US20090258021A1 - Genes associated with mechanical stress, expression products therefrom, and uses thereof - Google Patents

Genes associated with mechanical stress, expression products therefrom, and uses thereof Download PDF

Info

Publication number
US20090258021A1
US20090258021A1 US11/841,827 US84182707A US2009258021A1 US 20090258021 A1 US20090258021 A1 US 20090258021A1 US 84182707 A US84182707 A US 84182707A US 2009258021 A1 US2009258021 A1 US 2009258021A1
Authority
US
United States
Prior art keywords
seq
bone
ocp
sequence
polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/841,827
Inventor
Paz Einat
Orit Segev
Rami Skaliter
Elena Feinstein
Alexander Faerman
Aviva Samach
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Quark Pharmaceuticals Inc
Original Assignee
Quark Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2001/046400 external-priority patent/WO2002046364A2/en
Application filed by Quark Biotech Inc filed Critical Quark Biotech Inc
Priority to US11/841,827 priority Critical patent/US20090258021A1/en
Assigned to QUARK PHARMACEUTICALS, INC. reassignment QUARK PHARMACEUTICALS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: QUARK BIOTECH, INC.
Publication of US20090258021A1 publication Critical patent/US20090258021A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Definitions

  • This invention relates to mechanical stress induced genes and their functional equivalents, probes therefor, tests to identify such genes, expression products of such genes, uses for such genes and expression products, e.g., in diagnosis (for instance risk determination), treatment, prevention, or control, of osteoporosis or factors or processes which lead to osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis and bone fractures; and, to diagnosis, treatment, prevention, or control methods or processes, as well as compositions therefor and methods or processes for making and using such compositions, and receptors for such expression products and methods or processes for obtaining and using such receptors.
  • Bone is composed of a collagen-rich organic matrix impregnated with mineral, largely calcium and phosphate.
  • mineral largely calcium and phosphate.
  • Peak bone mass is mainly genetically determined, though dietary factors and physical activity can have positive effects. Peak bone mass is attained at the point when skeletal growth ceases, after which time bone loss starts.
  • Osteoporosis is a progressive and chronic disease characterized by low bone mass and structural deterioration of bone tissue, leading to bone fragility and an increased susceptibility to fractures of the hip, spine, and wrist (diminishing bone strength).
  • Bone loss occurs without symptoms.
  • the Consensus Development Conference ((1993) Am. J. Med. 94:646-650) defined osteoporosis as “a systemic skeletal disease characterized by low bone mass and microarchitectural deterioration of bone tissue, with a consequent increase in bone fragility and susceptibility to fracture.”
  • osteoporosis Common types include postmenopausal osteoporosis; and senile osteoporosis, which generally occurs in later life, e.g., 70+ years. See, e.g., U.S. Pat. No. 5,691,153. Osteoporosis is estimated to affect more than 25 million people in the United States (Rosen (1997) Calcif. Tis. Int. 60:225-228); and, at least one estimate asserts that osteoporosis affects 1 in 3 women. Keen et al. (1997) Drugs Aging 11:333-337.
  • Osteoporosis is also a major health problem in virtually all societies. Eisman (1996); Wark (1996) Maturitas 23:193-207; and U.S. Pat. No. 5,834,200. There is a20-30% mortality rate related to hip fractures in elderly women (U.S. Pat. No. 5,691,153); and, such a patient with a hip fracture has a 10-15% greater chance of dying than others of the same age. Further, although men suffer fewer hip injuries than women, men are 25% more likely than women to die within one year of the injury. See Spangler et al., supra. Also, about 20% of the patients who lived independently before a hip fracture remain confined in a long-term health care facility one year later. The treatment of osteoporosis and related fractures costs over $10 billion annually.
  • Osteoporosis treatment helps stop further bone loss and fractures.
  • Common therapeutics include HRT (hormone replacement therapy), bisphosphonates, e.g., alendronate (Fosamax), estrogen and estrogen receptor modulators, progestin, calcitonin, and vitamin D. While there may be numerous factors that determine whether any particular person will develop osteoporosis, a step towards prevention, control or treatment of osteoporosis is determining whether one is at risk for osteoporosis. Genetic factors also play an important role in the pathogenesis of osteoporosis. Ralston (1997); see also Keen et al. (1997); Eisman (1996); Rosen (1997); Cole (1998); Johnston et al.
  • Bone 17(2 Suppl)19S-22S Gong et al. (1996) Am. J. Hum. Genet. 59:146-151; and Wasnich (1996) Bone 18(3 Suppl): 179S-183S.
  • Some attribute 50-60% of total bone variation (bone mineral density: “BMD”), depending upon the bone area, to genetic effects. Livshits et al. (1996) Hum. Biol. 68:540-554. However, up to 85%-90% of the variance in bone mineral density may be genetically determined.
  • Cytokines are powerful regulators of bone resorption and formation under control of estrogen/testosterone, parathyroid hormone and 1,25(OH) 2 D3. Some cytokines primarily enhance osteoclastic bone resorption e.g. IL-1 (interleukin-1), TNF (tumor necrosis factor) and IL-6 (interleukin-6); while others primarily stimulate bone formation e.g. TGF- ⁇ (transforming growth factor- ⁇ ), IGF (insulin-like growth factor) and PDGF (platelet derived growth factor).
  • IL-1 interleukin-1
  • TNF tumor necrosis factor
  • IL-6 interleukin-6
  • TGF- ⁇ transforming growth factor- ⁇
  • IGF insulin-like growth factor
  • PDGF platelet derived growth factor
  • Bone develops via a number of processes. Mesenchymal cells can differentiate directly into bone, as occurs in the flat bones of the craniofacial skeleton; this process is termed intramembranous ossification.
  • cartilage provides a template for bone morphogenesis, as occurs in the majority of human bones. The cartilage template is replaced by bone in a process known as endochondral ossification. Reddi (1981) Collagen Rel. Res. 1:209-226.
  • Bone is also continuously modeled during growth and development and remodeled throughout the life of the organism in response to physical and chemical signals. Development and maintenance of cartilage and bone tissue during embryogenesis and throughout the lifetime of vertebrates is very complex.
  • Osteopenia has been defined as the appearance of decreased bone mineral content on radiography, but the term more appropriately refers to a phase in the continuum from decreased bone mass to fractures and infirmity.
  • Mechanotransduction in bone tissue involves several steps: 1) mechanochemical transduction of the signal; 2) cell-to-cell signaling; and 3) increased number and activity of osteoblasts.
  • Cell-to-cell signaling after mechanical stimulus involves prostaglandins, especially those produced by COX-2, and nitric oxide. Prostaglandins induce new bone formation by promoting both proliferation and differentiation of osteoprogenitor cells.
  • the present invention provides human mechanical stress induced genes and their functional equivalents, expression products of such genes, uses for such genes and expression products for treatment, prevention, control, of osteoporosis or factors or processes which are involved in bone diseases including, but not limited to, osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis and bone fracture.
  • the invention further provides diagnostic, treatment, prevention, control methods or processes as well as compositions.
  • the invention additionally provides an isolated nucleic acid molecule, and the complement thereof, encoding the protein 608 or a functional portion thereof or a polypeptide, which is at least substantially homologous thereto.
  • the invention encompasses an isolated nucleic acid molecule encoding human protein 608 (or “OCP”) or a functional portion thereof.
  • the invention further encompasses a method for preventing, treating or controlling osteoporosis or low bone density or other factors associated with, causing or contributing to bone diseases including, but not limited to, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis or symptoms thereof, or other conditions involving mechanical stress or a lack thereof, by administering to a subject in need thereof, a polypeptide or portion thereof provided herein; and accordingly, the invention comprehends uses of polypeptides in preparing a medicament or therapy for such prevention, treatment or control.
  • the invention also comprehends a method for preventing, treating or controlling osteoporosis or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or a lack thereof, by administering a composition comprising a gene or functional portion thereof, the expression product of that gene or a functional portion thereof, an antibody or portion thereof elicited by such an expression product or portion thereof, and, the invention thus further comprehends uses of such genes, expression products, antibodies, portions thereof, in the preparation of a medicament or therapy for such control, prevention or treatment.
  • the invention further encompasses methods of use of Adlican and a novel polypeptide Adlican-2 as described herein for any use of OCP.
  • the Adlican gene, or Adlican-2 gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene.
  • the invention further encompasses compositions comprising a physiologically acceptable excipient and at least one of Adlican, the Adlican gene and antibodies specific to Adlican, and at least one of Adlican-2, the Adlican-2 gene and antibodies specific to Adlican-2.
  • the invention additionally provides receptors for expression products of human mechanical stress induced genes and their functional equivalents, such as OCP and Adlican, and methods or processes for obtaining and using such receptors.
  • the invention also provides methods of using such receptors in assays, for instance for identifying proteins or polypeptides that bind to, associate with or block the receptors, and for testing the effects of such polypeptides.
  • FIG. 1 shows the rat 608 cDNA sequence (SEQ ID NO:1).
  • FIG. 2 shows the pcDNA3.1-608 construct.
  • FIG. 3 shows the OCP rat protein amino acid sequence (SEQ ID NO:2).
  • FIG. 4 shows the mouse OCP exon and intron map.
  • FIG. 5 shows the OCP map of exon-intron borders.
  • FIG. 6 shows the human OCP exon and intron list.
  • FIG. 7 shows the OCP human cDNA sequence (predicted coding region) (nucleotides 1-7796 of SEQ ID NO:6).
  • FIGS. 8A-8D show the percent identity between FIG. 8A . rat protein/human protein; FIG. 8B . rat protein/mouse protein; FIG. 8C . rat cDNA/human cDNA; and FIG. 8D . rat cDNA/mouse cDNA, based on the OCP human cDNA sequence of FIG. 7
  • FIG. 9 shows the partial mouse OCP protein amino acid sequence (236 aa) (SEQ ID NO:15).
  • FIG. 10 shows the OCP human protein amino acid sequence (2587 aa) (SEQ ID NO:16), based on the OCP human cDNA sequence of FIG. 7 .
  • FIGS. 11A-11B show a list of expression patterns of OCP in primary cells and various other cell lines.
  • A Northern blot of poly A+ RNA RT-PCR from rat primary calvaria cells and MC3T3 cells is shown. The main 8.9 kb transcript is present only in calvaria cells.
  • RT-PCR assays with specific OCP primers were performed on total RNA from various lines as indicated on the right side of the figure. In all assays similar amounts of GapDH RT-PCR products were detected in all RNA samples.
  • B no GapDH products were detected in any RNA samples, when RT was omitted.
  • ( ⁇ ) represents no expression of OCP, while (+) represents expression. When ( ⁇ +) are indicated, the expression of OCP is induced only upon specific conditions.
  • FIG. 12 shows responsiveness of CMF608 expression to mechanical stimulation by Northern blot analysis using polyA RNA from primary rat calvaria cells before and after mechanical stress (m.s.)—see left of Figure.
  • CMF608 is transcribed as a single RNA species of approximately 9 Kb.
  • tissue blot CMF608-specific 9 Kb mRNA transcript was hardly detectable in any other tissue type except for the bone (B)—see right of Figure.
  • FIG. 13 shows that OCP is an early marker of endochondral ossification in P7 rat femoral epiphysis.
  • FIG. 14 shows that OCP is induced during osteoblastic differentiation of bone marrow stroma cells and is a specific marker of early osteoblastic differentiation in bone marrow.
  • FIG. 15 shows in vivo regulation of OCP expression in bone marrow formation by various treatments.
  • the results shown are representative of three experiments using total cellular RNA from treated two-month old mice. The different treatments are indicated.
  • the RT-PCR products are marked. Control mice did not undergo any treatment.
  • the left lane represents negative control without the addition of RT
  • the central lane represents the OCP RT-PCR product
  • the right lane represents the GapDH RT-PCR product.
  • Bone formation is shown with blood loss and estrogen administration; bone loss is shown with sciatic neurotomy models.
  • FIG. 16 shows a low power photomicrograph of fractured bone one week after the operation. Note that well-developed woven bone and fibrocartilagenous callus formed at the fracture site. Bone marrow tissue was mainly destroyed by insertion of the wire used for the fracture immobilization. Marked areas are presented at higher magnification in the following figures.
  • FIGS. 17A-17B show photomicrographs of the central part of callus, FIG. 17A . brightfield and FIG. 17B . darkfield. Cells expressing the OCP gene can be seen in the fibrous part of the callus. There was no hybridization signal from chondrocytes.
  • FIGS. 18A-18B show photomicrographs of the callus area marked by 2 in FIG. 16 , FIG. 18A . brightfield and FIG. 18B . darkfield. Cells expressing the OCP gene can be seen in a highly vascularized subperiosteal area bordering the cartilagenous part of the callus.
  • FIGS. 19A-B show photomicrographs of the highly vascularized endosteal tissue. This was developed in reaction to the wire insertion (area 3 on FIG. 16 ), FIG. 19A . brightfield and FIG. 19B . darkfield. This tissue contains many cells expressing the OCP gene.
  • FIG. 20 shows a high power photomicrograph of perivascular cells.
  • the perivascular cells express the 608 gene within lacuna of woven bone arrowheads.
  • FIG. 21 shows a high power photomicrograph of periosteum covering the woven bone. Multiple cells display expression of the 608 gene in periosteum. Arrowheads point to two 608 expressing cells within the woven bone.
  • FIGS. 22A-22B show FIG. 22A . brightfield and FIG. 22B . darkfield photomicrographs of a section of fractured bone healed for 4 weeks. Multiple cells in periosteal tissue area of active remodeling of the cancellous bone covering the callus show a hybridization signal.
  • FIG. 23 shows the boxed area of FIG. 22 presented at higher magnification.
  • OCP-expressing cells are concentrated in vascular tissue that fills the cavities resulting from osteoclast activity (marked by asterisks).
  • FIG. 24 shows increased osteoblast differentiation in OCP-transfected ROS cells.
  • RT-PCR assays were with OCP, Cbfa1, ALP, BSP and GapDH specific primers as indicated above. The results shown are representative of two experiments using total cellular RNA from: (1) the stable OCP-expressed ROS cell line; and (2) the control ROS cell line (stable transfection with pcDNA).
  • the OCP RT-PCR product is 1020 bp
  • the Cbfa1 product is 289 bp
  • the ALP product is 226 bp
  • the BSP product is 1048 bp
  • the GapDH (control) product is 450 bp long.
  • M represents protein markers.
  • FIG. 25 shows increased osteoblast proliferation in OCP-transfected ROS cells.
  • FIG. 26 shows the sequences of the primer (SEQ ID NO:19) and QB3 (CMF608) (SEQ ID NO:20).
  • FIG. 27 shows the Adlican amino acid sequence (SEQ ID NO: 21).
  • FIG. 28 shows the Adlican DNA sequence (SEQ ID NO: 22)
  • FIG. 29 shows the predicted DNA sequence of the coding region-ORF of human OCP (SEQ ID NO: 23).
  • FIG. 30 shows the predicted amino acid sequence corresponding to the predicted coding region-ORF of human OCP (SEQ ID NO: 24).
  • FIG. 31 shows the sequence of the N-terminal 663 amino acid fragment derived from the OCP rat protein (SEQ ID NO: 25).
  • FIG. 32 shows the pCM-H-608-663-N-term construct map.
  • FIG. 33 shows the structure of the pKS H608 5′-2.4 Kb bAc#1 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3878).
  • ATCC American Type Culture Collection
  • FIG. 34 shows the physical sequence of the 5′ fragment (A) cloned into pBluescript KS to NotI (5′) and HindIII (3′) sites.
  • Fragment A is comprised of the 5′ region (2440 bp) of the complete human OCP sequence and includes, in addition, at the 5′ end, 21 nucleotides of the ⁇ -actin “Kozak” region (SEQ ID NO:26).
  • FIG. 35 shows the structure of the pKS H608 m.FRG.3.5 Kb#34 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3876)
  • FIG. 36 shows the physical sequence of the middle fragment (B) cloned into pBluescript KS to HindIII (5′) and SalI (3′) sites.
  • Fragment B is comprised of the central region (3518 bp) of the complete human OCP sequence (SEQ ID NO:27).
  • FIG. 37 shows the structure of the pM H608 3′-1.9 Kb HSTG#3.3 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3877).
  • ATCC American Type Culture Collection
  • FIG. 38 shows the physical sequence of the 3′ fragment (C) cloned into pMCS SV(A) to SalI (5′) and SpeI (3′) sites.
  • Fragment C is comprised of the 3′ region (1923 bp, not including the 3 bp stop codon) of the complete human OCP sequence and includes, at the 3′ end, 18 nucleotides coding for 6 Histidine residues (SEQ ID NO:28).
  • cloned fragment C contains a silent mutation (C>T transition) compared to the predicted sequence of human OCP ORF. This transition does not change the identity of the encoded amino acid residue.
  • FIG. 39 shows the predicted DNA sequence of Adlican-2 (SEQ ID NO:29). Bases 1555 and 5638 are presented as “g” but could be any other base.
  • FIG. 40 shows the predicted amino acid sequence of human Adlican-2 (SEQ ID NO:30).
  • FIG. 41 shows the amino acid sequence alignment of (i) human Adlican (SEQ ID NO: 21), (ii) human Adlican-2 full amino acid predicted sequence, as determined by the inventors (SEQ ID NO 30), (iii) deduced sequence (hLOC96359) of human Adlican-2 fragment of 539 amino acid residues as found in the database (residues 2036-2652 of SEQ ID NO 30), and (iv) deduced sequence (hLOC90792) of human Adlican-2-fragment of 617 amino acid residues as found in the database (residues 2114-2652 of SEQ ID NO 30).
  • FIG. 42 shows the complete physical DNA sequence of the coding region (ORF) of human OCP (SEQ ID NO: 31).
  • FIG. 43 shows the predicted amino acid sequence corresponding to the complete physical DNA sequence of the coding region (ORF) of human OCP (SEQ ID NO:32).
  • FIG. 44 shows the full rat 608 cDNA sequence (SEQ ID NO:33). This sequence is virtually identical to SEQ ID NO:1, but five unknown nucleotides (designated “n” in SEQ ID NO:1) have been identified.
  • the ORF is from position 575 to 8368.
