210 215 220
Met Asp Pro Gly Leu Ala Gly Leu Leu Gly Arg Gln Ala Pro Arg Ser 225 230 235 240 Arg Gln Pro Phe Met Val Thr Phe Phe Arg Ala Ser Gln Ser Pro Val
245 250 255
Arg Ala Pro Arg Ala Ala Arg Pro Leu Lys Arg Arg Gln Pro Lys Lys
260 265 270
Thr Asn Glu Leu Pro His Pro Asn Lys Leu Pro Gly Ile Phe Asp Asp
275 280 285
Gly His Gly Ser Arg Gly Arg Glu Val Cys Arg Arg His Glu Leu Tyr 290 295 300
Val Ser Phe Arg Asp Leu Gly Trp Leu Asp Trp Val Ile Ala Pro Gln 305 310 315 320 Gly Tyr Ser Ala Tyr Tyr Cys Glu Gly Glu Cys Ala Phe Pro Leu Asp
325 330 335
Ser Cys Met Asn Ala Thr Asn His Ala Ile Leu Gln Ser Leu Val His
340 345 350
Leu Met Lys Pro Asp Val Val Pro Lys Ala Cys Cys Ala Pro Thr Lys
355 360 365 Leu Ser Ala Thr Ser Val Leu Tyr Tyr Asp Ser Ser Asn Asn Val Ile 370 375 380
Leu Arg Lys His Arg Asn Met Val Val Lys Ala Cys Gly Cys His 385 390 395
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6418 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 1..6361
(D) OTHER INFORMATION: /note= "HOP-2 genomic sequence"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 1..837
(D) OTHER INFORMATION: /note= "EXON ONE"
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 884..885
(D) OTHER INFORMATION: /note= "A Gap Occurs Between Positions 884 and 885 in this Sequence"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 1088..1277
(D) OTHER INFORMATION: /note= "EXON TWO"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 1350..1814
(D) OTHER INFORMATION: /note= "EXON THREE"
(ix) FEATURE:
(A) NAME/KEY: misc_feature
(B) LOCATION: 1834..1835
(D) OTHER INFORMATION: /note= "A Gap Occurs Between Positions 1834 and 1835 in this Sequence" (ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 1883..2077
(D) OTHER INFORMATION: /note= "EXON FOUR"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 2902..2981
(D) OTHER INFORMATION: /note= "EXON FIVE"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 3507..3617
(D) OTHER INFORMATION: /note= "EXON SIX"
(ix) FEATURE:
(A) NAME/KEY: exon
(B) LOCATION: 6116..6361
(D) OTHER INFORMATION: /note= "EXON SEVEN"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
GGAATTCCGG CCACAGTGGC GCCGGCAGAG CAGGAGTGGC TGGAGGAGCT GTGGTTGGAG 60
CAGGAGGTGG CACGGCAGGG CTGGAGGGCT CCCTATGAGT GGCGGAGACG GCCCAGGAGG 120
CGCTGGAGCA ACAGCTCCCA CACCGCACCA AGCGGTGGCT GCAGGAGCTC GCCCATCGCC 180 CCTGCGCTGC TCGGACCGCG GCCACAGCCG GACTGGCGGG TACGGCGGCG ACAGACGGAT 240
TGGCCGAGAG TCCCAGTCCG CAGAGTAGCC CCGGCCTCGA GGCGGTGGCG TCCGCGTCCT 300
CTCGTCCAGG AGCCAGGACA GGTGTCGCGC GGCGGGCCGT CCAGGGACCG CGCTGAGGCC 360
GCGGTCGCCC GTCCCGCCCC GCCCCGCCGC CCGCCGCCCG CCGAGCCCAG CCTCCTTGCC 420
GTCGGGGCGT CCCCAGGCCC TGGGTCGGCC GCGGAGCCGA TGCGCGCCCG CTGAGCGCCC 480 CAGCTGAGCG CCCCCGGCCT GCCATGACCG CGCTCCCCGG CCCGCTCTGG CTCCTGGGCC 540
TGGCGCTATG CGCGCTGGGC GGGGGCGGCC CCGGCCTGCG ACCCCCGCCC GGCTGTCCCC 600
AGCGACGTCT GGGCGCGCGC GAGCGCCGGG ACGTGCAGCG CGAGATCCTG GCGGTGCTCG 660