  • FIG. 45 shows the OCP rat protein amino acid sequence corresponding to the above ORF sequence (SEQ ID NO:34). Three previously unknown amino acids have been identified, as compared to SEQ ID NO:2, where these amino acids are designated “Xaa”.
  • FIG. 46 shows that ALP, which is a biochemical serum marker of bone formation, is significantly increased in 3 month old 608 KO mice.
  • the present invention is related to the discovery of a novel gene, 608 (“OCP”), the expression of which is upregulated by mechanical stress on primary calvaria cells.
  • OCP novel gene, 608
  • Several functional features identify OCP as a most specific early marker of osteo- or chondro-progenitor cells as well as an inducer of osteoblast proliferation and differentiation.
  • RNA refers to RNA isolated from cell cultures, cultured tissues or cells or tissues isolated from organisms which are stimulated, differentiated, exposed to a chemical compound, infected with a pathogen, or otherwise stimulated.
  • translation is defined as the synthesis of protein encoded by an mRNA template.
  • stimulation of translation, transcription, stability or transportation of unknown target mRNA or stimulating element includes chemically, pathogenically, physically, or otherwise inducing or repressing an mRNA population encoded by genes derived from native tissues and/or cells under pathological and/or stress conditions.
  • stimulating the expression of an mRNA with a stress inducing element or “stressor” includes, but is not limited to, the application of an external cue, stimulus, or stimuli that stimulates or initiates translation of an mRNA stored as untranslated mRNA in the cells from the sample.
  • the stressor may cause an increase in stability of certain mRNAs, or induce the transport of specific mRNAs from the nucleus to the cytoplasm.
  • the stressor may also induce specific gene transcription.
  • stimulation can include induction and/or repression of genes under pathological and/or stress conditions.
  • the method utilizes a stimulus or stressor to identify unknown target genes regulated at the various possible levels by the stress inducing element or stressor.
  • nucleic acid molecules rat 608 and human 608 genes
  • the invention further comprehends isolated and/or purified nucleic acid molecules and isolated and/or purified polypeptides having at least about 70%, preferably at least about 75% or about 77% homology (“substantially homologous”); advantageously at least about 80% or about 83%, such as at least about 85% or about 87% homology (“significantly homologous”); for instance at least about 90% or about 93% homology (“highly homologous”); more advantageously at least about 95%, e.g., at least about 97%, about 98%, about 99% or even about 100% homology (“very highly homologous” to “100% (homologous”); or from about 84-100% homology considered “highly conserved”.
  • the invention also comprehends that these nucleic acid molecules and polypeptides can be used in the same fashion as the herein or aforementioned nucleic acid molecules and polypeptides.
  • Nucleotide sequence homology can be determined using the “Align” program of Myers and Miller, ((1988) CABIOS 4:11-17) and available at NCBI.
  • the term “homology” for instance, with respect to a nucleotide or amino acid sequence can indicate a quantitative measure of homology between two sequences.
  • the percent sequence homology can be calculated as (N ref ⁇ N dif )*100/N ref , wherein N dif is the total number of non-identical residues in the two sequences when aligned and wherein N ref is the number of residues in one of the sequences.
  • “homology” with respect to sequences can refer to the number of positions with identical nucleotides or amino acid residues divided by the number of nucleotides or amino acid residues in the shorter of the two sequences wherein alignment of the two sequences can be determined in accordance with the Wilbur and Lipman algorithm ((1983) Proc. Natl. Acad. Sci. USA 80:726), for instance, using a window size of 20 nucleotides, a word length of 4 nucleotides, and a gap penalty of 4, and computer-assisted analysis and interpretation of the sequence data including alignment can be conveniently performed using commercially available programs (e.g., IntelligeneticsTM Suite, Intelligenetics Inc., CA).
  • RNA sequences are said to be similar, or have a degree of sequence identity or homology with DNA sequences, thymidine (T) in the DNA sequence is considered equal to uracil (U) in the RNA sequence.
  • RNA sequences within the scope of the invention can be derived from DNA sequences or their complements, by substituting thymidine (T) in the DNA sequence with uracil (U).
  • amino acid sequence similarity or identity or homology can be determined, for instance, using the BlastP program (Altschul et al. Nucl. Acids Res. 25:3389-3402) and available at NCBI.
  • the following references provide algorithms for comparing the relative homology of amino acid residues of two proteins, and additionally, or alternatively, with respect to the foregoing, the teachings in these references can be used for determining percent homology. Smith et al. (1981) Adv. Appl. Math. 2:482-489; Smith et al. (1983) Nucl. Acids Res. 11:2205-2220; Devereux et al. (1984) Nucl. Acids Res. 12:387-395; Feng et al. (1987) J. Molec. Evol. 25:351-360; Higgins et al. (1989) CABIOS 5:151-153; and Thompson et al. (1994) Nucl. Acids Res. 22:4673-480.
  • inventive genes and expression products as well as genes identified by the herein disclosed methods and expression products thereof and the compositions comprising Adlican or the Adlican gene (including “functional” variations of such expression products, and truncated portions of herein defined genes such as portions of herein defined genes which encode a functional portion of an expression product) are useful in treating, preventing or controlling or diagnosing mechanical stress conditions or absence or reduced mechanical stress conditions.
  • Adlican including functional portions thereof, can be used in all methods suitable for OCP.
  • the sequence homology between Adlican and human OCP provides this novel use of the Adlican protein.
  • Adlican is provided, for instance, in AF245505.1:1.8487.
  • Adlican is named for “Adhesion protein with Leucine-rich repeats has immunoglobulin domains related to perleCAN”; and shows elevated expression in cartilage from osteoarthritis patients.
  • the Adlican gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene.
  • the invention further encompasses compositions comprising a physiologically acceptable excipient and at least one of Adlican, the Adlican gene and antibodies specific to Adlican.
  • OCP expression is related to proliferation and differentation of osteoblasts and chondrocytes.
  • the expression product of OCP, or cells or vectors expressing OCP may cause cells to selectively proliferate and differentiate and thereby increase or alter bone density. Detecting levels of OCP mRNA or expression and comparing it to “normal” non-osteopathic levels may allow one to detect subjects at risk for osteoporosis or lower levels of osteoblasts and chondrocytes.
  • the medicament or treatment can be any conventional medicament or treatment for osteoporosis.
  • the medicament or treatment can be the particular protein of the gene detected in the inventive methods, or that which inhibits that protein, e.g., binds to it.
  • the medicament or treatment can be a vector which expresses the protein of the gene detected in the inventive methods or that which inhibits expression of that gene; again, for instance, that which can bind to it and/or otherwise prevents its transcription or translation.
  • the selection of administering a protein or that which expresses it, or of administering that which inhibits the protein or the gene expression, can be done without undue experimentation, e.g., based on down-regulation or up-regulation as determined by inventive methods (e.g., in the osteoporosis model).
  • PCR comprising the methods of the invention is performed in a reaction mixture comprising an amount, typically between ⁇ 10 ng-200 ng template nucleic acid; 50-100 pmoles each oligonucleotide primer; 1-1.25 mM each deoxynucleotide triphosphate; a buffer solution appropriate for the polymerase used to catalyze the amplification reaction; and 0.5-2 Units of a polymerase, most preferably a thermostable polymerase (e.g., Taq polymerase or Tth polymerase).
  • a thermostable polymerase e.g., Taq polymerase or Tth polymerase
  • Antibodies may be used in various aspects of the invention, e.g., in detection or treatment or prevention methods. Antibodies can be monoclonal, polyclonal or recombinant for use in the immunoassays or other methods of analysis necessary for the practice of the invention.
  • antibody as used in the present invention is meant both poly- and mono-clonal complete antibodies as well as fragments thereof, such as Fab, F(ab′) 2 , and Fv, which are capable of binding the epitopic determinant. These antibody fragments retain the ability to selectively bind with its antigen or receptor and are exemplified as follows, inter alia:
  • the antibodies may be prepared against the immunogen or antigenic portion thereof for example a synthetic peptide based on the sequence, or prepared recombinantly by cloning techniques or the natural gene product and/or portions thereof may be isolated and used as the immunogen.
  • the genes are identified as set forth in the present invention and the gene product identified.
  • Immunogens can be used to produce antibodies by standard antibody production technology well known to those skilled in the art as described generally in Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.; and Borrebaeck (1992) Antibody Engineering—A Practical Guide, W.H. Freeman and Co.
  • Antibody fragments include Fab, F(ab′)2, Fv and scFv prepared by methods known to those skilled in the art. Bird et al. (1988) Science 242:423-426. Any peptide having sufficient flexibility and length can be used as an scFv linker. Usually the linker is selected to have little to no immunogenicity. Linker sequences can also provide additional functions, such as a means for attaching a drug or a solid support.
  • a host such as a rabbit or goat, is immunized with the immunogen or an immunogenic fragment thereof, generally with an adjuvant and, if necessary, coupled to a carrier; and antibodies to the immunogen are collected from the sera of the immunized animal.
  • the sera can be adsorbed against related immunogens so that no cross-reactive antibodies remain in the sera rendering the polyclonal antibody monospecific.
  • mAbs monoclonal antibodies
  • an appropriate donor generally a mouse
  • splenic antibody producing cells are isolated. These cells are fused to an immortal cell, such as a myeloma cell, to provide an immortal fused cell hybrid that secretes the antibody.
  • the cells are then cultured, in bulk, and the mAbs are harvested from the culture media for use.
  • Hybridoma cell lines provide a constant, inexpensive source of chemically identical antibodies and preparations of such antibodies can be easily standardized. Methods for producing mAbs are well known to those of ordinary skill in the art. See, e.g. U.S. Pat. No. 4,196,265.
  • mRNAs from antibody producing B lymphocytes of animals, or hybridomas are reverse-transcribed to obtain cDNAs. See generally, Huston et al. (1991) Met. Enzymol. 203:46-88; Johnson and Bird (1991) Met. Enzymol. 203:88-99; and Mernaugh and Memaugh (1995) In, Molecular Methods in Plant Pathology (Singh and Singh eds.) CRC Press Inc. Boca Raton, Fla., pp. 359-365).
  • Antibody cDNA which can be full or partial length, is amplified and cloned into a phage or a plasmid.
  • the cDNA can be a partial length of heavy and light chain cDNA, separated or connected by a linker.
  • the antibody, or antibody fragment is expressed using a suitable expression system to obtain recombinant antibody.
  • Antibody cDNA can also be obtained by screening pertinent expression libraries.
  • Antibodies can be bound to a solid support substrate or conjugated with a detectable moiety or be both bound and conjugated as is well known in the art.
  • conjugation of fluorescent or enzymatic moieties see, Johnston and Thorpe (1982) Immunochemistry in Practice, Blackwell Scientific Publications, Oxford.
  • the binding of antibodies to a solid support substrate is also well known in the art. See for a general discussion, Harlow and Lane (1988); and Borrebaeck (1992).
  • the detectable moieties contemplated with the present invention include, but are not limited to, fluorescent, metallic, enzymatic and radioactive markers such as biotin, gold, ferritin, alkaline phosphatase, ⁇ -galactosidase, peroxidase, urease, fluorescein, rhodamine, tritium, 13 C and iodination.
  • fluorescent, metallic, enzymatic and radioactive markers such as biotin, gold, ferritin, alkaline phosphatase, ⁇ -galactosidase, peroxidase, urease, fluorescein, rhodamine, tritium, 13 C and iodination.
  • Antibodies can also be used as an active agent in a therapeutic composition and such antibodies can be humanized, for instance, to enhance their effects. See, Huls et al. Nature Biotech. 17:1999. “Humanized” antibodies are antibodies in which at least part of the sequence has been altered from its initial form to render it more like human immunoglobulins. In one version, the H chain and L chain C regions are replaced with human sequence. In another version, the CDR regions comprise amino acid sequences from the antibody of interest, while the V framework regions have also been converted human sequences. See, for example, EP 0329400. In a third version, V regions are humanized by designing consensus sequences of human and mouse V regions, and converting residues outside the CDRs that are different between the consensus sequences. The invention encompasses humanized mAbs.
  • the expression product from the gene or portions thereof can be useful for generating antibodies such as monoclonal or polyclonal antibodies which are useful for diagnostic purposes or to block activity of expression products or portions thereof or of genes or a portion thereof, e.g., as therapeutics.
  • antibodies to the mouse or rat 608 polypeptide may also bind the human 608 polypeptide.
  • a preferred set of antibodies encompassed by this invention are antibodies which bind human 608 polypeptide but which do not bind rat 608 polypeptide.
  • Another preferred set of antibodies encompassed by this invention are antibodies which bind human 608 polypeptide but which do not bind mouse 608 polypeptide
  • genes of the present invention or portions thereof e.g., a portion thereof which expresses a protein which function the same as or analogously to the full length protein, or genes identified by the methods herein can be expressed recombinantly, e.g., in Escherichia coli or in another vector or plasmid for either in vivo expression or in vitro expression.
  • the methods for making and/or administering a vector or recombinant or plasmid for expression of gene products of genes of the invention or identified by the invention or a portion thereof either in vivo or in vitro can be any desired method, e.g., a method which is by or analogous to the methods disclosed in: U.S. Pat. Nos.
  • the expression product generated by vectors or recombinants can also be isolated and/or purified from infected or transfected cells; for instance, to prepare compositions for administration to patients. However, in certain instances, it may be advantageous to not isolate and/or purify an expression product from a cell; for instance, when the cell or portions thereof enhance the effect of the polypeptide.
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and may be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of the treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • An inventive vector or recombinant expressing a gene or a portion thereof identified herein or from a method herein can be administered in any suitable amount to achieve expression at a suitable dosage level, e.g., a dosage level analogous to the herein mentioned dosage levels (wherein the gene product is directly present).
  • the inventive vector or recombinant nucleotide can be administered to a patient or infected or transfected into cells in an amount of about at least 10 3 pfu; more preferably about 10 4 pfu to about 10 10 pfu, e.g., about 10 5 pfu to about 10 9 pfu, for instance about 10 6 pfu to about 10 8 pfu.
  • the dosage should be a sufficient amount of plasmid to elicit a response analogous to compositions wherein gene product or a portion thereof is directly present; or to have expression analogous to dosages in such compositions; or to have expression analogous to expression obtained in vivo by recombinant compositions.
  • suitable quantities of plasmid DNA in plasmid compositions can be 1 ⁇ g to 100 mg, preferably 0.1 to 10 mg, e.g., 500 ⁇ g, but lower levels such as 0.1 to 2 mg or preferably 1-10 ⁇ g may be employed.
  • Documents cited herein regarding DNA plasmid vectors can be consulted for the skilled artisan to ascertain other suitable dosages for DNA plasmid vector compositions of the invention, without undue experimentation.
  • compositions for administering vectors can be as in or analogous to such compositions in documents cited herein or as in or analogous to compositions herein described, e.g., pharmaceutical or therapeutic compositions and the like.
  • Gene therapy can refer to the transfer of genetic material (e.g. DNA or RNA) of interest into a host subject or patient to treat or prevent a genetic or acquired disease, condition or phenotype.
  • the particular gene that is to be used or which has been identified as the target gene is identified as set forth herein.
  • the genetic material of interest encodes a product (e.g. a protein, polypeptide, peptide or functional RNA) the production in vivo of which is desired.
  • the genetic material of interest can encode a hormone, receptor, enzyme, polypeptide or peptide of therapeutic value.
  • ex vivo gene therapy Two basic approaches to gene therapy have evolved: (1) ex vivo; and (2) in vivo gene therapy.
  • ex vivo gene therapy cells are removed from a patient, and while being cultured are treated in vitro.
  • a functional replacement gene is introduced into the cell via an appropriate gene delivery vehicle/method (transfection, homologous recombination, etc.) and, an expression system as needed and then the modified cells are expanded in culture and returned to the host/patient.
  • These genetically reimplanted cells have been shown to produce the transfected gene product in situ.
  • target cells are not removed from the subject; rather, the gene to be transferred is introduced into the cells of the recipient organism in situ, that is within the recipient.
  • the host gene is defective, the gene is repaired in situ.
  • Culver (1998) Antisense DNA & RNA Based Therapeutics, February, 1998, Coronado, Calif. These genetically altered cells have been shown to produce the transfected gene product in situ.
  • the gene expression vehicle is capable of delivery/transfer of heterologous nucleic acid into a host cell.
  • the expression vehicle may include elements to control targeting, expression and transcription of the nucleic acid in a cell-selective manner as is known in the art. It should be noted that often the 5′UTR and/or 3′UTR of the gene may be replaced by the 5′ UTR and/or 3′UTR of the expression vehicle. Therefore, as used herein, the expression vehicle may, as needed, not include the 5′UTR and/or 3′UTR shown in sequences herein and only include the specific amino acid coding region.
  • the expression vehicle can include a promoter for controlling transcription of the heterologous material and can be either a constitutive or inducible promoter to allow selective transcription. Enhancers that may be required to obtain necessary transcription levels can optionally be included. Enhancers are generally any non-translated DNA sequence that works contiguously with the coding sequence (in cis) to change the basal transcription level dictated by the promoter.
  • the expression vehicle can also include a selection gene as described herein.
  • Vectors can be introduced into cells or tissues by any one of a variety of known methods within the art. Such methods can be found generally described in Sambrook et al. (1989, 1992); Ausubel et al. (1989); Chang et al. (1995) Somatic Gene Therapy, CRC Press, Ann Arbor, Mich.; Vega et al. (1995) Gene Targeting, CRC Press, Ann Arbor, Mich.; Vectors: A Survey of Molecular Cloning Vectors and Their Uses, Butterworths, Boston Mass. (1988); and Gilboa et al. (1986) BioTech.
  • nucleic acids by infection offers advantages over the other listed methods. Higher efficiency can be obtained due to their infectious nature. Moreover, viruses are very specialized and typically infect and propagate in specific cell types. Thus, their natural specificity can be used to target the vectors to specific cell types in vivo or within a tissue or mixed cell culture. Viral vectors can also be modified with specific receptors or ligands to alter target specificity through receptor-mediated events.
  • Additional features can be added to the vector to ensure its safety and/or enhance its therapeutic efficacy.
  • Such features include, for example, markers that can be used to negatively select against cells infected with the recombinant virus.
  • An example of such a negative selection marker is the TK gene described above that confers sensitivity to the antibiotic gancyclovir. Negative selection is therefore a means by which infection can be controlled because it provides inducible suicide through the addition of antibiotic. Such protection ensures that if, for example, mutations arise that produce altered forms of the viral vector or recombinant sequence, cellular transformation will not occur.
  • Features that limit expression to particular cell types can also be included. Such features include, for example, promoter and regulatory elements that are specific for the desired cell type.
  • recombinant viral vectors are useful for in vivo expression of a desired nucleic acid because they offer advantages such as lateral infection and targeting specificity.
  • Lateral infection is inherent in the life cycle of, for example, retrovirus and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is that a large area becomes rapidly infected, most of which was not initially infected by the original viral particles. This is in contrast to vertical-type of infection in which the infectious agent spreads only through daughter progeny.
  • Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
  • a plasmid or DNA vector expressing the gene could be injected directly to the target tissue; alternatively a virus bearing a plasmid or DNA vector expressing the gene could be injected directly to the target tissue.
  • cells transfected with a plasmid or DNA vector expressing the gene could be injected directly to the target tissue. These transfected cells should preferably be the patient's own cells for example mesenchymal stem cells drawn from the bone marrow.
  • the pharmaceutically “effective amount” for purposes herein is thus determined by such considerations as are known in the art.
  • the amount must be effective to achieve improvement including but not limited to improved survival rate or more rapid recovery, or improvement or amelioration or elimination of symptoms and other indicators, e.g., of osteoporosis, for instance, improvement in bone density, as are selected as appropriate measures by those skilled in the art.
  • humans are treated generally longer than the mice or other experimental animals exemplified herein.
  • Human treatment has a length proportional to the length of the disease process and drug effectiveness.
  • the doses may be single doses or multiple doses over a period of several days, but single doses are preferred.
  • animal experiments e.g., rats, mice, and the like, to humans, by techniques from this disclosure and the knowledge in the art, without undue experimentation.
  • the present invention provides an isolated nucleic acid molecule containing nucleotides having a sequence set forth in at least one of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No.
  • PTA-3638 supplements thereof and a polynucleotide having a sequence that differs from SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No.
  • PTA-3638 due to the degeneracy of the genetic code or a sequence which hybridizes under stringent conditions to a sequence in a plasmid designated pCm-H608-663-N-term or a functional portion thereof or a polynucleotide which is at least substantially homologous thereto.
  • the nucleic acid molecule comprises a polynucleotide having at least 15 nucleotides from SEQ ID NO: 1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No. PTA-3638, preferably at least 50 nucleotides and more preferably at least 100 nucleotides.
  • the present invention further provides an isolated nucleic acid molecule containing nucleotides having a sequence set forth in at least one of SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO: 28, or SEQ ID NO:26 and SEQ ID NO:27 or SEQ ID NO:26 and SEQ ID NO:27 and SEQ ID NO: 28 or as inserted in a plasmid designated pKS H608 5′-2.4 Kb bAc#1 (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va.
  • ATCC American Type Culture Collection
  • the nucleic acid molecule comprises a polynucleotide having at least 15 nucleotides from SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO: 28, or SEQ ID NO:26 and SEQ ID NO:27 or SEQ ID NO:26 and SEQ ID NO:27 and SEQ ID NO: 28 or as inserted in a plasmid designated pKS H608 5′-2.4 Kb bAc#1, pKS H608 m.FRG.3.5 Kb#34 or pM H608 3′-1.9 Kb HSTG#3.3, preferably at least 50 nucleotides and more preferably at least 100 nucleotides.
  • the present invention also provides a composition of the isolated nucleic acid molecule, a vector comprising the isolated nucleic acid molecule, a composition containing said vector and a method for preventing, treating or controlling bone diseases including, but not limited to, osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis, bone fractures or low bone density or or other conditions involving mechanical stress or a lack thereof in a subject, comprising administering the inventive composition, or the inventive vector, and a method for preparing a polypeptide comprising expressing the isolated nucleic acid molecule or comprising expressing the polypeptide from the vector.
  • bone diseases including, but not limited to, osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis, bone fractures or low bone density or or or other conditions involving mechanical stress or a lack thereof in a subject, comprising administering the inventive composition, or the inventive vector, and a method for preparing a poly
  • the present invention further provides a method for preventing, treating or controlling osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or a lack thereof in a subject, comprising administering an isolated nucleic acid molecule or functional portion thereof or a polypeptide comprising an expression product of the gene or functional portion of the polypeptide or an antibody to the polypeptide or a functional portion of the antibody.
  • the isolated nucleic acid molecule encodes a 10 kD to 100 kD N-terminal cleavage product of the OCP protein.
  • the N-terminal cleavage product comprises of a polypeptide of about 25 kD. More preferably the N-terminal cleavage product comprises a polypeptide of about 70-80 kD, most preferably about 1-663 amino acids or about 1-741 amino acids of the OCP protein.
  • the present invention provides an isolated polypeptide encoded by the inventive polynucleotide. In one embodiment of the invention, the polypeptide is identified as human 608 protein, rat 608 protein, human Adlican-2 protein or a functional portion thereof or a polypeptide which is at least substantially homologous thereto.
  • this invention is directed to an isolated polypeptide wherein the functional portion comprises consecutive amino acids having a sequence set forth in SEQ ID NO:2, SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 30, SEQ ID NO: 32 or SEQ ID NO: 34.
  • Particular fragments of the polypeptide are about the first 663 amino acids or about the first 741 amino acids of the sequence set forth in SEQ ID NO:2, SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 30, SEQ ID NO: 32 or SEQ ID NO: 34.
  • Other particular fragments of the human 608 protein include amino acids 1-500, 501-1000, 1001-1500, 1501-2000, 2001-2500, and 2051-2623 of the sequence set forth in SEQ ID NO: 32. Further particular fragments of the human 608 protein include amino acids 250-749, 750-129, 1250-1749, 1750-2249 and 2250-2623 of the sequence set forth in SEQ ID NO: 32. Nucleic acid molecules (polynucleotides) encoding these particular fragments are also envisaged as aspects of the invention. Similar particular polypeptide fragments of the Adlican-2 protein (SEQ ID NO: 30), and similar particular polynucleotide fragments of the Adlican-2 nucleic acid (SEQ ID NO: 29) are also envisaged as aspects of the invention.
  • the present invention also provides a composition comprising one or of isolated polypeptides, an antibody specific for the polypeptide or a functional portion thereof, a composition comprising the antibody or a functional portion thereof, and a method for treating or preventing osteoporosis, or fracture healing, bone elongation, or periodontosis in a subject, comprising administering to the subject a N-terminal polypeptide having a molecular weight of between 10 kD and 100 kD, preferably about 25 kD to about 70-80 kD.
  • the present invention provides for a method of treating or preventing osteoarthritis, osteopetrosis, or osteosclerosis, comprising administering to a subject an effective amount of a chemical or a neutralizing mAbs that inhibit the activity of the N-terminal polypeptide having a molecular weight of between 10 kD and 30 kD, preferably about 25 kD.
  • the term “subject,” “patient,” “host” include, but are not limited to human, bovine, pig, mouse, rat, goat, sheep and horse.
  • compositions should be selected to be chemically inert with respect to the gene product and optional adjuvant or additive. This will present no problem to those skilled in chemical and pharmaceutical principles, or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein.
  • the present invention provides receptors of the expression products of human mechanical stress induced genes and their functional equivalents, such as OCP and Adlican, and methods or processes for obtaining and using such receptors.
  • the receptors of the present invention are those to which the expression products of mechanical stress induced genes and their functional equivalents bind or associate as determined by conventional assays, as well as in vivo. For example, binding of the polypeptides of the instant invention to receptors can be determined in vitro, using candidate receptor molecules that are associated with lipid membranes. See, e.g., Watson, J.
  • Scanning electron microscopy (“SEM”), x-ray crystallography and reactions using labelled polypeptides are examples of conventional means for determining whether polypeptides have bound or associated with a receptor molecule.
  • X-ray crystallography can provide detailed structural information to determine whether and to what extent binding or association has occurred. See, e.g., U.S. Pat. No. 6,037,117; U.S. Pat. No. 6,128,582 and U.S. Pat. No. 6,153,579.
  • crystallography including X-ray crystallography, provides three-dimensional structures that show whether a candidate polypeptide ligand can or would bind or associate with a target molecule, such as a receptor. See, e.g., WO 99/45379; U.S. Pat. Nos. 6,087,478 and 6,110,672. Such binding or association shows that the receptor molecule is the receptor for the candidate polypeptide.
  • the conventional means for obtaining the receptors include raising monoclonal antibodies (Mabs) to candidate receptors, purifying the receptors from a tissue sample by use of an affinity column, treatment with a buffer, and collection of the eluate receptor molecules.
  • Mabs monoclonal antibodies
  • Other means of isolating and purifying the receptors are conventional in the art, for instance isolation and purification by dialysis, salting out, and electrophoretic (e.g. SDS-PAGE) and chromatographic (e.g. ion-exchange and gel-filtration, in additional to affinity) techniques.
  • Sequencing of the isolated receptor involves methods known in the art, for instance directly sequencing a short N-terminal sequence of the receptor, constructing a nucleic-acid probe, isolating the receptor gene, and determining the entire amino-acid sequence of the receptor from the nucleic-acid sequence.
  • the entire receptor protein can be sequenced directly.
  • Automated Edman degradation is one conventional method used to partially or entirely sequence a receptor protein, facilitated by chemical or enzymatic cleavage.
  • Automated sequenators such as an ABI-494 Procise Sequencer (Applied Biosystems) can be used. See, generally, Stryer, Biochemistry, 50-58 (3d ed. 1988).
  • the invention provides methods for using such receptors in assays, for instance for identifying proteins or polypeptides that bind to, associate with or block the inventive receptors, determining binding constants and degree of binding, and for testing the effects of such polypeptides, for instance utilising membrane receptor preparations. See Watson (1998); Komesli-Sylviane (1998). For instance, FlashPlate® (Perkin-Elmer, Massachusetts, USA) technology can be used with the present invention to determine whether and to what degree candidate polypeptides bind to and are functional with respect to a receptor of the invention.
  • the gene and polypeptides of the invention can be employed as a diagnostic in several ways as follows:
  • DNA and/or RNA from bodily fluids or from a tissue, and preferably DNA from blood are tested. 5. Diagnosis of a disease associated with mutated 608 by PCR or RT PCR of DNA or RNA respectively. DNA and/or RNA from bodily fluids or from a tissue and preferably DNA from blood are tested
  • the sample is taken from a bodily fluid or from a tissue, preferably bone or cartilage tissue;
  • the bodily fluid is selected from the group of fluid consisting of blood, lymph fluid, ascites, serous fluid, pleural effusion, sputum, cerebrospinal fluid, lacrimal fluid, synovial fluid, saliva, stool, sperm and urine, preferably blood or urine.
  • Measurement of level of the 608 polypeptide may be determined by a method selected from the group consisting of immunohistochemistry, western blotting, ELISA, antibody microarray hybridization and targeted molecular imaging; antibodies have been described above. Such methods are well-known in the art, for example for immunohistochemistry: M. A.
  • Measurement of level of 608 polynucleotide may be determined by a method selected from: RT-PCR analysis, in-situ hybridization, polynucleotide microarray and Northern blotting.
  • Such methods are well-known in the art, for example for in-situ hybridization Andreeff & Pinkel (Editors) (1999), “Introduction to Fluorescence In Situ Hybridization: Principles and Clinical Applications”, John Wiley & Sons Inc.; and for Northern blotting Trayhurn (1996) “Northern blotting”, Proc Nutr Soc; 55(1B): 583-9 and Shifman & Stein(1995) “A reliable and sensitive method for non-radioactive Northern blot analysis of nerve growth factor mRNA from brain tissues”, Journal of Neuroscience Methods; 59: 205-208 inter alia.
  • the 608 gene expression pattern was studied by in situ hybridization on sections of bones from ovariectomized and sham-operated rats.
  • Female Wistar rats weighing 300-350g were subjected to ovariectomy under general anesthesia.
  • Control rats were operated on in the same way but ovaries were not excised as a sham operation.
  • the ectopic bone formation model was employed to study the bone development 608 gene expression pattern.
  • Rat bone marrow cells were seeded into cylinders of demineralized bone matrix prepared from rat tibiae. Cylinders were implanted subcutaneously into adult rats. After three weeks, rats were sacrificed and implants were decalcified and embedded into paraffin as described above for tibial bones.
  • the 6 ⁇ m sections were prepared and hybridized in situ. After hybridization, sections were dipped into nuclear track emulsion and exposed for three weeks at 4° C. Autoradiographs were developed, stained with hematoxylin-eosin and studied under microscopy using brightfield and darkfield illumination.
  • 608 gene expression For further assessment of cell and tissue specificity of 608 gene expression, an in situ hybridization study was performed on sections of multitissue block containing multiple samples of adult rat tissues. The 608 expression developmental pattern was studied on sagittal sections of mouse embryos of 12.5, 14.5 and 16.5 days postconception (dpc) stages.
  • the hybridization signal can be seen mainly in fibroblast-like cells found in several locations throughout the sections. Prominent accumulations of these cells can be seen in the area of periosteal modeling in metaphysis, and also in regions of active remodeling of compact bone in diaphysis: at the boundary between bone marrow and endosteal osteoblasts and in periosteum; also in close contact with osteoblasts.
  • Perivascular connective tissue filling Volkmann's canals in compact bone in diaphysis and epiphysis also contains 608-expressing cells. No hybridization was found within cancellous bone and in bone marrow. This hybridization pattern suggests that cells expressing 608 are associated with areas of remodeling of preexisting bone and are not involved in primary endochondral ossification.
  • 608 expressing cells can be seen in the perichondral fibrous ring of LaCroix. Some investigators regard this fibrous tissue as the aggregation of residual mesenchymal cells able to differentiate into both osteoblasts and chondrocytes. In this respect it is noteworthy that single cells expressing 608 can be seen in epiphyseal cartilage. These 608-expressing cells are rounded cells within the lateral segment of epiphysis (sometimes in close vicinity to the LaCroix ring) and flattened cells covering the articulate surface. Most cells in articulate cartilage and all chondrocytes on the growth plate do not show 608 expression. Ovariectomy did not alter the intensity and pattern of 608 expression in bone tissue.
  • 608 hybridization signal can be seen in some fibroblast-like cells either scattered within unmineralized connective tissue matrix or concentrated at the boundary between this tissue and osteoblasts of immature bone.
  • 608 gene expression patterns revealed by in situ hybridization in bone and cartilage indicate that its expression marks some skeletal tissue elements able to differentiate into two skeletal cell types—osteoblasts and chondrocytes. The terminal differentiation of these cells appears to be accompanied by down-regulation of 608 expression. The latter observation is supported by peculiar temporal pattern of 608 expression in primary cultures of osteogenic cells isolated from calvaria bones of rat fetuses. In these cultures, expression was revealed by in situ hybridization in the vast majority of cells after one and two weeks of incubation in vitro. Three and four week old cultures showing signs of ossification contained no 608 expressing cells. Significantly, no hybridization signal was found on sections of multitissue block hybridized to 608 probe suggesting high specificity of this gene expression for the skeletal tissue in adult organisms.
  • the hybridization signal here marks the condensed portion of sclerotomes.
  • Another area of the trunk showing a hybridization signal is comprised of a thin layer of mesenchymal cells in the anterior part of the thoracic body wall.
  • probe 608 gave no hybridization signal.
  • the 608 gene is transiently expressed by at least some mesenchymal and skeleton-forming cells. This expression is down-regulated at later stages of development. More detailed study of late embryonic and postnatal stages of development reveals the timing of appearance of cells expressing 608 in bone tissue.
  • the targeting vector used to produce OCP knockout mice included a “knock-in” of the ⁇ -galactosidase (LacZ) reporter gene into the OCP gene.
  • the LacZ gene was fused to the first exon of the OCP gene—a non-coding exon.
  • expression of LacZ is expected to depend on the OCP regulatory elements and to mark the cells expressing OCP.
  • LacZ staining was performed during embryonic development on OCP knockout mice.
  • the expression pattern revealed by this analysis reflects the activation of the OCP gene promoter, which results in expression of the knocked-in LacZ gene. This data in general supports the pattern detected by the in-situ hybridization described above.
  • dpc expression is seen in the apical ectodermal ridge (AER), in the forelimbs only. This specialized region, together with the zone of polarizing activity (ZPA), directs and coordinates the development of the limb bud.
  • AER apical ectodermal ridge
  • ZPA zone of polarizing activity
  • dpc expression in AER is maintained in the forelimbs and appears also in hindlimbs. In addition, it appears in precartilagenous condensations of the ribs, in mesenchymal tissue in the face, in mesenchymal tissue rostral to the forelimb, a region of future muscle development, and in the tip of the genital tubercle.
  • the 608 expression pattern during embryonic development is closely coupled with regions of bone and cartilage development. This expression pattern strongly suggests a role for 608 in bone metabolism.
  • OCP expression was upregulated approximately 3-fold by mechanical force. This was detected both by microarray analysis and by Northern blot analysis using poly (A)+ RNA from rat calvaria cells before and after the mechanical stress. In rat calvaria primary cells and in rat bone extract this gene was expressed as a main RNA species of approximately 8.9 kb and a minor RNA transcript of approximately 9 kb. The hybridization signal was not detected in any other rat RNA from various tissue sources, including testis, colon, intestine, kidney, stomach, thymus, lung, uterus, heart, brain, liver, eye, and lymph node.
  • the partial OCP rat cDNA clone (4007 bp long) isolated from a rat calvaria cDNA phage library was found to contain a 3356 bp open reading frame closed at the 3′ end. Comparison to public mouse databases revealed no sequence homologues.
  • a complete OCP rat cDNA clone was isolated from the rat calvaria cDNA library by a combination of 5′ RACE technique (Clontech), RT-PCR of 5′ cDNA fragments, and ligation of the latter products to the original 3′ clone.
  • the full rat cDNA clone that was generated (shown in FIG. 1-SEQ ID NO:1) was sequenced, and no mutations were found.