GGCTGCCTGG GCGGCCCCGG CCCCGCGCGC CACCCGCCGC CTCCCGGCTG CCCGCGTCCG 720
CGCCGCTCTT CATGCTGGAC CTGTACCACG CCATGGCCGG CGACGACGAC GAGGACGGCG 780 CGCCCGCGGA GCGGCGCCTG GGCCGCGCCG ACCTGGTCAT GAGCTTCGTT AACATGGGTG 840
AGTGCGGCGC CCGCGCGGGG ACCCTCGGAG TAAACTGGCT GCAGCTGCAG GGCCTCTTCT 900 GGCTCTACAC CCCGGGACCA AGCCTGGAAC AAACGTTTGC ACTAAATGAA GCCGGCCCCA 960
CCCAGGCCTC CCTGGGTCCG CTCCACCTTG AGTGGTGGGT GGCTGGGGGC GGTGGCTCAC 1020
ACCAGCTCTG CCCCCTCCAG AGCCCGAGCC ATTCTGAGTG CCAGCCCAGC GCTGCTTTGT 1080
CTTCTAGTGG AGCGAGACCG TGCCCTGGGC CACCAGGAGC CCCATTGGAA GGAGTTCCGC 1140
TTTGACCTGA CCCAGATCCC GGCTGGGGAG GCGGTCACAG CTGCGGAGTT CCGGATTTAC 1200 AAGGTGCCCA GCATCCACCT GCTCAACAGG ACCCTCCACG TCAGCATGTT CCAGGTGGTC 1260
CAGGAGCAGT CCAACAGGTG CCTTCCCCTT GGCCCGGGTG CCCACCTAAC CCCCCACCTC 1320
ACAGTCTCAT GGTCAAGGCA GCCCAGCAGG GAGTCGTGGT GGGTGAAAGA GAGCCTCAAA 1380
GATGGGAAGG ATGCTTGGCC CGAGGCCCTG CACTGTGGGA AGAGCCCCAG TGACAATCCT 1440
GACTTCAAGT CCCTGCCCTC CATCCTGCTG TGGGGACTTG GACATGGTCA CTGAGACTCA 1500 GTTTCCCCAT GTGTACACCT CTGTGGGCTG AGGCAATGAG ATGAGGCTCA GAAGGGCGCA 1560
GCCAGAGTCA GGTGGGAGAC GCTCCGGTGA CAGCCCCCAG CGGGCCCTGG AGACACGGAG 1620
GCAGCTGTGC CGGCCGCCGG TTAATTGTTC TTTCATGTCC ACAGGGGAGT CTGACTTGTT 1680
CTTTTTGGAT CTTCAGACGC TCCGAGCTGG AGACGAGGGC TGGCTGGTGC TGGATGTCAC 1740
AGCAGCCAGT GACTGCTGGT TGCTGAAGCG TCACAAGGAC CTGGGACTCC GCCTCTATGT 1800 GGAGACTGAG GACGGTGAGG CTGGGGCTCT GCAGCTGCAG AGCCACTGCC CGTGAGTGAC 1860
CCCTCTCTCC TTTCTGTCTC AGGGCACAGC GTGGATCCTG GCCTGGCCGG CCTGCTGGGT 1920
CAACGGGCCC CACGCTCCCA ACAGCCTTTC GTGGTCACTT TCTTCAGGGC CAGTCCGAGT 1980
CCCATCCGCA CCCCTCGGGC AGTGAGGCCA CTGAGGAGGA GGCAGCCGAA GAAAAGCAAC 2040
GAGCTGCCGC AGGCCAACCG ACTCCCAGGG ATCTTTGGTG AGGGTCGGGC AGGCTGGGGC 2100 GAGGCTGTGG CTGTCTGGCT GAGAGAGGCA GGGCGAGAAC CAAGTGGTGG CCCAGAGCCC 2160
AGAGCCTCAG GCTAGGTCGG TTCAAGCTGA CGGCCACTCT CCAGCCACCT TTCCTGACAC 2220 CATCTTGGCC CTGATGCACC CTGGTGACCG GCACTCCGAG GCCTGTCCTG GCTGTCCCTG 2280
CTGCCAGAAG TCTCCCTCTC TCCCCCTGGC TCCTCCGGGT CTTTCTCAGG AGCCTCCTTC 2340 AGAATCAGCT GCCCCTTCCC TGGGAGCCGC AGCCCCTCAT GACCTGCGGT TGTGCCTGGG 2400
CACCTGTGGA TCCTCGGTTG CTTATGCGAT TTTCTCCCCA ACTGGCCAAG CTTCAGGATC 2460
AGGGACAGGC CTGACCCAAC CCCGTGCCCT CCTTCCCAGG GAGTCGGCCC TTGACTGGCC 2520
TGGTCGTGAG CCACTTGAAC CTCGGGAATG GGTGTGGCAG GAGAGGGTGG GCTGGAGTCA 2580
CAGGGGTCTC CAGAGAGGAG GAGGCACAGG ATGGCCGAGG GTCCTGCTGG GCTGTTTACT 2640 GGAGCATAAA GATGCTCATA GGCTGAAGGA CAGGGGAGGA CTGGGCACAG TGTCACTCTA 2700
GCCATTGGGA GCCATGGCAG GCTTCTGAGC TGGGTCATGG TACAAGCAGA GTTCCAGGGA 2760
TGGGCTTTAT GAGCCAAATG GTTTCCTGTC ATTCATTTAT TTGACAAATG TGCTCATCAG 2820
GGCATCCCCC ACCCTGGTAC CCCATAGTAG CTGCACACAG CAGGAACCCC AGAAAAGACC 2880
TTGCCCCTTC TGTCCCTGCA GATGACGTCC ACGGCTCCCA CGGCCGGCAG GTCTGCCGTC 2940 GGCACGAGCT CTACGTCAGC TTCCAGGACC TCGGCTGGCT GGTAATTGCT GACTCTCCTT 3000
GTTTCTGAAA TGACAATCAC CACCTGTAGA TCAGAAGTGA ATCTGCAGGG AGGACATAGA 3060