  • the full sequence stretch is 8883 bp long and contains an ORF (nt 575-8366) for a 2597 amino acid residue protein.
  • the cDNA does not contain a polyadenylation site, but contains a 3′ poly A stretch.
  • the 608 encodes a large protein that appears to be a part of the extra-cellular matrix.
  • the gene may be actively involved in supporting osteoblast differentiation. Another option is that it is expressed in regions were remodeling takes place. Such an hypothesis is also compatible with a role in directing osteoclast action and thus it may be a target for inhibition by small molecules.
  • osteoblast function overwhelms In normal bone formation, activation of osteoblasts leads to secretion of various factors that attract osteoclast precursors or mature osteoclasts to the sites of bone formation to initiate the process of bone resorption. In normal bone formation both functions are balanced. Imbalance to any side causes either osteitis deformans (osteoblast function overwhelms) or osteoporosis (osteoclast function overwhelms).
  • osteoblast activators mechanical force stimulation—is actually applied in the present model.
  • increased expression of several genes known to respond to mechanical stress by transcriptional upregulation were found. They include tenascin, endothelin and possibly trombospondin.
  • TNT transcription-translation
  • mice genomic Bac clones containing the mouse OCP gene promoter region and part of the coding region were identified, based on their partial homology to the 5′UTR region of the rat-608 cDNA. These clones (23-261L4 and 23-241H7 with ⁇ 200 Kb average insert length) were bought from TIGR.
  • the rat OCP cDNA sequence is homologous to the human genomic DNA sequence located on chromosome 3. Based on the homology and bioinformatic analysis ( FIG. 6 ), a putative cDNA sequence was generated. FIG. 7 . The highest similarity is evident between nt 1-1965 (1-655 a.a); 2179-2337 (727-779 a.a); and 4894-7833 (1635 a.a.-end) as presented in the table shown in FIG. 8 . On the protein level, no homologues were found in the data bank.
  • the deduced OCP protein was generated following the alignment of the rat, mouse and human cDNA sequences and the equivalent rat, mouse and human amino acid sequences, respectively.
  • the following alignments were made: (a) alignment of rat, human, and mouse OCP cDNA coding regions (rat cDNA: SEQ ID NO:7; human 5+3 corrected: SEQ ID NO:8; and mus cDNA 5: SEQ ID NO:9)
  • the deduced OCP protein( FIG. 10 ) contains the following features
  • leucine-rich repeat region (aa 28-280). This region can be divided into N-terminal and C-terminal domains of leucine-rich repeats (aa 28-61 and 219-280, respectively). Between them, there are six leucine-rich repeat outliers (aa 74-96, 98-120, 122-144, 146-168, 178-200, 202-224). Leucine rich repeats are usually found in extracellular portions of a number of proteins with diverse functions. These repeats are thought to be involved in protein-protein interactions. Each leucine-rich repeat is composed of ⁇ -sheet and ⁇ -helix. Such units form elongated non-globular structures;
  • immunoglobulin C-2 type repeats at amino acid positions 488-558, 586-652, 1635-1704, 1732-1801, 1829-1898, 1928-1997, 2025-2100, 2128-2194, 2233-2294, 2324-2392, 2419-2487, 2515-2586.
  • two Ig-like repeats are found immediately downstream of a leucine-rich region, while the remaining 10 repeats are clustered at the protein's C-terminus.
  • Immunoglobulin C-2 type repeats are involved in protein-protein interaction and are usually found in extracellular protein portions;
  • NLS nuclear localization domains
  • OCP belongs to the Ig superfamily.
  • OCP is a serine-rich protein (10.3% versus av. 6.3%), with a central nuclear prediction domain and an N-terminal extracellular prediction domain.
  • 608 RNA is bone-specific. Moreover, it seems to be specific to bone progenitors (as judged by their location in bone and involvement in normal bone modeling and remodeling processes) that do not yet express the known bone-specific markers. To further prove the relevance of 608-expressing cells to osteogenic lineage, the patterns of 608 expression in the animal model of bone fracture healing that imply the activation of bone formation processes were studied.
  • Acute inflammatory cells migrate to the region, as do polymorphonuclear leukocytes and then macrophages.
  • the cells that participate directly in fracture repair during the second phase are of mesenchymal origin and are pluripotent. These cells form collagen, cartilage and bone. Some cells are derived from the cambium layer of the periosteum and form the earliest bone. Endosteal cells also participate. However, the majority of cells directly taking part in fracture healing enter the fracture site with the granulation tissue that invades the region from surrounding vessels.
  • the invading cells produce tissue known as “callus” (made up of fibrous tissue, cartilage, and young, immature fibrous bone), rapidly enveloping the ends of the bone, with a resulting gradual increase in stability of the fracture fragments.
  • Cartilage thus formed will eventually be resorbed by a process that is indistinguishable except for its lack of organization from endochondral bone formation.
  • Bone will be formed by those cells having an adequate oxygen supply and subjected to the relevant mechanical stimuli.
  • the periosteum is regarded as a source of undifferentiated progenitors participating in callus formation at the site of bone fracture.
  • the hybridization signal disappeared slowly during further differentiation stages of fracture healing (three and four weeks) and was retained only in the vascularized connective tissue.
  • 22 displays brightfield (left) and darkfield (right) photomicrographs of a section of fractured bone healed for 4 weeks. In these later healing stages, the mature callus tissue was found to be comprised mainly by cancellous bone undergoing remodeling into compact bone, with little if any cartilage or woven bone present.
  • the volume of the vascularized periosteal tissue is decreased but multiple cells in the periosteal tissue area of active remodeling of the cancellous bone covering the callus, show hybridization signal. This tissue covers the center of the callus and is also entrapped within the bone. See FIGS. 22 and 23 .
  • the box in FIG. 22 is enlarged in FIG. 23 .
  • no hybridization signal was found in chondrocytes and osteoblasts.
  • FIGS. 17 and 23 Several OCP expressing cells are concentrated in the vascular tissue that fills the cavities resulting from osteoclast activity (marked by asterisks).
  • bacs L4 and H7 were restricted with three different enzymes: BamHI, Bgl II and SauIIIA. The resulting fragments were cloned into the BamHI site of pKS. Ligation mixes were transformed into bacteria ( E. coli -DH5 ⁇ ) and 1720 colonies were plated onto nitrocellulose filters which were screened with 32 P-labeled PCR fragment spanning the mouse-OCP-exonl. Positive colonies were isolated.
  • the 14C10 clone is longer than the OCP “Lexicon” clone by ⁇ 8 Kb at the 5′ end.
  • the PCR product was cut by BamHI and NotI and the resulting 1.4 Kb fragment was ligated to pMCSIE into BamHI/NotI sites upstream to the EGFP reporter gene.
  • the resulting clone was designated pMCSIEm608prm1.4.
  • Clone p14C10 was cut by XbaI and BamHI and the excised 4.088 Kb fragment was ligated into the BamHI and XbaI sites of pMCSIEm608prm1.4, upstream to the 1.4 Kb insert.
  • the resulting clone was designated pMCSIEm608prm5.5 and contains 5552 nucleotides of the mouse 608 promoter and UTR upstream to EGFP.
  • the insert of pMCSIEm608prm5.5 clone was completely sequenced.
  • pMCSIEm608prm14.5 contains a 14.5 Kb fragment of the mouse-OCP promoter and UTR upstream to EGFP.
  • Plasmid pMCSIEm608prm14.5 was restricted by NotI and the cohesive ends of the linearized plasmid were filled and turned into blunt ends.
  • the 14.5 Kb insert was then excised by cutting the linear plasmid by SalI.
  • the purified 14.5 Kb fragment was ligated to the XhoI and HindIII (filled in) sites of pGL3-basic upstream to the luciferase gene to create the construct designated pGL3basicm608prm14.5.
  • SEQ ID NO:18 depicts 4610 bp that have been sequenced.
  • TF binding DNA elements were analyzed for similarity upstream of human and mouse OCP ATG using the DiAlign program of Genomatix GmbH.
  • the genomic pieces used are the proprietary mouse genomic OCP and reverse complement of AC024886 92001 to 11 1090.
  • the locations of the ATG in these DNA pieces are:
  • Some of the main “master gene” binding sites are the osteoblast-/chondrocyte-specific Cbfa1 factor; the chondrocyte-specific SOX 9 factor; the myoblast-specific Myo-D and Myo-F factors; the brain- and bone-specific WT1; Egr 3 and Egr 2 factors (Egr superfamily); the vitamin D-responsive (VDR) factor; the adipocyte-specific PPAR factor; and the ubiquitous activator SP1.
  • gene 608 was tested in primary cells and in cell lines with regard to expression of various markers of osteogenic and chondrogenic lineages. The results of this analysis are summarized in the following table and showed that expression of 608 is restricted to committed early osteoprogenitor cells.
  • FIG. 14 OCP was initially discovered as being upregulated during mechanical stress in calvaria cells.
  • mechanical stimulation caused by mild (287 ⁇ g) centrifugation markedly induced OCP mRNA accumulation.
  • FIG. 15 Other osteoblastic marker genes (osteopontin, ALP (staining—not shown) and Cbfa1) were transcriptionally augmented by this procedure.
  • FIG. 15 Other osteoblastic marker genes (osteopontin, ALP (staining—not shown) and Cbfa1) were transcriptionally augmented by this procedure.
  • the RT-PCR product of a non-osteoblastic marker gene (GAP-DH) was used as a control to compare RNA levels between samples. No increased expression was noticed when the latter primers were used. No expression was detected in non-osteoblastic cells ( FIG. 14 ), suggesting that OCP expression is specifically induced in osteogenesis. Responsiveness of CMF608 expression to mechanical stimulation was confirmed by Northern blot analysis using polyA RNA from primary rat calvaria cells before and after mechanical stress ( FIG. 12 .).
  • FIG. 17 A strong fluorescence signal was observed all over the second ossification center using OCP probes.
  • FIG. 17 the hybridization signal delineates periosteal and perichondrial tissue in a way similar to that found earlier in adult bones. Surrounding mature chondrocytes displayed no signal. A very faint signal was observed using the osteocalcin probe which is a marker of mature osteoblasts.
  • OCP osteoprogenitor cells that initiate endochondral ossification during bone development.
  • Osteogenic cells are believed to derive from precursor cells present in the marrow stroma and along the bone surface.
  • Blood loss a condition that stimulates hemopoietic stem cells, activates osteoprogenitor cells in the bone marrow and initiates a systemic osteogenic response.
  • High-dose estrogen administration also increases de novo medullary bone formation possibly via stimulation of generation of osteoblasts from bone marrow osteoprogenitor cells.
  • skeletal unweighting whether due to space-flight, prolonged bed-rest, paralysis or cast immobilization leads to bone loss in humans and laboratory animal models.
  • OCP cell marker
  • Bone formation should be augmented in trabecular bone and cortical bone in osteoporotic patients.
  • the latter cells normally differentiate to mature osteoblasts embedded in the trabecular and cortical bone matrix.
  • RNA from mouse and rat bone marrow extracted total RNA from mouse and rat bone marrow immediately after obtaining it and after cultivation for up to 15 days in culture.
  • No OCP-specific RT-PCR product was detected with RNA from freshly obtained bone marrow (both in adherent and non-adherent) cells.
  • a faint signal was found after 5 days in culture, and it was further enhanced when RNA from cells grown for 15 days in culture was used.
  • ALP (alkaline phosphatase) expression an osteoblastic marker was also found to be enhanced after 15 days.
  • adherent and non-adherent cells were reseeded, and RNA extractions were prepared 5 and 15 days later.
  • RNA extracted from originally adherent cells suggesting the existence of less mature progenitors in the non-adherent population of bone marrow cells.
  • the RT-PCR product of a non-osteoblastic marker gene (GAP-DH) was used as a control to compare RNA levels between samples.
  • bone marrow progenitor cells do not express OCP, but differentiate to more committed cells that do express this gene.
  • C3H10T1/2 cells were transfected with the following constructs containing the CMV promoter:
  • 608-663 a.a Construct containing 5′ untranslated region of ⁇ -actin, the OCP coding region from ATG at position 1 to the amino acid at position 663 of FIG. 3 (SEQ ID NO:2) and 3′Flag Tag.
  • the functional portion of the mammalian OCP expressed using this construct contains the first 663 amino acids of the OCP polypeptide sequence, plus several additional amino acids of the 3′Flag tag
  • pCm-H608-663Nterm which has the 5′ untranslated region of ⁇ -actin, the human OCP coding region from which encodes polypeptide from the ATG at position 1 to the amino acid at position 663 of FIG. 30 (SEQ ID NO:24) but no Flag Tag; this construct was deposited in the ATCC on Aug. 14, 2001 under ATCC Number PTA-3638.
  • pCMV-neo as negative control. This is the empty plasmid into which the 608-663aa was cloned to create vector #1 above. It serves as negative control to show that the effects are not caused by any other part of the #1 construct but by expression of the 608-663aa.
  • a readout system is created to identify small molecules that can either activate or inactivate the OCP bone-precursor-specific promoter
  • AC024886 A DNA sequence encoding a fragment of human OCP named AC024886 is found in htgs database but not in nt. There is no genomic DNA corresponding to the rat cDNA. Alignment of AC024886 against the rat cDNA using BLAST shows two areas of long alignment (and several shorter areas):
  • AC024886 was wrongly assembled in the region upstream of position 6462 (according to the rat cDNA), it was in the incorrect orientation. Using the incorrect orientation provided incorrect coding sequence and does not yield the human OCP protein.
  • Genbank report on AC024886 was as follows:
  • mice and human predicted cDNAs were modified in order to allow frame shifts that allow a good multiple alignment of the human, mouse and rat proteins. Alignment was done using CLUSTALX and Pretty.
  • Polyclonal antibodies specific to the whole 608 putative protein are prepared by methods well-known in the art (the structure of 608 resembles that of growth factor precursors). Polyclonal antibodies are identified and the recombinant active form of 608 is prepared. The activities of the polyclonal antibodies are tested in vivo in mice. The antibodies can be used for the identification of the active form of this protein which is likely to constitute a fraction of the 608 protein.
  • the homology between the rat and human N-terminal portions of the 608 protein is especially significant within the first 250 amino acids.
  • KCKKDR (aa 242-247 and 240-245, in rat and human proteins, respectively). Stretches of basic amino acids frequently serve as protease cleavage sites. The fact that such a stretch is found on the boundary of more or less conserved sequences and the fact that it occurs within the C-terminal LRR, a generally conserved domain, suggests an underlying biological significance.
  • the 608 protein may undergo post-translational processing through the cleavage of its highly conserved N-terminal portion and this portion may be an active part of the 608 protein or possess at least part of its biological activities. Since the resulting 25 kD protein preserves the signal peptide, it would be secreted.
  • the biologically active 25 kD N-terminal cleavage product of 608 can thus be used for treatment and/or prevention of osteoporosis, fracture healing, bone elongation and periodontosis.
  • the fragment can be used for treatment and/or prevention of osteoarthritis, osteopetrosis, and osteosclerosis.
  • Adlican is a recently described protein. Crowl and Luk (2000) Arthritis Biol. Res. Adlican, a proteoglycan, was derived from placenta. The full amino acid sequence of Adlican is disclosed and identified as AF245505.1:1.8487, and is hereby incorporated by reference into this application; see FIG. 27 .
  • Adlican The structure of Adlican was analyzed using methods described herein and found to have leucine-rich repeats and immunoglobulin regions similar to those of the OCP protein.
  • Adlican in any manner described herein for the OCP protein. These functions and uses have not been disclosed previously for Adlican. They include use of Adlican, or a functional portion thereof, for preventing, treating or controlling osteoporosis, or for fracture healing, bone elongation or treatment of osteopenia, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or lack thereof in a subject.
  • Adlican gene can likewise be used for any purpose described herein for an OCP gene.
  • Compositions comprising the Adlican gene, Adlican or antibodies specific for Adlican and physiologically acceptable excipients are likewise encompassed by the invention.
  • excipients are known in the art and include saline, phosphate buffered saline and Ringer's solutions.
  • the corresponding amino acid sequence of the encoded OCP protein thus has an additional two amino acids, as shown in FIG. 30 ,(SEQ ID NO:24) where these 2 additional amino acids are underlined.
  • the 663 amino acid construct described in Example 14 was expressed in 293T cells. Western blot analysis of the medium, using antibody to the Flag tag, showed the presence of the 663 amino acid polypeptide. This polypeptide was purified from the medium, using a column of anti-Flag tag antibodies.
  • the purified 663 polypeptide showed a proliferative effect on W-20-17, a mouse bone marrow stromal cell line. This effect was reproduced with two 663-polypeptide batches in 5 independent experiments.
  • the activity of proliferation of bone marrow stromal cells demonstrated in the above experiments could be indicative of pre-osteoblastic proliferation activity induced by the 663 amino acid polypeptide.
  • the 663 polypeptide activity could be mimicking the complete 608 protein in vivo activity.
  • the 663 polypeptide activity could have a dominant negative effect, i.e. an effect that inhibits the whole 608 protein in vivo activity.
  • the 663 polypeptide could be used to induce proliferation of pre-osteoblastic stromal cells. This activity could help restore the pre-osteoblastic cell population that is known to be depleted in old-age or senile osteoporosis.
  • SEQ ID NO: 2 and SEQ ID NO: 34 depict the amino acid sequence of the rat 608 polypeptide. There is an RGD sequence at positions 729-731, and there is a putative cleavage motif subtilisin-like proprotein convertase (SPC) consensus sequence at positions 735-741.
  • SPC subtilisin-like proprotein convertase
  • the 608 protein was partially cleaved by SPC, in 293HEK cells. This putative peptide also contained the RGD sequence. Many adhesive proteins, present in extracellular matrices and in the blood, contain this tripeptide as their cell recognition site. Therefore, the 608 peptide comprising 1-741 amino acids, or a shorter fragment of the 608 protein containing the RGD sequence, may be a much more effective drug than the 663 amino acid fragment.
  • the RGD and RxxRxxR (viz. R-aa1-aa2-R-aa3-aa4-R, i.e., SPC cleavage site) sequences are present in the human 608 protein sequence but are not present in Adlican or in Adlican-2.
  • a polyclonal antibody against the rat 608 fragment comprising amino acid residues 1-312 was prepared by methods well-known in the art. This antibody was used to identify 608 peptides on Western blots. Several 608 sequences were expressed in cells derived from the transiently transfected 293T kidney cell line. The sequences were rat full length 608 polypeptide, rat 608 polypeptide fragment comprising amino acid residues 1-1634, and rat 608 polypeptide fragment comprising amino acid residues 1-663. The antibody identified a peptide of about 90 kDa in all three constructs produced. This peptide was detected by the anti-608 antibody in the conditioned medium of the cells, and not in cell extracts.
  • the 608 full length and the 608 1-1634 aa proteins produced in 293T cells were cleaved and secreted into the medium.
  • the cleaved products appeared to be of identical size.
  • the 608 1-663 aa protein was also secreted into the medium, but appeared to be slightly smaller than the cleaved full length and 1-1634 aa proteins.
  • mouse calvaria cells cultured in vitro were analyzed by western blotting with the antibody to the 608 fragment 1-312aa.
  • No 608 specific band was detected in cell extracts.
  • a band of approximately 350 kDa was detected by the anti 608 1-312aa.
  • the size of this band correlates with the protein size expected from the full length 608 protein. This analysis probably indicates that the 608 full-length protein is secreted.
  • Adlican is a recently described protein.
  • the Adlican protein has LRR (Leucine-rich-repeats) and immunoglobulin regions highly similar to those of the OCP protein.
  • LRR Leucine-rich-repeats
  • immunoglobulin regions highly similar to those of the OCP protein.
  • the overall homology found between the amino acid residues of the indicated regions in the two human proteins is as shown in Example 19.
  • the deduced Adlican protein comprises the following features:
  • LRR region (26-205 aa) This region can be divided into N-terminal and C-terminal domains of LRR (aa 26-59 and 217-276, respectively). Between them, there are six LRR (aa 55-77, 78-101, 102-125, 126-149, 150-173, 182-205).
  • Adlican is a good candidate as an inducer of osteoblast proliferation and differentiation.
  • Adlican expression causes proliferation and differentation of osteoblasts and chondrocytes
  • the expression product of Adlican, or cells or vectors expressing Adlican are monitored to determine if they cause cells to selectively proliferate and differentiate and thereby increase or alter bone density. Detecting levels of Adlican mRNA or expression and comparing it to “normal” non-osteopathic levels will allow screening amd detection of individuals who may be at risk for developing osteoporosis or lower levels of osteoblasts and chondrocytes.
  • the deduced Adlican-2 protein (Genomic location: Yq11.21) was generated following the alignment (shown in FIG. 41 ) comparing Adlican-2 predicted sequences ( FIGS. 39 and 40 ) and the equivalent human Adlican amino acid sequences ( FIG. 27 ).
  • This DNA molecule and the encoded polypeptide are novel molecules and constitute an integral part of this invention.
  • a Y chromosome BAC clone (gi 8748884) shows 93% homology to Human Adlican.
  • Two mRNA sequences 100% homologous to this BAC clone were submitted to the gene bank (gi 14719942, and gi 14719940).
  • the sequence of these clones is not based on cDNA sequences, but on human genomic data and they cover a short stretch of the nucleotide sequence in the C-terminal Ig region.
  • the sequence alignment of Adlican and Adlican-2 exists along the entire Adlican sequence with one possible exception. Alignment along aa 66-215 of Adlican may be missing from the Adlican-2 molecule. This is the area of the 6 LRR (leucine-rich repeats).
  • the invention therefore encompasses the use of Adlican-2 in any manner described herein for the OCP protein.
  • No functions or uses have been disclosed previously for Adlican-2.
  • the proposed uses include use of Adlican-2, or a functional portion thereof, for preventing, treating or controlling osteoporosis, or of fracture healing, bone elongation or treatment of osteopenia, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving maechanical stress or lack thereof in a subject.
  • Adlican-2 can be used for treatment and/or prevention of osteoarthritis, osteopetrosis, and osteosclerosis.
  • Adlican-2 gene can likewise be used for any purpose described herein for an OCP gene.
  • Compositions comprising the Adlican-2 gene, Adlican-2 or antibodies specific for Adlican-2 and physiologically acceptable excipients are likewise encompassed by the invention.
  • excipients are known in the art and include saline, phosphate buffered saline and Ringer's solutions.
  • the first section of the gene to be cloned was a small fragment corresponding to the first 663 amino acids, creating the plasmid described in Example 14 (ATCC Number PTA-3638), and giving a corrected predicted sequence.
  • FIG. 42 shows the physical DNA sequence of the coding region-ORF of human OCP (SEQ ID NO: 31) having 7872 base pairs, including the stop codon.
  • the sequence contains a silent mutation (C>T transition) at position 6729 compared to the predicted sequence of human OCP ORF. This transition does not change the identity of the encoded amino acid residue.
  • FIG. 43 shows the predicted amino acid sequence corresponding to the physical DNA sequence of the coding region-ORF of human OCP (SEQ ID NO:32), having 2623 amino acids.
  • FIGS. 34 , 36 and 38 The three plasmids harboring the 5′ fragment (A), middle fragment (B) and 3′ fragment (C) are depicted in FIGS. 34 , 36 and 38 respectively, and were deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession numbers PTA-3878, PTA-3876 and PTA-3877 respectively.
  • ATCC American Type Culture Collection
  • FIG. 34 shows the physical sequence of the 5′ fragment (A) cloned into pBluescript KS to NotI (5′) and HindIII (3′) sites.
  • Fragment A is comprised of the 5′ region (2440 bp) of the complete human OCP sequence and includes, in addition, at the 5′ end, 21 nucleotides of the ⁇ -actin “Kozak” region (nucleotides 9-29) followed by the ATG initiation coNotI (5′) and HindIII (3′) sites are located at nucleotides 1-8 and 2464-2469 respectively (SEQ ID NO:26).
  • FIG. 36 shows the physical sequence of the middle fragment (B) cloned into pBluescript KS to HindIII (5′) and SalI (3′) sites.
  • Fragment B is comprised of the central region (3518 bp) of the complete human OCP sequence; the HindIII (5′) and SalI (3′) sites are located at nucleotides 1-6 and 3513-3518 respectively (SEQ ID NO:27).
  • FIG. 38 shows the physical sequence of the 3′ fragment (C) cloned into pMCS SV(A) to SalI (5′) and SpeI (3′) sites.
  • Fragment C is comprised of the 3′ region (1923 bp, not including the 3 bp stop codon) of the complete human OCP sequence and includes at the 3′ end, 18 nucleotides coding for 6 Histidine residues, nucleotides 1924-1941, followed by the TGA stop codon.
  • the SalI (5′) and SpeI (3′) sites are located at nucleotides 1-6 and 1945-1950 respectively (SEQ ID NO:28).
  • cloned fragment C contains a silent mutation (C>T transition) at nucleotide 783 compared to the predicted sequence of human OCP ORF; this transition does not change the identity of the encoded amino acid residue.
  • the sequence of fragment C does not include the 3 nucleotide stop codon at the 3′ end, since it is interrupted by 18 nucleotides coding for 6 Histidine residues.
  • Knockout mice deleted of the 608 gene were prepared by the method of Wattler et. al. BioTechniques 26:1150-1160, 1999. Comparison of 608 knockout (KO) mice to age, sex, and treatment matched wild type (WT) mice was performed to test the effect of 608 absence on bone parameters. Bone parameters of KO and WT were compared in untreated 3 and 4 months old females. KO and WT bone parameters were also compared in 3 months old female mice 5 weeks post ovariectomy (post-menopausal osteoporosis model).
  • the bone-related phenotypes were evaluated using two analyses: Peripheral Quantitative Computed Tomography (PQCT) of femur and tibia Rosen H N et. Al. Calcif. Tissue Int. 57:35-39, 1995) and serum Alkaline phosphatase (ALP) (Farley J R et. al. J Bone Miner Res 9:497-508, 1994.
  • PQCT scanning is a 2D X-ray analysis that measures bone mineral density (BMD), bone mineral content (BMC), and bone geometric parameters.
  • Serum ALP is a biochemical marker of bone formation.
  • pQCT scanning was performed for two groups of mice. pQCT of 3 months old untreated (sham operated) female mice gave parameters that were significantly different between WT and KO by two-way ANOVA analysis (pvalue ⁇ 0.05), as shown in the Table below.
  • the bone related phenotype of untreated 608 KO females is as follows:
  • the 608KO values were higher compared to WT. This may implicate an inhibitory role for the 608 gene in bone metabolism.
  • KO mice appeared to have some basic anatomical differences compared to the bones of WT mice. This observation is based on trends seen in parameters reflecting bone geometry, such as total slice area, periosteal circumference, cortical area and thickness. Compared to untreated WT mice, an increase in the femur metaphysis area was observed in KO animals, both in 4 month-old and 3 month-old mice. Distal femur total BMD was notably unaffected by genotype despite the differences in bone size. Similar increases in geometric parameters were noted in KO mice compared to WT at the femur diaphysis (cortical area) and at the tibia diaphysis (cortical thickness) in 3 month-old mice. At 4 months old there is also a significant increase in cortical BMD of both femur and tibia.
  • the effects on bone mass and biochemical markers of bone turnover noted in the KO mice appear to be indicative of a protective effect of the KO phenotype on bone loss following ovariectomy, although the effects were small.
  • a trend to a genotype-related prevention of bone loss in the distal femur metaphysis relative to the ovariectomized controls was observed in KO animals.
  • a slight partial prevention of bone loss relative to the ovariectomized control group was observed in KO mice at the endocortical surface of femur and tibia metaphysis, although the effects were not marked.
  • the function of the 608 protein could be to promote proliferation of the undifferentiated osteoprogenitor cell population.
  • This hypothesis is based on the proliferative effect of the 608 1-663aa polypeptide on mouse bone marrow cell line, as shown in Example 21. In the absence of this protein the balance between proliferation and differentiation of osteoprogenitors is changed in favor of differentiation and therefore the increased bone parameters are obtained at a young age. It could be that in aged mice this change in balance causes a decrease in bone parameters due to the normal decrease in osteoprogenitors that occurres with aging. If this hypothesis is correct an intermittent administration of the 608 protein or fragments of it could be used as a treatment for osteoporosis. Administration of the 608 polypeptide would cause proliferation of osteoprogenitors. When 608 level is allowed to decrease to normal levels, differentiation could take place.
  • Timing of intermittent treatment may be daily e.g. daily administration, preferably by injection, preferably subcutaneous, as opposed to continuous administration e.g. by infusion.
  • Other examples may be administration every other day, or every few days, or even once a week or once a month.
  • parathyroid hormone (1-34 amino acid)
  • daily subcutaneous injections of 20-40 ⁇ g were considered intermittent administration as opposed to continuous infusions; see Neer R. M. et. al. 2001, The New England Journal of Medicine. 344: 1434-1441, Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis.
  • 663aa fragment may act as an inhibitor of 608 function, as discussed in Example 21.

Abstract

The disclosure relates to human and mechanical stress induced genes, in particular gene 608, and functional equivalents, probes therefor, tests to identify such genes, polypeptide expression products of such genes, antibodies to the polypeptides, uses for such genes, expression products and antibodies, e.g., in diagnosis (for instance risk determination), treatment, prevention, or control, of osteoporosis or fractures; and to diagnostic, treatment, prevention, or control methods or processes, as well as compositions therefor and methods or processes for making and using such compositions, and receptors therefor and methods or processes for obtaining and using such receptors.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a divisional of U.S. patent application Ser. No. 10/454,351, filed Jun. 4, 2003, which, in turn, was a continuation-in-part of International Application No. PCT/US01/46400, filed Dec. 4, 2001, the entire contents of which are hereby incorporated by reference, and also a continuation-in-part of U.S. patent application Ser. No. 09/312,216, filed May 14, 1999, the entire contents of which are also hereby incorporated by reference. Each document or reference cited in those applications is hereby expressly incorporated herein by reference. Documents or references are also cited in the following text, and these documents or references (“herein-cited documents or references”), as well as each document or reference cited in each of the herein-cited documents or references, are hereby expressly incorporated herein by reference.
  • FIELD OF THE INVENTION
  • This invention relates to mechanical stress induced genes and their functional equivalents, probes therefor, tests to identify such genes, expression products of such genes, uses for such genes and expression products, e.g., in diagnosis (for instance risk determination), treatment, prevention, or control, of osteoporosis or factors or processes which lead to osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis and bone fractures; and, to diagnosis, treatment, prevention, or control methods or processes, as well as compositions therefor and methods or processes for making and using such compositions, and receptors for such expression products and methods or processes for obtaining and using such receptors.
  • BACKGROUND OF THE INVENTION
  • Bone is composed of a collagen-rich organic matrix impregnated with mineral, largely calcium and phosphate. Two major forms of bone exist, compact cortical bone forms the external envelopes of the skeleton and trabecular or medullary bone forms plates that traverse the internal cavities of the skeleton. The responses of these two forms to metabolic influences and their susceptibility to fracture differ.
  • Bone undergoes continuous remodeling (turnover, renewal) throughout life. Mechanical and electrical forces, hormones and local regulatory factors influence remodeling. Bone is renewed by two opposing activities that are coupled in time and space. Parfitt (1979) Calcify Tis. Int. 28:1-5. These activities, resorption and formation, are contained within a temporary anatomic structure known as a bone-remodeling unit. Parfitt (1981) Res. Staff Physic. Dec.: 60-72. Within a given bone-remodeling unit, old bone is resorbed by osteoclasts. The resorbed cavity created by osteoclasts is subsequently filled with new bone by osteoblasts, synthesizing bone organic matrix.
  • Peak bone mass is mainly genetically determined, though dietary factors and physical activity can have positive effects. Peak bone mass is attained at the point when skeletal growth ceases, after which time bone loss starts.
  • In contrast to the positive balance that occurs during growth, in osteoporosis, the resorbed cavity is not completely refilled by bone. Parfitt (1988), Osteoporosis: Etiology, Diagnosis, and Management (Riggs and Melton, eds.) Raven Press, New York, pp. 74-93. Osteoporosis, or porous bone, is a progressive and chronic disease characterized by low bone mass and structural deterioration of bone tissue, leading to bone fragility and an increased susceptibility to fractures of the hip, spine, and wrist (diminishing bone strength).
  • Bone loss occurs without symptoms. The Consensus Development Conference ((1993) Am. J. Med. 94:646-650) defined osteoporosis as “a systemic skeletal disease characterized by low bone mass and microarchitectural deterioration of bone tissue, with a consequent increase in bone fragility and susceptibility to fracture.”
  • Common types of osteoporosis include postmenopausal osteoporosis; and senile osteoporosis, which generally occurs in later life, e.g., 70+ years. See, e.g., U.S. Pat. No. 5,691,153. Osteoporosis is estimated to affect more than 25 million people in the United States (Rosen (1997) Calcif. Tis. Int. 60:225-228); and, at least one estimate asserts that osteoporosis affects 1 in 3 women. Keen et al. (1997) Drugs Aging 11:333-337. Moreover, life expectancy has increased, and in the western world, 17% of women are now over 50 years of age: a woman can expect to live one third of her life after menopause. Thus, some estimate that 1 out of every 2 women and 1 out of 5 men will eventually develop osteoporosis; and, that 75 million people in the U.S., Japan and Europe have osteoporosis. The World Summit of Osteoporosis Societies estimates that more than 200 million people worldwide are afflicted with the disease. The actual incidence of the disease is difficult to estimate since the condition is often asymptomatic until a bone fracture occurs. It is believed that there are over 1.5 million osteoporosis-associated bone fractures per year in the U.S. Of these, 300,000 are hip fractures that usually require hospitalization and surgery and may result in lengthy or permanent disability or even death. See a minireview by Spangler et al. “The Genetic Component of Osteoporosis” (1997) Cambridge Scientific Abstracts”.
  • Osteoporosis is also a major health problem in virtually all societies. Eisman (1996); Wark (1996) Maturitas 23:193-207; and U.S. Pat. No. 5,834,200. There is a20-30% mortality rate related to hip fractures in elderly women (U.S. Pat. No. 5,691,153); and, such a patient with a hip fracture has a 10-15% greater chance of dying than others of the same age. Further, although men suffer fewer hip injuries than women, men are 25% more likely than women to die within one year of the injury. See Spangler et al., supra. Also, about 20% of the patients who lived independently before a hip fracture remain confined in a long-term health care facility one year later. The treatment of osteoporosis and related fractures costs over $10 billion annually.
  • Osteoporosis treatment helps stop further bone loss and fractures. Common therapeutics include HRT (hormone replacement therapy), bisphosphonates, e.g., alendronate (Fosamax), estrogen and estrogen receptor modulators, progestin, calcitonin, and vitamin D. While there may be numerous factors that determine whether any particular person will develop osteoporosis, a step towards prevention, control or treatment of osteoporosis is determining whether one is at risk for osteoporosis. Genetic factors also play an important role in the pathogenesis of osteoporosis. Ralston (1997); see also Keen et al. (1997); Eisman (1996); Rosen (1997); Cole (1998); Johnston et al. (1995) Bone 17(2 Suppl)19S-22S; Gong et al. (1996) Am. J. Hum. Genet. 59:146-151; and Wasnich (1996) Bone 18(3 Suppl): 179S-183S. Some attribute 50-60% of total bone variation (bone mineral density: “BMD”), depending upon the bone area, to genetic effects. Livshits et al. (1996) Hum. Biol. 68:540-554. However, up to 85%-90% of the variance in bone mineral density may be genetically determined.
  • Studies have shown from family histories, twin studies, and racial factors, that there may be a predisposition for osteoporosis. Jouanny et al. (1995) Arthritis Rheum. 38:61-67; Garnero et al. (1996) J. Clin. Endrocrinol. Metab. 81:140-146; Cummings (1996) Bone 18(3 Suppl): 165S-167S; and Lonzer et al. (1996) Clin. Pediatr. 35:185-189. Several candidate genes may be involved in this, most probably multigenic, process.