ATCATGGTGA CTTCAATTTT CTTATGTATT TTTTTCTTCT GTGTTTTCCA AGTTTTCTAA 3120
AGTGAGAATA TGGTGAGAAA GGGTTTTGTT GTTGTTGTTG TGTTTTTTGT TTTTTTTTAA 3180
AAACCCATGA AAATGAAGAC TGAATCAACC AACTAAGCTG TCAGCATTGC CGCAGGGTAA 3240 CTGAGACCTC CCTGCATTGG CTACGACTGC AGCTCTGGGA GGTGTGGGCA GGGGAGGGCC 3300
GGCTGGGGAG GGCCGGCTGG GGAGGGGACA CAAAGTGAAG ATGGGGGTTG TTGGGCCTGA 3360
GCTCCTGCCC AGCCTTTTCC GCCGGGGTTC CTGGGTGGAT TCAAGCCTCT TGGGGGAGAC 3420
GCGCTGCAGG GCTGGAGGAT GGGCTTTGGG CCCTGAGGCT CAGGGAGGAG CACATGGATG 3480
GGACTCACCT TCTCCCTTGC CCCCAGGACT GGGTCATCGC TCCCCAAGGC TACTCGGCCT 3540 ATTACTGTGA GGGGGAGTGC TCCTTCCCAC TGGACTCCTG CATGAATGCC ACCAACCACG 3600
CCATCCTGCA GTCCCTGGTC GATACCGTCG CCCATCCTGC CCAGCCCCCT GGTGGAGGCC 3660 CTGCAGAGAG GGGTCTGGTC CAGCCAGCCG GGAGGCAGTG AGGCCACCTG CTCCATGTCT 3720
CGGGGCTTTG TCTGCACAGA GTCAGTAACG TCGCTAACTT CCCACAGCTC TGCAGGAACT 3780 GGTCCTCATA CAGCCACACT ACTACACATA GACCCACACC CAAACACGGA CACACGTGAA 3840
CAGTCGCGTA TCATGCCTGT TCTATGCACT GAACAAACTC CTGTGGGACA CTTACACACC 3900
TGCGTGCGGC GCTCAGAGGC ACAGCACATG AAACAGATGT GTACACTGTG TGGGGGCTGT 3960
GTGATCTTAA CACACGGGCC CCCGAGTACG CTGGCAAGTC TGACCGCCCG TGATATGTGC 4020
GCACAGTGTG TGGGGTGTGC GTGTGCATCA CCCACCTGTG CCGCACCACA GGTAGGAAGC 4080 TTCTAGATGG TGTGGCTCTC AACCTTTTGG CTTTTTCCCG CAGTTTCTCT CTTGGCTGTC 4140
TGTGTTTTCT CTGGATCCCC TGGCTTTTGA TGCCGTTGGT GTCTGGGGCA ACCTTAAAGG 4200
ACAAAAGCAG GCTTCTGATG GGATCACTGG TGCTGCTCAC CACTGAGTGC TCGTGTGTTT 4260
GCGGATTCTG GCACCGAGGC TTCCTTCTAG AAGTTTTTAC CTAGAATCCC AGTTCCTGGT 4320
ATTGCACAGC CTTATGTTTT CCTCTTAGGA GGTTCAACGG TGATGCCTTG ATCAGGCGCA 4380 GTGGCTCACC CTGTAATCGC AGCACACGAG CCCAGAAGTT CAAGACAAGC CTGAGCAACA 4440
CAGCAAAACC CTGTCTCTAA AATAAAAATT AAAACACACA CACACACACA CACACACACA 4500
CACACACGTG CGCACACAAT GCCTTGGTGT GAGAGGAAAG AAATTACCAA AAGCTGCTCT 4560
GAGCCTATGA TAATACTTCC TTTCTGGGCA GTCAAATGGT GTTTGCTGGA CACCCTGGAG 4620
CCATCTCCTT GGAAAGGCCC AGGGGTGATG AGGAGCTCCG TCGGGGTGGC CTGGCCAGCA 4680 CCTTTATGCC GTGTGGTTCT CACAGCTGCA TGTGTGGGAG GTACATGGGA AGGTGACTGC 4740
ACCTGCGCTC CTGGACTCCA TCTCCTCTGC CCTTGCCCCT GCCCCTCACG TGCAACTAGA 4800
GTGAGTGCTC ACAGCCTACA GGGCAGCAAA CAGGCACTGT GCTCTAGGGG AGGCTGTCGG 4860
TGGGCACAGA AGCAAACCAA CCGTGGAGTT GACACCTCCT GTGAGGAAGA GCAGACGAGC 4920
CGTGCCGTCA GTGGAGTGAG ACTGGGCCCA GCTCTCCACA CAAGGAGGGG CACGTCAGCA 4980 GCTGGAGGAG GAATGTTCCA GAAGGAGCAA GTGCAAGGCC CTAAGACAGG AGCAGGCTGG 5040
CCCTAAGTTC AGGGCAGGGG AGGAGAGGGG CTGGGTGCAG TGAAGGGGAG GAGAGTGGAG 5100 GGAGGTGATC CGGGGTGATA GGCCAGCTCC CGTAGCCTGG GTTCCCTGGG AAGAGGGTGG 5160
ATTTTATTCC AAGCAACCCC AGAGGCTGTC AGAGGTCTTC AGCAAAGAGT GTCCTTGGTC 5220 TGCGTCACCC TCCAGAAGGA CCTTTCTGGC TTGGGGAGGT CGCGGGAGTG GAAGGCAGAG 5280
GAGCAGGGGA TGAGTGAGGG CTGCTGTGGT CACCTGGCAG GTGATGGCAG CTCGACTGGG 5340
CAGGTGGTCC GAGGCAGCAC GGAGGTGGAG GTTGAGCCAG GGGCTGCTCT CAGGGAAGGG 5400
AGGAGGCGAA AGGAGTCATC CAGGAGGCCT CCCAGGCGGG AGCTATGATG TCAGGGCGGG 5460
AGGAATTCTA TGTTCCACTG AGGCCTCATT AGACCCCCAA GTGCAGAAGT GGGAAGGGGA 5520 GCAGGATCCG CAAGTCTGGA GTTCAGAAGA GAGGTCCAAG CTGAGCCAGG GGAGTGGAGA 5580
GGTGCGGGCC AATGCAGGGC CTTGAAGTGC TGAGGGCGGA TCGAGTCCTC TGGGAGAAGG 5640
AGCAGCACAG GAGAGGGGGC GAGGCTGGCT CCCAGAGCCT GGGGAGGGAG GCAGGTGTGG 5700
GGAGGCAGAG CTTGGGGGGG TCTGAAGGGC TATAAGAAGA CAGTGGTCCT TCCAGGTTCC 5760
CCCTTGGACC TCACTAAGGG CACAAACCTG GCCATGAGGT TCTCCTTCCC ATTATCCCCA 5820 GGAGGAAGTC TGAGCCCTTG GCCTGGGACT CGAGGCCCCT CATTAGTGCC CTGCCCACCT 5880
GCCCCACACC CTGGGGCTGC CATGTATCCC TCCCTGGGCA CTGTGGGCAC CACAGCTCCC 5940
GCTCCCAGAG CTCTCAGGGC TGCTCTTATT CCTGTTAATA ATTCTTATTA TTGTGCTGCT 6000
CCCATGTGGC TTGGAGATGG CCAGGGCAGG GAGCAGGTGG AGCTGGGGCG GGCTAGGTGG 6060
GTCCTCAGAG GAGGCCACTG GCTCATGCCC CTGCCTGTGC TCCCTTCCTG GCCAGGTGCA 6120 CCTGATGAAG CCAAACGCAG TCCCCAAGGC GTGCTGTGCA CCCACCAAGC TGAGCGCCAC 6180
CTCTGTGCTC TACTATGACA GCAGCAACAA CGTCATCCTG CGCAAGCACC GCAACATGGT 6240
GGTCAAGGCC TGCGGCTGCC ACTGAGTCAG CCCGCCCAGC CCTACTGCAG CCACCCTTCT 6300
CATCTGGATC GGGCCCTGCA GAGGCAGAAA ACCCTTAAAT GCTGTCACAG CTCAAGCAGG 6360
AGTGTCAGGG GCCCTCACTC TCTGTGCCTA CTTCCTGTCA GGCTTCTGGT CCTTTCTC 6418 (2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 97 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(ix) FEATURE:
(A) NAME/KEY: Protein
(B) LOCATION: 1..97
(D) OTHER INFORMATION: /label= Generic-Seq-7
/note= "wherein each Xaa is independently selected from a group of one or more specified amino acids as defined in the specification."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
Leu Xaa Xaa Xaa Phe Xaa Xaa Xaa Gly Trp Xaa Xaa Xaa Xaa Xaa Xaa 1 5 10 15
Pro Xaa Xaa Xaa Xaa Ala Xaa Tyr Cys Xaa Gly Xaa Cys Xaa Xaa Pro
20 25 30
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Asn His Ala Xaa Xaa Xaa Xaa Xaa
35 40 45
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys Cys Xaa Pro 50 55 60
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Leu Xaa Xaa Xaa Xaa Xaa Xaa Xaa 65 70 75 80
Val Xaa Leu Xaa Xaa Xaa Xaa Xaa Met Xaa Val Xaa Xaa Cys Xaa Cys
85 90 95
Xaa (2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 102 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(ix) FEATURE:
(A) NAME/KEY: Protein
(B) LOCATION: 1..102
(D) OTHER INFORMATION: /label= Generic-Seq-8
/note= "wherin each Xaa is independently selected from a group of one or more specified amino acids as defined in the specification."
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:
Cys Xaa Xaa Xaa Xaa Leu Xaa Xaa Xaa Phe Xaa Xaa Xaa Gly Trp Xaa 1 5 10 15
Xaa Xaa Xaa Xaa Xaa Pro Xaa Xaa Xaa Xaa Ala Xaa Tyr Cys Xaa Gly
20 25 30
Xaa Cys Xaa Xaa Pro Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Asn His Ala
35 40 45
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 50 55 60
Xaa Cys Cys Xaa Pro Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Leu Xaa Xaa 65 70 75 80
Xaa Xaa Xaa Xaa Xaa Val Xaa Leu Xaa Xaa Xaa Xaa Xaa Met Xaa Val
85 90 95
Xaa Xaa Cys Xaa Cys Xaa
100