  • Cytokines are powerful regulators of bone resorption and formation under control of estrogen/testosterone, parathyroid hormone and 1,25(OH)2D3. Some cytokines primarily enhance osteoclastic bone resorption e.g. IL-1 (interleukin-1), TNF (tumor necrosis factor) and IL-6 (interleukin-6); while others primarily stimulate bone formation e.g. TGF-β (transforming growth factor-β), IGF (insulin-like growth factor) and PDGF (platelet derived growth factor).
  • There is need for clinical and epidemiological research for the prevention and treatment of osteoporosis for gaining greater knowledge of factors controlling bone cell activity and regulation of bone mineral and matrix formation and remodeling.
  • Bone develops via a number of processes. Mesenchymal cells can differentiate directly into bone, as occurs in the flat bones of the craniofacial skeleton; this process is termed intramembranous ossification. Alternatively, cartilage provides a template for bone morphogenesis, as occurs in the majority of human bones. The cartilage template is replaced by bone in a process known as endochondral ossification. Reddi (1981) Collagen Rel. Res. 1:209-226. Bone is also continuously modeled during growth and development and remodeled throughout the life of the organism in response to physical and chemical signals. Development and maintenance of cartilage and bone tissue during embryogenesis and throughout the lifetime of vertebrates is very complex. It is widely accepted that a multitude of factors, from systemic hormones to local regulatory factors such as the members of the TGF-β superfamily, cytokines and prostaglandins, act in concert to regulate the continuous processes of bone formation and bone resorption. Disturbance of the balance between osteoblastic bone deposition and osteoclastic bone resorption is responsible for many skeletal diseases.
  • Diseases of bone loss are a major public health problem especially for women in all Western communities. The most common cause of osteopenia is osteoporosis; other causes include osteomalacia and bone disease related to hyperparathyroidism. Osteopenia has been defined as the appearance of decreased bone mineral content on radiography, but the term more appropriately refers to a phase in the continuum from decreased bone mass to fractures and infirmity.
  • It is estimated that 30 million Americans are at risk for osteoporosis, the most common among these diseases, and there are probably 100 million people similarly at risk worldwide. Melton (1995) Bone Min. Res. 10:175. These numbers are growing as the proportion of the elderly in the world population increases. Despite recent successes with drugs that inhibit bone resorption, there is a clear need for specific anabolic agents that will considerably increase bone formation in people who have already suffered substantial bone loss. There are no such drugs currently approved.
  • Mechanical stimulation induces new bone formation in vivo and increases osteoblastic differentiation and metabolic activity in culture. Mechanotransduction in bone tissue involves several steps: 1) mechanochemical transduction of the signal; 2) cell-to-cell signaling; and 3) increased number and activity of osteoblasts. Cell-to-cell signaling after mechanical stimulus involves prostaglandins, especially those produced by COX-2, and nitric oxide. Prostaglandins induce new bone formation by promoting both proliferation and differentiation of osteoprogenitor cells.
  • OBJECTS AND SUMMARY OF THE INVENTION
  • In a search for agents that enhance osteoblast proliferation/differentiation and bone formation, mechanical force was employed as an osteogenesis inducer and a proprietary gene discovery methodology was carried out to detect genes that are specifically expressed in very early osteo-, chondro-progenitor cells.
  • The present invention provides human mechanical stress induced genes and their functional equivalents, expression products of such genes, uses for such genes and expression products for treatment, prevention, control, of osteoporosis or factors or processes which are involved in bone diseases including, but not limited to, osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis and bone fracture. The invention further provides diagnostic, treatment, prevention, control methods or processes as well as compositions.
  • The invention additionally provides an isolated nucleic acid molecule, and the complement thereof, encoding the protein 608 or a functional portion thereof or a polypeptide, which is at least substantially homologous thereto. The invention encompasses an isolated nucleic acid molecule encoding human protein 608 (or “OCP”) or a functional portion thereof.
  • The invention further encompasses a method for preventing, treating or controlling osteoporosis or low bone density or other factors associated with, causing or contributing to bone diseases including, but not limited to, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis or symptoms thereof, or other conditions involving mechanical stress or a lack thereof, by administering to a subject in need thereof, a polypeptide or portion thereof provided herein; and accordingly, the invention comprehends uses of polypeptides in preparing a medicament or therapy for such prevention, treatment or control.
  • The invention also comprehends a method for preventing, treating or controlling osteoporosis or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or a lack thereof, by administering a composition comprising a gene or functional portion thereof, the expression product of that gene or a functional portion thereof, an antibody or portion thereof elicited by such an expression product or portion thereof, and, the invention thus further comprehends uses of such genes, expression products, antibodies, portions thereof, in the preparation of a medicament or therapy for such control, prevention or treatment.
  • Analogously with the OCP-related description above, the invention further encompasses methods of use of Adlican and a novel polypeptide Adlican-2 as described herein for any use of OCP. The Adlican gene, or Adlican-2 gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene. The invention further encompasses compositions comprising a physiologically acceptable excipient and at least one of Adlican, the Adlican gene and antibodies specific to Adlican, and at least one of Adlican-2, the Adlican-2 gene and antibodies specific to Adlican-2.
  • The invention additionally provides receptors for expression products of human mechanical stress induced genes and their functional equivalents, such as OCP and Adlican, and methods or processes for obtaining and using such receptors. The invention also provides methods of using such receptors in assays, for instance for identifying proteins or polypeptides that bind to, associate with or block the receptors, and for testing the effects of such polypeptides. These and other embodiments are disclosed or are obvious from and encompassed by, the Detailed Description which follows the Brief Description of the Figures below.
  • BRIEF DESCRIPTION OF THE FIGURES
  • The following Detailed Description, given by way of example, but not intended to limit the invention to specific embodiments described, may be understood in conjunction with the accompanying Figures, in which
  • FIG. 1 shows the rat 608 cDNA sequence (SEQ ID NO:1).
  • FIG. 2 shows the pcDNA3.1-608 construct.
  • FIG. 3 shows the OCP rat protein amino acid sequence (SEQ ID NO:2).
  • FIG. 4 shows the mouse OCP exon and intron map.
  • FIG. 5 shows the OCP map of exon-intron borders.
  • FIG. 6 shows the human OCP exon and intron list.
  • FIG. 7 shows the OCP human cDNA sequence (predicted coding region) (nucleotides 1-7796 of SEQ ID NO:6).
  • FIGS. 8A-8D show the percent identity between FIG. 8A. rat protein/human protein; FIG. 8B. rat protein/mouse protein; FIG. 8C. rat cDNA/human cDNA; and FIG. 8D. rat cDNA/mouse cDNA, based on the OCP human cDNA sequence of FIG. 7
  • FIG. 9 shows the partial mouse OCP protein amino acid sequence (236 aa) (SEQ ID NO:15).
  • FIG. 10 shows the OCP human protein amino acid sequence (2587 aa) (SEQ ID NO:16), based on the OCP human cDNA sequence of FIG. 7.
  • FIGS. 11A-11B show a list of expression patterns of OCP in primary cells and various other cell lines. A. Northern blot of poly A+ RNA RT-PCR from rat primary calvaria cells and MC3T3 cells is shown. The main 8.9 kb transcript is present only in calvaria cells. RT-PCR assays with specific OCP primers were performed on total RNA from various lines as indicated on the right side of the figure. In all assays similar amounts of GapDH RT-PCR products were detected in all RNA samples. In addition, B. no GapDH products were detected in any RNA samples, when RT was omitted. (−) represents no expression of OCP, while (+) represents expression. When (−+) are indicated, the expression of OCP is induced only upon specific conditions.
  • FIG. 12 shows responsiveness of CMF608 expression to mechanical stimulation by Northern blot analysis using polyA RNA from primary rat calvaria cells before and after mechanical stress (m.s.)—see left of Figure. In these cells, CMF608 is transcribed as a single RNA species of approximately 9 Kb. On tissue blot, CMF608-specific 9 Kb mRNA transcript was hardly detectable in any other tissue type except for the bone (B)—see right of Figure.
  • FIG. 13 shows that OCP is an early marker of endochondral ossification in P7 rat femoral epiphysis.
  • FIG. 14 shows that OCP is induced during osteoblastic differentiation of bone marrow stroma cells and is a specific marker of early osteoblastic differentiation in bone marrow.
  • FIG. 15 shows in vivo regulation of OCP expression in bone marrow formation by various treatments. The results shown are representative of three experiments using total cellular RNA from treated two-month old mice. The different treatments are indicated. The RT-PCR products are marked. Control mice did not undergo any treatment. In each treatment group the left lane represents negative control without the addition of RT, the central lane represents the OCP RT-PCR product and the right lane represents the GapDH RT-PCR product. Bone formation is shown with blood loss and estrogen administration; bone loss is shown with sciatic neurotomy models.
  • FIG. 16 shows a low power photomicrograph of fractured bone one week after the operation. Note that well-developed woven bone and fibrocartilagenous callus formed at the fracture site. Bone marrow tissue was mainly destroyed by insertion of the wire used for the fracture immobilization. Marked areas are presented at higher magnification in the following figures.
  • FIGS. 17A-17B show photomicrographs of the central part of callus, FIG. 17A. brightfield and FIG. 17B. darkfield. Cells expressing the OCP gene can be seen in the fibrous part of the callus. There was no hybridization signal from chondrocytes.
  • FIGS. 18A-18B show photomicrographs of the callus area marked by 2 in FIG. 16, FIG. 18A. brightfield and FIG. 18B. darkfield. Cells expressing the OCP gene can be seen in a highly vascularized subperiosteal area bordering the cartilagenous part of the callus.
  • FIGS. 19A-B show photomicrographs of the highly vascularized endosteal tissue. This was developed in reaction to the wire insertion (area 3 on FIG. 16), FIG. 19A. brightfield and FIG. 19B. darkfield. This tissue contains many cells expressing the OCP gene.
  • FIG. 20 shows a high power photomicrograph of perivascular cells. The perivascular cells express the 608 gene within lacuna of woven bone arrowheads.
  • FIG. 21 shows a high power photomicrograph of periosteum covering the woven bone. Multiple cells display expression of the 608 gene in periosteum. Arrowheads point to two 608 expressing cells within the woven bone.
  • FIGS. 22A-22B show FIG. 22A. brightfield and FIG. 22B. darkfield photomicrographs of a section of fractured bone healed for 4 weeks. Multiple cells in periosteal tissue area of active remodeling of the cancellous bone covering the callus show a hybridization signal.
  • FIG. 23 shows the boxed area of FIG. 22 presented at higher magnification. Several OCP-expressing cells are concentrated in vascular tissue that fills the cavities resulting from osteoclast activity (marked by asterisks).
  • FIG. 24 shows increased osteoblast differentiation in OCP-transfected ROS cells. RT-PCR assays were with OCP, Cbfa1, ALP, BSP and GapDH specific primers as indicated above. The results shown are representative of two experiments using total cellular RNA from: (1) the stable OCP-expressed ROS cell line; and (2) the control ROS cell line (stable transfection with pcDNA). The OCP RT-PCR product is 1020 bp, the Cbfa1 product is 289 bp, the ALP product is 226 bp, the BSP product is 1048 bp and the GapDH (control) product is 450 bp long. M represents protein markers.
  • FIG. 25 shows increased osteoblast proliferation in OCP-transfected ROS cells.
  • FIG. 26 shows the sequences of the primer (SEQ ID NO:19) and QB3 (CMF608) (SEQ ID NO:20).
  • FIG. 27 shows the Adlican amino acid sequence (SEQ ID NO: 21).
  • FIG. 28 shows the Adlican DNA sequence (SEQ ID NO: 22)
  • FIG. 29 shows the predicted DNA sequence of the coding region-ORF of human OCP (SEQ ID NO: 23).
  • FIG. 30 shows the predicted amino acid sequence corresponding to the predicted coding region-ORF of human OCP (SEQ ID NO: 24).
  • FIG. 31 shows the sequence of the N-terminal 663 amino acid fragment derived from the OCP rat protein (SEQ ID NO: 25).
  • FIG. 32 shows the pCM-H-608-663-N-term construct map.
  • FIG. 33 shows the structure of the pKS H608 5′-2.4 Kb bAc#1 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3878).
  • FIG. 34 shows the physical sequence of the 5′ fragment (A) cloned into pBluescript KS to NotI (5′) and HindIII (3′) sites. Fragment A is comprised of the 5′ region (2440 bp) of the complete human OCP sequence and includes, in addition, at the 5′ end, 21 nucleotides of the β-actin “Kozak” region (SEQ ID NO:26).
  • FIG. 35 shows the structure of the pKS H608 m.FRG.3.5 Kb#34 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3876)
  • FIG. 36 shows the physical sequence of the middle fragment (B) cloned into pBluescript KS to HindIII (5′) and SalI (3′) sites. Fragment B is comprised of the central region (3518 bp) of the complete human OCP sequence (SEQ ID NO:27).
  • FIG. 37 shows the structure of the pM H608 3′-1.9 Kb HSTG#3.3 construct (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3877).
  • FIG. 38 shows the physical sequence of the 3′ fragment (C) cloned into pMCS SV(A) to SalI (5′) and SpeI (3′) sites. Fragment C is comprised of the 3′ region (1923 bp, not including the 3 bp stop codon) of the complete human OCP sequence and includes, at the 3′ end, 18 nucleotides coding for 6 Histidine residues (SEQ ID NO:28). Also cloned fragment C contains a silent mutation (C>T transition) compared to the predicted sequence of human OCP ORF. This transition does not change the identity of the encoded amino acid residue.
  • FIG. 39 shows the predicted DNA sequence of Adlican-2 (SEQ ID NO:29). Bases 1555 and 5638 are presented as “g” but could be any other base.
  • FIG. 40 shows the predicted amino acid sequence of human Adlican-2 (SEQ ID NO:30).
  • FIG. 41 shows the amino acid sequence alignment of (i) human Adlican (SEQ ID NO: 21), (ii) human Adlican-2 full amino acid predicted sequence, as determined by the inventors (SEQ ID NO 30), (iii) deduced sequence (hLOC96359) of human Adlican-2 fragment of 539 amino acid residues as found in the database (residues 2036-2652 of SEQ ID NO 30), and (iv) deduced sequence (hLOC90792) of human Adlican-2-fragment of 617 amino acid residues as found in the database (residues 2114-2652 of SEQ ID NO 30).
  • FIG. 42 shows the complete physical DNA sequence of the coding region (ORF) of human OCP (SEQ ID NO: 31).
  • FIG. 43 shows the predicted amino acid sequence corresponding to the complete physical DNA sequence of the coding region (ORF) of human OCP (SEQ ID NO:32).
  • FIG. 44 shows the full rat 608 cDNA sequence (SEQ ID NO:33). This sequence is virtually identical to SEQ ID NO:1, but five unknown nucleotides (designated “n” in SEQ ID NO:1) have been identified. The ORF is from position 575 to 8368.
  • FIG. 45 shows the OCP rat protein amino acid sequence corresponding to the above ORF sequence (SEQ ID NO:34). Three previously unknown amino acids have been identified, as compared to SEQ ID NO:2, where these amino acids are designated “Xaa”.
  • FIG. 46 shows that ALP, which is a biochemical serum marker of bone formation, is significantly increased in 3 month old 608 KO mice.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is related to the discovery of a novel gene, 608 (“OCP”), the expression of which is upregulated by mechanical stress on primary calvaria cells. Several functional features identify OCP as a most specific early marker of osteo- or chondro-progenitor cells as well as an inducer of osteoblast proliferation and differentiation.
  • As used herein, the same gene of the invention may be referred to either as “608” or “OCP.” RNA refers to RNA isolated from cell cultures, cultured tissues or cells or tissues isolated from organisms which are stimulated, differentiated, exposed to a chemical compound, infected with a pathogen, or otherwise stimulated. As used herein, translation is defined as the synthesis of protein encoded by an mRNA template.
  • As used herein, stimulation of translation, transcription, stability or transportation of unknown target mRNA or stimulating element, includes chemically, pathogenically, physically, or otherwise inducing or repressing an mRNA population encoded by genes derived from native tissues and/or cells under pathological and/or stress conditions. In other words, stimulating the expression of an mRNA with a stress inducing element or “stressor” includes, but is not limited to, the application of an external cue, stimulus, or stimuli that stimulates or initiates translation of an mRNA stored as untranslated mRNA in the cells from the sample. The stressor may cause an increase in stability of certain mRNAs, or induce the transport of specific mRNAs from the nucleus to the cytoplasm. The stressor may also induce specific gene transcription. In addition to stimulating translation of mRNA from genes in native cells/tissues, stimulation can include induction and/or repression of genes under pathological and/or stress conditions. The method utilizes a stimulus or stressor to identify unknown target genes regulated at the various possible levels by the stress inducing element or stressor.
  • More in particular, with respect to nucleic acid molecules (rat 608 and human 608 genes) and polypeptides expressed from them, the invention further comprehends isolated and/or purified nucleic acid molecules and isolated and/or purified polypeptides having at least about 70%, preferably at least about 75% or about 77% homology (“substantially homologous”); advantageously at least about 80% or about 83%, such as at least about 85% or about 87% homology (“significantly homologous”); for instance at least about 90% or about 93% homology (“highly homologous”); more advantageously at least about 95%, e.g., at least about 97%, about 98%, about 99% or even about 100% homology (“very highly homologous” to “100% (homologous”); or from about 84-100% homology considered “highly conserved”. The invention also comprehends that these nucleic acid molecules and polypeptides can be used in the same fashion as the herein or aforementioned nucleic acid molecules and polypeptides.
  • Nucleotide sequence homology can be determined using the “Align” program of Myers and Miller, ((1988) CABIOS 4:11-17) and available at NCBI. Alternatively or additionally, the term “homology” for instance, with respect to a nucleotide or amino acid sequence, can indicate a quantitative measure of homology between two sequences. The percent sequence homology can be calculated as (Nref−Ndif)*100/Nref, wherein Ndif is the total number of non-identical residues in the two sequences when aligned and wherein Nref is the number of residues in one of the sequences. Hence, AGTCAGTC has a sequence similarity of 75% to AATCAATC(Nref=8; Ndif=2).