Claims

What is claimed is:
1. A substantially pure protein comprising the
amino acid sequence defined by residues 303 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
2. The protein of claim 1 wherein said amino acid sequence is defined by residues 298 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
3. The protein of claim 2 wherein said amino acid sequence is defined by residues 264 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
4. The protein of claim 3 wherein said amino acid sequence is defined by residues 261 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
5. The protein of claim 4 wherein said amino acid sequence is defined by residues 18 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
6. The protein of claim 5 wherein said amino acid sequence is defined by residues 1 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
7. A substantially pure antibody which binds to an epitope on a protein encoded by a nucleic acid comprising the DNA sequence defined by bases 69- 1265 of Seq. ID No. 1.
8. A substantially pure nucleic acid comprising
part or all of the DNA sequence defined by bases 1 to 1674 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
9. A substantially pure nucleic acid encoding a
morphogenically active protein, said nucleic acid comprising a DNA sequence that hybridizes to part or all of the DNA sequence defined by bases 120 to 848 of Seq. ID No. 1, under
stringent conditions, including allelic, species and other amino acid sequence variants thereof.
10. A substantially pure nucleic acid comprising
part or all of the DNA sequence defined by bases 120 to 848 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
11. A substantially pure nucleic acid encoding a
morphogenic protein, said nucleic acid
comprising a DNA sequence defined by bases 975 to 1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
12. The nucleic acid of claim 11 wherein said DNA sequence is defined by bases 960 to 1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
13. The nucleic acid of claim 12 wherein said DNA sequence is defined by bases 858 to 1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
14. The nucleic acid of claim 13 wherein said DNA sequence is defined by bases 849 to 1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
15. The nucleic acid of claim 14 wherein said DNA sequence is defined by bases 120 to 1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
16. The nucleic acid of claim 15 wherein said DNA sequence is defined by bases 69-1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
17. A vector comprising at least part of the nucleic acid sequence defined by bases 69-1265 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof, wherein said sequence is sufficient to encode a
morphogenic protein.
18. A cell transformed with the vector of claim 17.
19. A cell adapted to express a nucleic acid
comprising the sequence defined by bases
975-1265 of Seq. ID No. 1, including allelic, species or amino acid sequence variants thereof.
20. A substantially pure morphogenic protein encoded by at least part of the nucleic acid sequence of Seq. ID No. 1 including allelic, species and other amino acid sequence variants thereof.
21. A composition for increasing the progenitor cell population in a mammal comprising:
progenitor cells, stimulated ex vivo by exposure to a morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said progenitor cells are stimulated to proliferate.
22. A composition for inducing tissue growth in a mammal comprising:
progenitor cells, stimulated by exposure to a morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said progenitor cells, when disposed in vivo within a tissue locus, are capable of tissue-specific differentiation and
proliferation within said locus.
23. The composition of claim 21 or 22 wherein said morphogenically active fragment comprises amino acid residues 303 to 399 of Seq. ID No. 1 or allelic, species and other variants thereof.
24. The composition of claim 23 wherein said
morphogenically active fragment comprises amino acid residues 298 to 399 of Seq. ID No. 1 or allelic, species and other variants thereof.
25. The composition of claim 24 wherein said
morphogenically active fragment comprises amino acid residues 264 to 399 of Seq. ID No. 1 or allelic, species and other variants thereof.
26. The composition of claim 25 wherein said
morphogenically active fragment comprises amino acid residues 261 to 399 of Seq. ID No. 1 or allelic, species and other variants thereof.
27. The composition of claim 26 wherein said
morphogenically active fragment comprises amino acid residues 18 to 399 of Seq. ID No. 1 or allelic, or species and other amino acid sequence variants thereof.
28. The composition of claim 27 wherein said
morphogenically active fragment comprises amino acid residues 1 to 399 of Seq. ID No. 1 or allelic, species and other amino acid sequence variants thereof.
29. The composition of claim 21 or 22 wherein said progenitor cells are hemopoietic pluripotential stem cells.
30. The composition of claim 21 or 22 wherein said progenitor cells are of mesenchymal origin.
31. A composition for inducing the formation of
replacement tissue at a tissue locus in a mammal comprising:
a biocompatible, acellular matrix having
components specific for said tissue and capable of providing a morphogenically permissive, tissue-specific environment; and
a morphogenically active fragment of OP-3, or allelic or species variants thereof, for
inducing the developmental cascade of tissue morphogenesis at said locus when absorbed on said matrix and provided to a tissue-specific locus requiring replacement tissue.
32. A composition for inducing the formation of
replacement tissue at a tissue locus in a mammal comprising:
a biocompatible, acellular matrix capable of providing a morphogenically permissive
environment; and a morphogenically active fragment of OP-3, or allelic or species variants thereof, for inducing the developmental cascade of tissue morphogenesis at said locus when absorbed on said matrix and provided to a tissue-specific locus requiring replacement tissue.
33. The composition of claim 31 or 32 wherein said morphogenically active fragment comprises amino acid residues 303 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
34. The composition of claim 33 wherein said
morphogenically active fragment comprises amino acid residues 298 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
35. The composition of claim 34 wherein said
morphogenically active fragment comprises amino acid residues 264 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
36. The composition of claim 35 wherein said
morphogenically active fragment comprises amino acid residues 261 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
37. The composition of claim 36 wherein said
morphogenically active fragment comprises amino acid residues 18 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
38. The composition of claim 32 wherein said
morphogenically active fragment comprises amino acid residues 1 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
39. The composition of claim 31 or 32 wherein said matrix is biodegradable.
40. The composition of claim 31 or 32 wherein said matrix is derived from organ-specific tissue.
41. The composition of claim 31 or 32 wherein said matrix comprises collagen and cell attachment factors selected from the group consisting of glycosammoglycans and proteoglycans .
42. The composition of claim 31 or 32 wherein said matrix defines a structure which permits the attachment, differentiation and proliferation of migratory progenitor cells from the body of said mammal .
43. A method of increasing a population of
progenitor cells comprising the step of:
contacting progenitor cells with a
morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said progenitor cells are stimulated to proliferate.
44. The method of claim 43 further comprising the step of:
supplying said stimulated progenitor cells to a mammal to increase the progenitor cell
population in said mammal.
45. A method of inducing tissue growth in a mammal comprising the step of:
contacting progenitor cells with a
morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said progenitor cells, when provided to a tissue-specific locus in a mammal, are capable of tissue-specific differentiation and
proliferation at said locus.
46. The method of claim 43 or 45 wherein said
progenitor cells are of mesenchymal origin.
47. A method of maintaining the phenotypic
expression of differentiated cells in a mammal comprising the steps of:
contacting said differentiated cells with a morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said cells are stimulated to express their phenotype.
48. The method of claim 47 wherein said
differentiated cells are senescent or quiescent cells.
49. A method of inducing tissue growth at a tissue locus in a mammal comprising:
providing said locus with a morphogenically active fragment of OP-3, or allelic or species variants thereof, at a concentration and for a time sufficient such that said morphogenically active fragment, when provided to a
morphogenically permissive tissue-specific locus, is capable of inducing the developmental cascade of tissue morphogenesis at said locus.
50. The method of claim 49 wherein said tissue is hepatic tissue, and said tissue locus is the liver.
51. The method of claim 49 wherein said tissue is cartilage or bone tissue, and said tissue locus is osteoporotic bone.
52. The method of claim 49 wherein said OP-3, or
allelic or species variants thereof, is provided to said locus in association with a
biocompatible, acellular matrix.
53. The method of claim 52 wherein said matrix has components specific for said tissue.
54. The method of claim 52 wherein said matrix is biodegradable.
55. The method of claim 52 wherein said matrix is derived from organ-specific tissue.
56. The method of claim 52 wherein said matrix
comprises collagen and cell attachment factors specific for said tissue.
57. The method of claim 52 wherein said matrix
defines a structure which permits the
attachment, differentiation and proliferation of migratory progenitor cells from the body of said mammal .
58. The method of claims 43, 45, 47 or 49 wherein said morphogenically active fragment comprises amino acid residues 303 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
59. The method of claim 58 wherein said
morphogenically active fragment comprises amino acid residues 298 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
60. The method of claim 59 wherein said
morphogenically active fragment comprises amino acid residues 264 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
61. The method of claim 60 wherein said
morphogenically active fragment comprises amino acid residues 261 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
62. The method of claim 61 wherein said
morphogenically active fragment comprises amino acid residues 18 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
63. The method of claim 62 wherein said
morphogenically active fragment comprises amino acid residues 1 to 399 of Seq. ID No. 1,
including allelic, species and other amino acid sequence variants thereof.
64. A method of producing a morphogenically active protein comprising the steps of:
transfecting cells with a nucleic acid sequence encoding a morphogenic protein comprising the amino acid residues 303 to 399 of Seq. ID No. 1; culturing said cells in a suitable culture medium;
expressing said morphogenic protein from said nucleic acid; and
isolating and purifying said protein from said culture medium.
65. The method of claim 64 wherein said morphogenic protein comprises amino acid residues 298 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
66. The method of claim 65 wherein said morphogenic protein comprises amino acid residues 264 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
67. The method of claim 66 wherein said morphogenic protein comprises amino acid residues 261 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
68. The method of claim 67 wherein said morphogenic protein comprises amino acid residues 18 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
69. The method of claim 68 wherein said morphogenic protein comprises amino acid residues 1 to 399 of Seq. ID No. 1, including allelic, species and other amino acid sequence variants thereof.
70. The protein of claim 1, 2 or 3 wherein said
amino acid sequence variant has an amino acid substitution for the serine at position 315 or the cysteine at position 338 in Seq. ID No. 1.
71. The protein of claim 70 wherein said amino acid sequence variant has a tryptophan residue in place of the serine at position 315 in Seq. ID No. 1.
72. The protein of claim 70 wherein said cysteine residue at position 338 in Seq. ID No. 1 is substituted for an amino acid selected from the group consisting of tyrosine, histidine,
isoleucine and serine.
73. A chimeric morphogen comprising the amino acid sequence of claim 1, 2 or 3.
74. The composition of claim 22, 23, 31 or 32
wherein said morphogen amino acid sequence variant has an amino acid substitution for the serine at position 315 or the cysteine at position 338 in Seq. ID No. 1.
75. The method of claim 43, 45, 47 or 49 wherein said morphogen amino acid sequence variant has an amino acid substitution for the serine at position 315 or the cysteine at position 338 in Seq. ID No. 1.
76. A morphogen comprising an amino acid sequence defined by Generic Sequence 7 or 8 (Seq. ID Nos. 12 or 13.)
77. The protein of claims 1, 2, 3 or 4 wherein said protein comprises a dimeric protein species complexed with a peptide comprising a pro region of a member of the morphogen family, or an allelic, species or other sequence variant thereof.
78. The protein of claim 77 wherein said dimeric
protein species is noncovalently complexed with said peptide.
79. The protein of claim 77 wherein said dimeric
protein species is complexed with two said peptides .
80. The protein of claim 77 wherein said peptide
comprises at least the first 18 amino acids of a sequence defining said pro region.
81. The protein of claim 80 wherein said peptide
comprises the full length form of said pro region.
82. The protein of claim 77 wherein said peptide
comprises a nucleic acid that hybridizes under stringent conditions with a DNA defined by nucleotides 136-192 of Seq. ID No. 3 or
nucleotides 157-211 of Seq. ID No. 7.
83. the protein of claim 77 wherein said peptide comprises at least the first 18 amino acid of the pro region of OP3 (Seq. ID No. 1).
84. The protein of claim 77 wherein said complex is further stabilized by exposure to a basic amino acid, a detergent or a carrier protein.
PCT/US1993/010520 1992-11-03 1993-11-02 Op-3-induced morphogenesis WO1994010203A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP6511392A JPH08503198A (en) 1992-11-03 1993-11-02 OP-3 induced morphogenesis
AU55900/94A AU681362B2 (en) 1992-11-03 1993-11-02 OP-3-induced morphogenesis
EP94901244A EP0672064A1 (en) 1992-11-03 1993-11-02 Op-3-induced morphogenesis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US97109192A 1992-11-03 1992-11-03
US971,091 1992-11-03
US2933593A 1993-03-04 1993-03-04
US029,335 1993-03-04
US4051093A 1993-03-31 1993-03-31
US040,510 1993-03-31