  • Alternatively or additionally, “homology” with respect to sequences can refer to the number of positions with identical nucleotides or amino acid residues divided by the number of nucleotides or amino acid residues in the shorter of the two sequences wherein alignment of the two sequences can be determined in accordance with the Wilbur and Lipman algorithm ((1983) Proc. Natl. Acad. Sci. USA 80:726), for instance, using a window size of 20 nucleotides, a word length of 4 nucleotides, and a gap penalty of 4, and computer-assisted analysis and interpretation of the sequence data including alignment can be conveniently performed using commercially available programs (e.g., Intelligenetics™ Suite, Intelligenetics Inc., CA). When RNA sequences are said to be similar, or have a degree of sequence identity or homology with DNA sequences, thymidine (T) in the DNA sequence is considered equal to uracil (U) in the RNA sequence. RNA sequences within the scope of the invention can be derived from DNA sequences or their complements, by substituting thymidine (T) in the DNA sequence with uracil (U).
  • Additionally or alternatively, amino acid sequence similarity or identity or homology can be determined, for instance, using the BlastP program (Altschul et al. Nucl. Acids Res. 25:3389-3402) and available at NCBI. The following references provide algorithms for comparing the relative homology of amino acid residues of two proteins, and additionally, or alternatively, with respect to the foregoing, the teachings in these references can be used for determining percent homology. Smith et al. (1981) Adv. Appl. Math. 2:482-489; Smith et al. (1983) Nucl. Acids Res. 11:2205-2220; Devereux et al. (1984) Nucl. Acids Res. 12:387-395; Feng et al. (1987) J. Molec. Evol. 25:351-360; Higgins et al. (1989) CABIOS 5:151-153; and Thompson et al. (1994) Nucl. Acids Res. 22:4673-480.
  • As to uses, the inventive genes and expression products as well as genes identified by the herein disclosed methods and expression products thereof and the compositions comprising Adlican or the Adlican gene (including “functional” variations of such expression products, and truncated portions of herein defined genes such as portions of herein defined genes which encode a functional portion of an expression product) are useful in treating, preventing or controlling or diagnosing mechanical stress conditions or absence or reduced mechanical stress conditions.
  • As described herein, Adlican, including functional portions thereof, can be used in all methods suitable for OCP. The sequence homology between Adlican and human OCP provides this novel use of the Adlican protein. Adlican is provided, for instance, in AF245505.1:1.8487. Adlican is named for “Adhesion protein with Leucine-rich repeats has immunoglobulin domains related to perleCAN”; and shows elevated expression in cartilage from osteoarthritis patients. The Adlican gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene. The invention further encompasses compositions comprising a physiologically acceptable excipient and at least one of Adlican, the Adlican gene and antibodies specific to Adlican.
  • OCP expression is related to proliferation and differentation of osteoblasts and chondrocytes. The expression product of OCP, or cells or vectors expressing OCP may cause cells to selectively proliferate and differentiate and thereby increase or alter bone density. Detecting levels of OCP mRNA or expression and comparing it to “normal” non-osteopathic levels may allow one to detect subjects at risk for osteoporosis or lower levels of osteoblasts and chondrocytes.
  • The medicament or treatment can be any conventional medicament or treatment for osteoporosis. Alternatively, or additionally, the medicament or treatment can be the particular protein of the gene detected in the inventive methods, or that which inhibits that protein, e.g., binds to it. Similarly, additionally, or alternatively, the medicament or treatment can be a vector which expresses the protein of the gene detected in the inventive methods or that which inhibits expression of that gene; again, for instance, that which can bind to it and/or otherwise prevents its transcription or translation. The selection of administering a protein or that which expresses it, or of administering that which inhibits the protein or the gene expression, can be done without undue experimentation, e.g., based on down-regulation or up-regulation as determined by inventive methods (e.g., in the osteoporosis model).
  • In the practice of the invention, one can employ general methods in molecular biology. Standard molecular biology techniques known in the art and not specifically described are generally followed as in Sambrook et al. (1989, 1992) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York; and Ausubel et al. (1989) Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, Md.
  • PCR comprising the methods of the invention is performed in a reaction mixture comprising an amount, typically between <10 ng-200 ng template nucleic acid; 50-100 pmoles each oligonucleotide primer; 1-1.25 mM each deoxynucleotide triphosphate; a buffer solution appropriate for the polymerase used to catalyze the amplification reaction; and 0.5-2 Units of a polymerase, most preferably a thermostable polymerase (e.g., Taq polymerase or Tth polymerase).
  • Antibodies may be used in various aspects of the invention, e.g., in detection or treatment or prevention methods. Antibodies can be monoclonal, polyclonal or recombinant for use in the immunoassays or other methods of analysis necessary for the practice of the invention. By the term “antibody” as used in the present invention is meant both poly- and mono-clonal complete antibodies as well as fragments thereof, such as Fab, F(ab′)2, and Fv, which are capable of binding the epitopic determinant. These antibody fragments retain the ability to selectively bind with its antigen or receptor and are exemplified as follows, inter alia:
      • (1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield a light chain and a portion of the heavy chain;
      • (2) (Fab′)2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab′2) is a dimer of two Fab fragments held together by two disulfide bonds;
      • (3) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and
      • (4) Single chain antibody (SCA), defined as a genetically engineered molecule containing the variable region of the light chain and the variable region of the heavy chain linked by a suitable polypeptide linker as a genetically fused single chain molecule.
  • Conveniently, the antibodies may be prepared against the immunogen or antigenic portion thereof for example a synthetic peptide based on the sequence, or prepared recombinantly by cloning techniques or the natural gene product and/or portions thereof may be isolated and used as the immunogen. The genes are identified as set forth in the present invention and the gene product identified. Immunogens can be used to produce antibodies by standard antibody production technology well known to those skilled in the art as described generally in Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.; and Borrebaeck (1992) Antibody Engineering—A Practical Guide, W.H. Freeman and Co. Antibody fragments, as mentioned above, include Fab, F(ab′)2, Fv and scFv prepared by methods known to those skilled in the art. Bird et al. (1988) Science 242:423-426. Any peptide having sufficient flexibility and length can be used as an scFv linker. Usually the linker is selected to have little to no immunogenicity. Linker sequences can also provide additional functions, such as a means for attaching a drug or a solid support.
  • For producing polyclonal antibodies, a host, such as a rabbit or goat, is immunized with the immunogen or an immunogenic fragment thereof, generally with an adjuvant and, if necessary, coupled to a carrier; and antibodies to the immunogen are collected from the sera of the immunized animal. The sera can be adsorbed against related immunogens so that no cross-reactive antibodies remain in the sera rendering the polyclonal antibody monospecific.
  • For producing monoclonal antibodies (mAbs), an appropriate donor, generally a mouse, is hyperimmunized with the immunogen and splenic antibody producing cells are isolated. These cells are fused to an immortal cell, such as a myeloma cell, to provide an immortal fused cell hybrid that secretes the antibody. The cells are then cultured, in bulk, and the mAbs are harvested from the culture media for use. Hybridoma cell lines provide a constant, inexpensive source of chemically identical antibodies and preparations of such antibodies can be easily standardized. Methods for producing mAbs are well known to those of ordinary skill in the art. See, e.g. U.S. Pat. No. 4,196,265.
  • For producing recombinant antibodies, mRNAs from antibody producing B lymphocytes of animals, or hybridomas are reverse-transcribed to obtain cDNAs. See generally, Huston et al. (1991) Met. Enzymol. 203:46-88; Johnson and Bird (1991) Met. Enzymol. 203:88-99; and Mernaugh and Memaugh (1995) In, Molecular Methods in Plant Pathology (Singh and Singh eds.) CRC Press Inc. Boca Raton, Fla., pp. 359-365). Antibody cDNA, which can be full or partial length, is amplified and cloned into a phage or a plasmid. The cDNA can be a partial length of heavy and light chain cDNA, separated or connected by a linker. The antibody, or antibody fragment, is expressed using a suitable expression system to obtain recombinant antibody. Antibody cDNA can also be obtained by screening pertinent expression libraries.
  • Antibodies can be bound to a solid support substrate or conjugated with a detectable moiety or be both bound and conjugated as is well known in the art. For a general discussion of conjugation of fluorescent or enzymatic moieties see, Johnston and Thorpe (1982) Immunochemistry in Practice, Blackwell Scientific Publications, Oxford. The binding of antibodies to a solid support substrate is also well known in the art. See for a general discussion, Harlow and Lane (1988); and Borrebaeck (1992). The detectable moieties contemplated with the present invention include, but are not limited to, fluorescent, metallic, enzymatic and radioactive markers such as biotin, gold, ferritin, alkaline phosphatase, β-galactosidase, peroxidase, urease, fluorescein, rhodamine, tritium, 13C and iodination.
  • Antibodies can also be used as an active agent in a therapeutic composition and such antibodies can be humanized, for instance, to enhance their effects. See, Huls et al. Nature Biotech. 17:1999. “Humanized” antibodies are antibodies in which at least part of the sequence has been altered from its initial form to render it more like human immunoglobulins. In one version, the H chain and L chain C regions are replaced with human sequence. In another version, the CDR regions comprise amino acid sequences from the antibody of interest, while the V framework regions have also been converted human sequences. See, for example, EP 0329400. In a third version, V regions are humanized by designing consensus sequences of human and mouse V regions, and converting residues outside the CDRs that are different between the consensus sequences. The invention encompasses humanized mAbs.
  • The expression product from the gene or portions thereof can be useful for generating antibodies such as monoclonal or polyclonal antibodies which are useful for diagnostic purposes or to block activity of expression products or portions thereof or of genes or a portion thereof, e.g., as therapeutics.
  • Note that some antibodies to the mouse or rat 608 polypeptide may also bind the human 608 polypeptide. A preferred set of antibodies encompassed by this invention are antibodies which bind human 608 polypeptide but which do not bind rat 608 polypeptide. Another preferred set of antibodies encompassed by this invention are antibodies which bind human 608 polypeptide but which do not bind mouse 608 polypeptide
  • The genes of the present invention or portions thereof, e.g., a portion thereof which expresses a protein which function the same as or analogously to the full length protein, or genes identified by the methods herein can be expressed recombinantly, e.g., in Escherichia coli or in another vector or plasmid for either in vivo expression or in vitro expression. The methods for making and/or administering a vector or recombinant or plasmid for expression of gene products of genes of the invention or identified by the invention or a portion thereof either in vivo or in vitro can be any desired method, e.g., a method which is by or analogous to the methods disclosed in: U.S. Pat. Nos. 4,603,112; 4,769,330; 5,174,993; 5,505,941; 5,338,683; 5,494,807; 4,394,448; 4,722,848; 4,745,051; 4,769,331; 5,591,639; 5,589,466; 4,945,050; 5,677,178; 5,591,439; 5,552,143; and 5,580,859; U.S. patent application Serial No. 920,197, filed Oct. 16, 1986; WO 94/16716; WO 96/39491; WO91/11525; WO 98/33510; WO 90/01543; EP 0 370 573; EP 265785; Paoletti (1996) Proc. Natl. Acad. Sci. USA 93:11349-11353; Moss (1996) Proc. Natl. Acad. Sci. USA 93:11341-11348; Richardson (Ed) (1995) Methods in Molecular Biology 39, “Baculovirus Expression Protocols,” Humana Press Inc.; Smith et al. (1983) Mol. Cell. Biol. 3:2156-2165; Pennock et al. (1984) Mol. Cell. Biol. 4:399-406; RoizmanProc. Natl. Acad. Sci. USA 93:11307-11312; Andreansky et al. Proc. Natl. Acad. Sci. USA 93:11313-11318; Robertson et al. Proc. Natl. Acad. Sci. USA 93:11334-11340; Frolov et al. Proc. Natl. Acad. Sci. USA 93:11371-11377; Kitson et al. (1991) J. Virol. 65:3068-3075; Grunhaus et al. (1992) Sem. Virol. 3:237-52; Ballay et al. (1993) EMBO J. 4:3861-65; Graham (1990) Tibtech 8:85-87; Prevec et al. J. Gen. Virol. 70:429-434; Felgner et al. (1994) J. Biol. Chem. 269:2550-2561; (1993) Science 259:1745-49; McClements et al. (1996) Proc. Natl. Acad. Sci. USA 93:11414-11420; Ju et al. (1998) Diabetologia 41:736-739; and Robinson et al. (1997) Sem. Immunol. 9:271-283.
  • The expression product generated by vectors or recombinants can also be isolated and/or purified from infected or transfected cells; for instance, to prepare compositions for administration to patients. However, in certain instances, it may be advantageous to not isolate and/or purify an expression product from a cell; for instance, when the cell or portions thereof enhance the effect of the polypeptide.
  • As used herein, “treatment” refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and may be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of the treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • An inventive vector or recombinant expressing a gene or a portion thereof identified herein or from a method herein can be administered in any suitable amount to achieve expression at a suitable dosage level, e.g., a dosage level analogous to the herein mentioned dosage levels (wherein the gene product is directly present). The inventive vector or recombinant nucleotide can be administered to a patient or infected or transfected into cells in an amount of about at least 103 pfu; more preferably about 104 pfu to about 1010 pfu, e.g., about 105 pfu to about 109 pfu, for instance about 106 pfu to about 108 pfu. In plasmid compositions, the dosage should be a sufficient amount of plasmid to elicit a response analogous to compositions wherein gene product or a portion thereof is directly present; or to have expression analogous to dosages in such compositions; or to have expression analogous to expression obtained in vivo by recombinant compositions. For instance, suitable quantities of plasmid DNA in plasmid compositions can be 1 μg to 100 mg, preferably 0.1 to 10 mg, e.g., 500 μg, but lower levels such as 0.1 to 2 mg or preferably 1-10 μg may be employed. Documents cited herein regarding DNA plasmid vectors can be consulted for the skilled artisan to ascertain other suitable dosages for DNA plasmid vector compositions of the invention, without undue experimentation.
  • Compositions for administering vectors can be as in or analogous to such compositions in documents cited herein or as in or analogous to compositions herein described, e.g., pharmaceutical or therapeutic compositions and the like.
  • Thus, the invention comprehends in vivo gene expression which is sometimes termed “gene therapy.” Gene therapy can refer to the transfer of genetic material (e.g. DNA or RNA) of interest into a host subject or patient to treat or prevent a genetic or acquired disease, condition or phenotype. The particular gene that is to be used or which has been identified as the target gene is identified as set forth herein. The genetic material of interest encodes a product (e.g. a protein, polypeptide, peptide or functional RNA) the production in vivo of which is desired. For example, the genetic material of interest can encode a hormone, receptor, enzyme, polypeptide or peptide of therapeutic value. For a review see, in general, the text “Gene Therapy” (Advances in Pharmacology 40, Academic Press, 1997).
  • Two basic approaches to gene therapy have evolved: (1) ex vivo; and (2) in vivo gene therapy. In ex vivo gene therapy cells are removed from a patient, and while being cultured are treated in vitro. Generally, a functional replacement gene is introduced into the cell via an appropriate gene delivery vehicle/method (transfection, homologous recombination, etc.) and, an expression system as needed and then the modified cells are expanded in culture and returned to the host/patient. These genetically reimplanted cells have been shown to produce the transfected gene product in situ. In in vivo gene therapy, target cells are not removed from the subject; rather, the gene to be transferred is introduced into the cells of the recipient organism in situ, that is within the recipient. Alternatively, if the host gene is defective, the gene is repaired in situ. Culver (1998) Antisense DNA & RNA Based Therapeutics, February, 1998, Coronado, Calif. These genetically altered cells have been shown to produce the transfected gene product in situ.
  • The gene expression vehicle is capable of delivery/transfer of heterologous nucleic acid into a host cell. The expression vehicle may include elements to control targeting, expression and transcription of the nucleic acid in a cell-selective manner as is known in the art. It should be noted that often the 5′UTR and/or 3′UTR of the gene may be replaced by the 5′ UTR and/or 3′UTR of the expression vehicle. Therefore, as used herein, the expression vehicle may, as needed, not include the 5′UTR and/or 3′UTR shown in sequences herein and only include the specific amino acid coding region.
  • The expression vehicle can include a promoter for controlling transcription of the heterologous material and can be either a constitutive or inducible promoter to allow selective transcription. Enhancers that may be required to obtain necessary transcription levels can optionally be included. Enhancers are generally any non-translated DNA sequence that works contiguously with the coding sequence (in cis) to change the basal transcription level dictated by the promoter. The expression vehicle can also include a selection gene as described herein.
  • Vectors can be introduced into cells or tissues by any one of a variety of known methods within the art. Such methods can be found generally described in Sambrook et al. (1989, 1992); Ausubel et al. (1989); Chang et al. (1995) Somatic Gene Therapy, CRC Press, Ann Arbor, Mich.; Vega et al. (1995) Gene Targeting, CRC Press, Ann Arbor, Mich.; Vectors: A Survey of Molecular Cloning Vectors and Their Uses, Butterworths, Boston Mass. (1988); and Gilboa et al. (1986) BioTech. 4:504-512, as well as other documents cited herein and include, for example, stable or transient transfection, lipofection, electroporation and infection with recombinant viral vectors. In addition, see U.S. Pat. No. 4,866,042 for vectors involving the central nervous system; and also U.S. Pat. Nos. 5,464,764 and 5,487,992 for positive-negative selection methods.
  • Introduction of nucleic acids by infection offers advantages over the other listed methods. Higher efficiency can be obtained due to their infectious nature. Moreover, viruses are very specialized and typically infect and propagate in specific cell types. Thus, their natural specificity can be used to target the vectors to specific cell types in vivo or within a tissue or mixed cell culture. Viral vectors can also be modified with specific receptors or ligands to alter target specificity through receptor-mediated events.
  • Additional features can be added to the vector to ensure its safety and/or enhance its therapeutic efficacy. Such features include, for example, markers that can be used to negatively select against cells infected with the recombinant virus. An example of such a negative selection marker is the TK gene described above that confers sensitivity to the antibiotic gancyclovir. Negative selection is therefore a means by which infection can be controlled because it provides inducible suicide through the addition of antibiotic. Such protection ensures that if, for example, mutations arise that produce altered forms of the viral vector or recombinant sequence, cellular transformation will not occur. Features that limit expression to particular cell types can also be included. Such features include, for example, promoter and regulatory elements that are specific for the desired cell type.