Publications (2)

Publication Number Publication Date
WO1994010203A2 true WO1994010203A2 (en) 1994-05-11
WO1994010203A3 WO1994010203A3 (en) 1994-08-18

Family

ID=27363460

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/010520 WO1994010203A2 (en) 1992-11-03 1993-11-02 Op-3-induced morphogenesis

Country Status (5)

Country Link
EP (1) EP0672064A1 (en)
JP (1) JPH08503198A (en)
AU (1) AU681362B2 (en)
CA (1) CA2147598A1 (en)
WO (1) WO1994010203A2 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998051354A2 (en) * 1997-05-15 1998-11-19 Stryker Corporation Compositions for morphogen-induced osteogenesis
WO2005089829A2 (en) 2004-03-10 2005-09-29 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
EP1719531A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
US7147839B2 (en) 1998-05-29 2006-12-12 Curis, Inc. Methods for evaluating tissue morphogenesis and activity
WO2008082563A2 (en) 2006-12-21 2008-07-10 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
EP1988395A1 (en) 1997-05-30 2008-11-05 Curis, Inc. Methods for evaluating tissue morphogenesis and morphogenic activity
EP0957943B2 (en) 1997-02-07 2008-11-26 Stryker Corporation Matrix-free osteogenic devices, implants and methods of use thereof
US7498170B2 (en) * 1994-08-12 2009-03-03 The Regents Of The University Of Michigan Bone precursor cells: compositions and methods
US7524817B2 (en) 1996-05-06 2009-04-28 Stryker Corporation Therapies for chronic renal failure
WO2010093925A2 (en) 2009-02-12 2010-08-19 Stryker Corporation PERIPHERAL ADMINISTRATION OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS FOR TREATMENT OF SYSTEMIC DISORDERS AND DISEASE
WO2010093941A2 (en) 2009-02-12 2010-08-19 Stryker Corporation COMPOSITIONS AND METHODS FOR MINIMALLY-INVASIVE SYSTEMIC DELIVERY OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS
WO2010110974A1 (en) 2009-03-24 2010-09-30 Stryker Corporation Methods and compositions for tissue engineering
WO2011031856A1 (en) 2009-09-09 2011-03-17 Stryker Corporation Bmp -7 for use in treating pain induced by injuries and diseases of an articular joint
EP2298335A1 (en) 2004-05-25 2011-03-23 Stryker Corporation Use of morphogenic proteins for treating cartilage defects
WO2011035094A1 (en) 2009-09-17 2011-03-24 Stryker Corporation Buffers for controlling the ph of bone morphogenetic proteins
US7947649B2 (en) 2008-04-14 2011-05-24 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
US7956028B2 (en) 2006-12-14 2011-06-07 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
US7964561B2 (en) 2007-06-29 2011-06-21 Advanced Technologies And Regenerative Medicine, Llc Protein formulations for use at elevated temperatures
WO2011087768A1 (en) 2009-12-22 2011-07-21 Stryker Corporation Bmp-7 variants with reduced immunogenicity
US8058237B2 (en) 2007-08-07 2011-11-15 Advanced Technologies & Regenerative Medicine, LLC Stable composition of GDF-5 and method of storage
EP2540310A1 (en) 2006-05-17 2013-01-02 Stryker Corporation Methods of treating cartilage defects using a soluble morphogenic protein complex
US8372805B1 (en) 1997-03-20 2013-02-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
ITMI20112437A1 (en) * 2011-12-30 2013-07-01 Bionest Ltd USE OF CYTOKINES, GROWTH FACTORS AND ANTIBACTERIAL FACTORS AS CAVITY SUBSTRATE MATERIAL FOR THE THERAPY AND TOOTH INFLAMMATION THERAPY
WO2013098331A1 (en) * 2011-12-30 2013-07-04 Bionest Ltd. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins c3a/c4a, and chemotactic factors
US8741840B2 (en) 2008-02-13 2014-06-03 Washington University BMP-7 for use in treating neointimal hyperplasia
US8748378B2 (en) 1997-05-05 2014-06-10 Stryker Corporation Therapies for acute renal failure
US10098927B2 (en) 2011-12-30 2018-10-16 Innomed S.A. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins C3A/C4A, immunoglobulins and chemotactic factors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989009788A2 (en) * 1988-04-08 1989-10-19 Creative Biomolecules, Inc. Biosynthetic osteogenic proteins and osteogenic devices containing them
WO1990003733A1 (en) * 1988-10-11 1990-04-19 International Genetic Engineering, Inc. Osteogenic factors
WO1992015323A1 (en) * 1991-03-11 1992-09-17 Creative Biomolecules, Inc. Protein-induced morphogenesis
WO1993005751A2 (en) * 1991-08-30 1993-04-01 Creative Biomolecules, Inc. Osteogenic proteins in the treatment of bone deseases
WO1994003600A1 (en) * 1992-07-31 1994-02-17 Creative Biomolecules, Inc. Morphogenic protein soluble complex and composition thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5011961A (en) * 1990-06-13 1991-04-30 Dow Corning Limited Method of making organosilbutadiyne polymers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989009788A2 (en) * 1988-04-08 1989-10-19 Creative Biomolecules, Inc. Biosynthetic osteogenic proteins and osteogenic devices containing them
WO1990003733A1 (en) * 1988-10-11 1990-04-19 International Genetic Engineering, Inc. Osteogenic factors
WO1992015323A1 (en) * 1991-03-11 1992-09-17 Creative Biomolecules, Inc. Protein-induced morphogenesis
WO1993005751A2 (en) * 1991-08-30 1993-04-01 Creative Biomolecules, Inc. Osteogenic proteins in the treatment of bone deseases
WO1994003600A1 (en) * 1992-07-31 1994-02-17 Creative Biomolecules, Inc. Morphogenic protein soluble complex and composition thereof

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7498170B2 (en) * 1994-08-12 2009-03-03 The Regents Of The University Of Michigan Bone precursor cells: compositions and methods
US8017580B2 (en) 1996-05-06 2011-09-13 Stryker Corporation Therapies for chronic renal failure
US8377878B2 (en) 1996-05-06 2013-02-19 Stryker Corporation Therapies for chronic renal failure
US7524817B2 (en) 1996-05-06 2009-04-28 Stryker Corporation Therapies for chronic renal failure
EP0957943B2 (en) 1997-02-07 2008-11-26 Stryker Corporation Matrix-free osteogenic devices, implants and methods of use thereof
EP1719532A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
EP1719531A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
US8802626B2 (en) 1997-03-20 2014-08-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
US8372805B1 (en) 1997-03-20 2013-02-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
US8354376B2 (en) 1997-03-20 2013-01-15 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
US8748378B2 (en) 1997-05-05 2014-06-10 Stryker Corporation Therapies for acute renal failure
WO1998051354A2 (en) * 1997-05-15 1998-11-19 Stryker Corporation Compositions for morphogen-induced osteogenesis
WO1998051354A3 (en) * 1997-05-15 1999-03-11 Creative Biomolecules Inc Compositions for morphogen-induced osteogenesis
EP1988395A1 (en) 1997-05-30 2008-11-05 Curis, Inc. Methods for evaluating tissue morphogenesis and morphogenic activity
EP2309261A1 (en) 1997-05-30 2011-04-13 Stryker Corporation Methods for evaluating tissue morphogenesis and morphogenic activity
US7147839B2 (en) 1998-05-29 2006-12-12 Curis, Inc. Methods for evaluating tissue morphogenesis and activity
US8372419B2 (en) 2004-03-10 2013-02-12 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
WO2005089829A2 (en) 2004-03-10 2005-09-29 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
EP2298335A1 (en) 2004-05-25 2011-03-23 Stryker Corporation Use of morphogenic proteins for treating cartilage defects
EP2540310A1 (en) 2006-05-17 2013-01-02 Stryker Corporation Methods of treating cartilage defects using a soluble morphogenic protein complex
US8435943B2 (en) 2006-12-14 2013-05-07 Advanced Technogies And Regenerative Medicine, Llc Protein stabilization formulations
US8895506B2 (en) 2006-12-14 2014-11-25 DePuy Synthes Products, LLC Protein stabilization formulations
US7956028B2 (en) 2006-12-14 2011-06-07 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
WO2008082563A2 (en) 2006-12-21 2008-07-10 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
US7964561B2 (en) 2007-06-29 2011-06-21 Advanced Technologies And Regenerative Medicine, Llc Protein formulations for use at elevated temperatures
US8058237B2 (en) 2007-08-07 2011-11-15 Advanced Technologies & Regenerative Medicine, LLC Stable composition of GDF-5 and method of storage
US8741840B2 (en) 2008-02-13 2014-06-03 Washington University BMP-7 for use in treating neointimal hyperplasia
US7947649B2 (en) 2008-04-14 2011-05-24 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
WO2010093941A2 (en) 2009-02-12 2010-08-19 Stryker Corporation COMPOSITIONS AND METHODS FOR MINIMALLY-INVASIVE SYSTEMIC DELIVERY OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS
WO2010093925A2 (en) 2009-02-12 2010-08-19 Stryker Corporation PERIPHERAL ADMINISTRATION OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS FOR TREATMENT OF SYSTEMIC DISORDERS AND DISEASE
WO2010110974A1 (en) 2009-03-24 2010-09-30 Stryker Corporation Methods and compositions for tissue engineering
WO2011031856A1 (en) 2009-09-09 2011-03-17 Stryker Corporation Bmp -7 for use in treating pain induced by injuries and diseases of an articular joint
WO2011035094A1 (en) 2009-09-17 2011-03-24 Stryker Corporation Buffers for controlling the ph of bone morphogenetic proteins
WO2011087768A1 (en) 2009-12-22 2011-07-21 Stryker Corporation Bmp-7 variants with reduced immunogenicity
ITMI20112437A1 (en) * 2011-12-30 2013-07-01 Bionest Ltd USE OF CYTOKINES, GROWTH FACTORS AND ANTIBACTERIAL FACTORS AS CAVITY SUBSTRATE MATERIAL FOR THE THERAPY AND TOOTH INFLAMMATION THERAPY
WO2013098331A1 (en) * 2011-12-30 2013-07-04 Bionest Ltd. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins c3a/c4a, and chemotactic factors
US9492512B2 (en) 2011-12-30 2016-11-15 Innomed S.A. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins C3A/C4A, and chemotactic factors
EP3173087A1 (en) * 2011-12-30 2017-05-31 Innomed S.A. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins c3a/c4a, and chemotactic factors
US10098927B2 (en) 2011-12-30 2018-10-16 Innomed S.A. Combination of growth factors, cytokines, antibacterial/antiviral factors, stem cell stimulating factors, complement proteins C3A/C4A, immunoglobulins and chemotactic factors

Also Published As

Publication number Publication date
EP0672064A1 (en) 1995-09-20
JPH08503198A (en) 1996-04-09
WO1994010203A3 (en) 1994-08-18
CA2147598A1 (en) 1994-05-11
AU681362B2 (en) 1997-08-28
AU5590094A (en) 1994-05-24

Similar Documents

Publication Publication Date Title
AU681362B2 (en) OP-3-induced morphogenesis
AU660019B2 (en) Protein-induced morphogenesis
US6333312B1 (en) Treatment to prevent loss of and/or increase bone mass in metabolic bone diseases
US6800603B2 (en) Morphogen-induced neural cell adhesion
US6153583A (en) OP-3 induced morphogenesis
AU681594B2 (en) Morphogen-induced nerve regeneration and repair
AU4599793A (en) Prosthetic devices having enhanced osteogenic properties
US6495513B1 (en) Morphogen-enhanced survival and repair of neural cells
US6949505B1 (en) Morphogen-induced dendritic growth
WO1993005751A2 (en) Osteogenic proteins in the treatment of bone deseases
US5652118A (en) Nucleic acid encoding a novel morphogenic protein, OP-3
US20030105004A1 (en) Novel morphogenic protein compositions of matter
EP0652953A1 (en) Morphogenic protein soluble complex and composition thereof
AU5162393A (en) Morphogen-induced liver regeneration
AU2862492A (en) Morphogenic protein screening method
WO1994006399A1 (en) Morphogen-induced periodontal tissue regeneration
US6211146B1 (en) 60A protein-induced morphogenesis
AU5290893A (en) Morphogen treatment of gastrointestinal ulcers
US20030224979A1 (en) Treatment to prevent loss of and/or increase bone mass in metabolic bone diseases
AU670558C (en) Osteogenic proteins in the treatment of bone diseases
AU670334B2 (en) Morphogen-induced periodontal tissue regeneration
WO1994006447A2 (en) 60a protein-induced morphogenesis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AT AU BB BG BR CA CH CZ DE DK ES FI GB HU JP KP KR LK LU MG MN MW NL NO NZ PL PT RO RU SD SE SK UA

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2147598

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1994901244

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1994901244

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1994901244

Country of ref document: EP