  • In addition, recombinant viral vectors are useful for in vivo expression of a desired nucleic acid because they offer advantages such as lateral infection and targeting specificity. Lateral infection is inherent in the life cycle of, for example, retrovirus and is the process by which a single infected cell produces many progeny virions that bud off and infect neighboring cells. The result is that a large area becomes rapidly infected, most of which was not initially infected by the original viral particles. This is in contrast to vertical-type of infection in which the infectious agent spreads only through daughter progeny. Viral vectors can also be produced that are unable to spread laterally. This characteristic can be useful if the desired purpose is to introduce a specified gene into only a localized number of targeted cells.
  • In particular, use of the 608 gene (or a functional fragment thereof) for treatment of osteoporosis, and/or osteoarthritis, and/or osteopetrosis, and/or osteosarcoma, and/or fracture healing is envisaged using gene therapy methods. As described above, a plasmid or DNA vector expressing the gene could be injected directly to the target tissue; alternatively a virus bearing a plasmid or DNA vector expressing the gene could be injected directly to the target tissue. Another embodiment is that cells transfected with a plasmid or DNA vector expressing the gene could be injected directly to the target tissue. These transfected cells should preferably be the patient's own cells for example mesenchymal stem cells drawn from the bone marrow.
  • Delivery of gene products (products from herein defined genes: genes identified herein or by inventive methods or portions thereof) and/or antibodies or portions thereof and/or agonists or antagonists (collectively or individually “therapeutics”), and compositions comprising the same, as well as of compositions comprising a vector expressing gene products, can be done without undue experimentation from this disclosure and the knowledge in the art.
  • The pharmaceutically “effective amount” for purposes herein is thus determined by such considerations as are known in the art. The amount must be effective to achieve improvement including but not limited to improved survival rate or more rapid recovery, or improvement or amelioration or elimination of symptoms and other indicators, e.g., of osteoporosis, for instance, improvement in bone density, as are selected as appropriate measures by those skilled in the art.
  • It is noted that humans are treated generally longer than the mice or other experimental animals exemplified herein. Human treatment has a length proportional to the length of the disease process and drug effectiveness. The doses may be single doses or multiple doses over a period of several days, but single doses are preferred. Thus, one can scale up from animal experiments, e.g., rats, mice, and the like, to humans, by techniques from this disclosure and the knowledge in the art, without undue experimentation.
  • The present invention provides an isolated nucleic acid molecule containing nucleotides having a sequence set forth in at least one of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No. PTA-3638, supplements thereof and a polynucleotide having a sequence that differs from SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No. PTA-3638 due to the degeneracy of the genetic code or a sequence which hybridizes under stringent conditions to a sequence in a plasmid designated pCm-H608-663-N-term or a functional portion thereof or a polynucleotide which is at least substantially homologous thereto. In a preferred embodiment, the nucleic acid molecule comprises a polynucleotide having at least 15 nucleotides from SEQ ID NO: 1, SEQ ID NO:3, SEQ ID NO: 6, SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 29 or SEQ ID NO: 31 or as inserted in a plasmid designated pCm-H-608-663-N-term, deposited under ATCC Accession No. PTA-3638, preferably at least 50 nucleotides and more preferably at least 100 nucleotides.
  • The present invention further provides an isolated nucleic acid molecule containing nucleotides having a sequence set forth in at least one of SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO: 28, or SEQ ID NO:26 and SEQ ID NO:27 or SEQ ID NO:26 and SEQ ID NO:27 and SEQ ID NO: 28 or as inserted in a plasmid designated pKS H608 5′-2.4 Kb bAc#1 (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3878), pKS H608 m.FRG.3.5 Kb#34 (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3876) or pM H608 3′-1.9 Kb HSTG#3.3 (deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession number PTA-3877), supplements thereof and a polynucleotide having a sequence that differs from SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO: 28, or SEQ ID NO:26 and SEQ ID NO:27 or SEQ ID NO:26 and SEQ ID NO:27 and SEQ ID NO: 28 or as inserted in a plasmid designated pKS H608 5′-2.4 Kb bAc#1, pKS H608 m.FRG.3.5 Kb#34 or pM H608 3′-1.9 Kb HSTG#3.3 due to the degeneracy of the genetic code or a sequence which hybridizes under stringent conditions to a sequence in a plasmid designated pKS H608 5′-2.4 Kb bAc#1, pKS H608 m.FRG.3.5 Kb#34 or pM H608 3′-1.9 Kb HSTG#3.3 or a functional portion thereof or a polynucleotide which is at least substantially homologous thereto. In a preferred embodiment, the nucleic acid molecule comprises a polynucleotide having at least 15 nucleotides from SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO: 28, or SEQ ID NO:26 and SEQ ID NO:27 or SEQ ID NO:26 and SEQ ID NO:27 and SEQ ID NO: 28 or as inserted in a plasmid designated pKS H608 5′-2.4 Kb bAc#1, pKS H608 m.FRG.3.5 Kb#34 or pM H608 3′-1.9 Kb HSTG#3.3, preferably at least 50 nucleotides and more preferably at least 100 nucleotides.
  • The present invention also provides a composition of the isolated nucleic acid molecule, a vector comprising the isolated nucleic acid molecule, a composition containing said vector and a method for preventing, treating or controlling bone diseases including, but not limited to, osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis, bone fractures or low bone density or or other conditions involving mechanical stress or a lack thereof in a subject, comprising administering the inventive composition, or the inventive vector, and a method for preparing a polypeptide comprising expressing the isolated nucleic acid molecule or comprising expressing the polypeptide from the vector.
  • The present invention further provides a method for preventing, treating or controlling osteoporosis, osteopenia, osteopetrosis, osteosclerosis, osteoarthritis, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or a lack thereof in a subject, comprising administering an isolated nucleic acid molecule or functional portion thereof or a polypeptide comprising an expression product of the gene or functional portion of the polypeptide or an antibody to the polypeptide or a functional portion of the antibody. In one embodiment of the invention, the isolated nucleic acid molecule encodes a 10 kD to 100 kD N-terminal cleavage product of the OCP protein. Preferably, the N-terminal cleavage product comprises of a polypeptide of about 25 kD. More preferably the N-terminal cleavage product comprises a polypeptide of about 70-80 kD, most preferably about 1-663 amino acids or about 1-741 amino acids of the OCP protein. The present invention provides an isolated polypeptide encoded by the inventive polynucleotide. In one embodiment of the invention, the polypeptide is identified as human 608 protein, rat 608 protein, human Adlican-2 protein or a functional portion thereof or a polypeptide which is at least substantially homologous thereto. More particularly this invention is directed to an isolated polypeptide wherein the functional portion comprises consecutive amino acids having a sequence set forth in SEQ ID NO:2, SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 30, SEQ ID NO: 32 or SEQ ID NO: 34. Particular fragments of the polypeptide are about the first 663 amino acids or about the first 741 amino acids of the sequence set forth in SEQ ID NO:2, SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 30, SEQ ID NO: 32 or SEQ ID NO: 34. Other particular fragments of the human 608 protein include amino acids 1-500, 501-1000, 1001-1500, 1501-2000, 2001-2500, and 2051-2623 of the sequence set forth in SEQ ID NO: 32. Further particular fragments of the human 608 protein include amino acids 250-749, 750-129, 1250-1749, 1750-2249 and 2250-2623 of the sequence set forth in SEQ ID NO: 32. Nucleic acid molecules (polynucleotides) encoding these particular fragments are also envisaged as aspects of the invention. Similar particular polypeptide fragments of the Adlican-2 protein (SEQ ID NO: 30), and similar particular polynucleotide fragments of the Adlican-2 nucleic acid (SEQ ID NO: 29) are also envisaged as aspects of the invention.
  • The present invention also provides a composition comprising one or of isolated polypeptides, an antibody specific for the polypeptide or a functional portion thereof, a composition comprising the antibody or a functional portion thereof, and a method for treating or preventing osteoporosis, or fracture healing, bone elongation, or periodontosis in a subject, comprising administering to the subject a N-terminal polypeptide having a molecular weight of between 10 kD and 100 kD, preferably about 25 kD to about 70-80 kD.
  • The present invention provides for a method of treating or preventing osteoarthritis, osteopetrosis, or osteosclerosis, comprising administering to a subject an effective amount of a chemical or a neutralizing mAbs that inhibit the activity of the N-terminal polypeptide having a molecular weight of between 10 kD and 30 kD, preferably about 25 kD.
  • As used herein, the term “subject,” “patient,” “host” include, but are not limited to human, bovine, pig, mouse, rat, goat, sheep and horse.
  • Those skilled in the art will recognize that the components of the compositions should be selected to be chemically inert with respect to the gene product and optional adjuvant or additive. This will present no problem to those skilled in chemical and pharmaceutical principles, or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein.
  • The present invention provides receptors of the expression products of human mechanical stress induced genes and their functional equivalents, such as OCP and Adlican, and methods or processes for obtaining and using such receptors. The receptors of the present invention are those to which the expression products of mechanical stress induced genes and their functional equivalents bind or associate as determined by conventional assays, as well as in vivo. For example, binding of the polypeptides of the instant invention to receptors can be determined in vitro, using candidate receptor molecules that are associated with lipid membranes. See, e.g., Watson, J. et al., Development of FlashPlate® technology to measure (35S) GTP gamma S binding to Chinese hamster ovary cell membranes expressing the cloned human 5-HT1B receptor, Journal of Biomolecular Screening. Summer, 1998; 3 (2) 101-105; Komesli-Sylviane et al., Chimeric extracellular domain of type II transforming growth factor (TGF)-beta receptor fused to the Fc region of human immunoglobulin as a TGF-beta antagonist, European Journal of Biochemistry. June, 1998; 254 (3) 505-513. See, generally, Darnell et al., Molecular Cell Biology, 644-646, Scientific American Books, New York (1986). Scanning electron microscopy (“SEM”), x-ray crystallography and reactions using labelled polypeptides are examples of conventional means for determining whether polypeptides have bound or associated with a receptor molecule. For instance, X-ray crystallography can provide detailed structural information to determine whether and to what extent binding or association has occurred. See, e.g., U.S. Pat. No. 6,037,117; U.S. Pat. No. 6,128,582 and U.S. Pat. No. 6,153,579. Further, crystallography, including X-ray crystallography, provides three-dimensional structures that show whether a candidate polypeptide ligand can or would bind or associate with a target molecule, such as a receptor. See, e.g., WO 99/45379; U.S. Pat. Nos. 6,087,478 and 6,110,672. Such binding or association shows that the receptor molecule is the receptor for the candidate polypeptide.
  • With the disclosures in the present specification of the inventive genes, expression products and uses thereof, those skilled in the art can obtain by conventional methods the receptors for the inventive expression products. The conventional means for obtaining the receptors include raising monoclonal antibodies (Mabs) to candidate receptors, purifying the receptors from a tissue sample by use of an affinity column, treatment with a buffer, and collection of the eluate receptor molecules. Other means of isolating and purifying the receptors are conventional in the art, for instance isolation and purification by dialysis, salting out, and electrophoretic (e.g. SDS-PAGE) and chromatographic (e.g. ion-exchange and gel-filtration, in additional to affinity) techniques. Such methods can be found generally described in Stryer, Biochemistry, 44-50, W.H. Freeman & Co., New York (3d ed. 1988); Darnell et al., Molecular Cell Biology, 77-80 (1986); Alberts et al., Molecular Biology of the Cell, 167-172, 193 Garland Publishing, New York (2d ed. 1989).
  • Sequencing of the isolated receptor involves methods known in the art, for instance directly sequencing a short N-terminal sequence of the receptor, constructing a nucleic-acid probe, isolating the receptor gene, and determining the entire amino-acid sequence of the receptor from the nucleic-acid sequence. Alternatively, the entire receptor protein can be sequenced directly. Automated Edman degradation is one conventional method used to partially or entirely sequence a receptor protein, facilitated by chemical or enzymatic cleavage. Automated sequenators, such as an ABI-494 Procise Sequencer (Applied Biosystems) can be used. See, generally, Stryer, Biochemistry, 50-58 (3d ed. 1988).
  • The invention provides methods for using such receptors in assays, for instance for identifying proteins or polypeptides that bind to, associate with or block the inventive receptors, determining binding constants and degree of binding, and for testing the effects of such polypeptides, for instance utilising membrane receptor preparations. See Watson (1998); Komesli-Sylviane (1998). For instance, FlashPlate® (Perkin-Elmer, Massachusetts, USA) technology can be used with the present invention to determine whether and to what degree candidate polypeptides bind to and are functional with respect to a receptor of the invention.
  • Diagnostics:
  • The gene and polypeptides of the invention can be employed as a diagnostic in several ways as follows:
  • 1. Diagnosis of osteoarthritis by detection of 608 protein or parts of it, or detection of 608 RNA in synovial fluid.
    2. Diagnosis of osteoporosis by detection of 608 protein or fragments thereof, or detection of 608 RNA, preferably in a blood sample.
    3. Diagnosis of a fracture by detection of 608 protein or fragments thereof, or detection of 608 RNA, preferably in a blood sample.
    4. Diagnosis of succeptibility to osteoporosis, and/or osteoarthritis, and/or osteopetrosis, and/or osteosarcoma associated with mutated 608 by PCR or RT PCR of DNA or RNA respectively. DNA and/or RNA from bodily fluids or from a tissue, and preferably DNA from blood are tested.
    5. Diagnosis of a disease associated with mutated 608 by PCR or RT PCR of DNA or RNA respectively. DNA and/or RNA from bodily fluids or from a tissue and preferably DNA from blood are tested
  • The diagnostic methods to be utilized are described in more detail as follows.
  • In diagnosis, the sample is taken from a bodily fluid or from a tissue, preferably bone or cartilage tissue; the bodily fluid is selected from the group of fluid consisting of blood, lymph fluid, ascites, serous fluid, pleural effusion, sputum, cerebrospinal fluid, lacrimal fluid, synovial fluid, saliva, stool, sperm and urine, preferably blood or urine. Measurement of level of the 608 polypeptide may be determined by a method selected from the group consisting of immunohistochemistry, western blotting, ELISA, antibody microarray hybridization and targeted molecular imaging; antibodies have been described above. Such methods are well-known in the art, for example for immunohistochemistry: M. A. Hayat (2002) Microscopy, Immunohistochemistry and Antigen Retrieval Methods: For Light and Electron Microscopy, Kluwer Academic Publishers; Brown C (1998): “Antigen retrieval methods for immunohistochemistry”, Toxicol Pathol; 26(6): 830-1); for western blotting: Laemmeli UK(1970): “Cleavage of structural proteins during the assembley of the head of a bacteriophage T4”, Nature; 227: 680-685; and Egger & Bienz(1994) “Protein (western) blotting”, Mol Biotechnol; 1(3): 289-305); for ELISA: Onorato et al.(1998) “Immunohistochemical and ELISA assays for biomarkers of oxidative stress in aging and disease”, Ann NY Acad Sci 20; 854: 277-90); for antibody microarray hybridization: Huang(2001) “Detection of multiple proteins in an antibody-based protein microarray system, Immunol Methods 1; 255 (1-2): 1-13); and for targeted molecular imaging: Thomas (2001). Targeted Molecular Imaging in Oncology, Kim et al (Eds)., Springer Verlag, inter alia.
  • Measurement of level of 608 polynucleotide may be determined by a method selected from: RT-PCR analysis, in-situ hybridization, polynucleotide microarray and Northern blotting. Such methods are well-known in the art, for example for in-situ hybridization Andreeff & Pinkel (Editors) (1999), “Introduction to Fluorescence In Situ Hybridization: Principles and Clinical Applications”, John Wiley & Sons Inc.; and for Northern blotting Trayhurn (1996) “Northern blotting”, Proc Nutr Soc; 55(1B): 583-9 and Shifman & Stein(1995) “A reliable and sensitive method for non-radioactive Northern blot analysis of nerve growth factor mRNA from brain tissues”, Journal of Neuroscience Methods; 59: 205-208 inter alia.
  • A better understanding of the present invention and of its many advantages will be had from the following examples, given by way of illustration and as a further description of the invention.
  • EXPERIMENTAL DETAILS Example 1 608 Gene Expression by In Situ Hybridization
  • The 608 gene expression pattern was studied by in situ hybridization on sections of bones from ovariectomized and sham-operated rats. Female Wistar rats weighing 300-350g were subjected to ovariectomy under general anesthesia. Control rats were operated on in the same way but ovaries were not excised as a sham operation.
  • Three weeks after the operation, rats were sacrificed and tibia were excised together with the knee joint. Bones were fixed for three days in 4% paraformaldehyde and then decalcified for four days in a solution containing 5% formic acid and 10% formalin. Decalcified bones were postfixed in 10% formalin for three days and embedded into paraffin.
  • The ectopic bone formation model was employed to study the bone development 608 gene expression pattern. Rat bone marrow cells were seeded into cylinders of demineralized bone matrix prepared from rat tibiae. Cylinders were implanted subcutaneously into adult rats. After three weeks, rats were sacrificed and implants were decalcified and embedded into paraffin as described above for tibial bones.
  • The 6 μm sections were prepared and hybridized in situ. After hybridization, sections were dipped into nuclear track emulsion and exposed for three weeks at 4° C. Autoradiographs were developed, stained with hematoxylin-eosin and studied under microscopy using brightfield and darkfield illumination.
  • For further assessment of cell and tissue specificity of 608 gene expression, an in situ hybridization study was performed on sections of multitissue block containing multiple samples of adult rat tissues. The 608 expression developmental pattern was studied on sagittal sections of mouse embryos of 12.5, 14.5 and 16.5 days postconception (dpc) stages.
  • Microscopic study of hybridized sections of long bones revealed a peculiar pattern of 608 probe hybridization. The hybridization signal can be seen mainly in fibroblast-like cells found in several locations throughout the sections. Prominent accumulations of these cells can be seen in the area of periosteal modeling in metaphysis, and also in regions of active remodeling of compact bone in diaphysis: at the boundary between bone marrow and endosteal osteoblasts and in periosteum; also in close contact with osteoblasts. Perivascular connective tissue filling Volkmann's canals in compact bone in diaphysis and epiphysis also contains 608-expressing cells. No hybridization was found within cancellous bone and in bone marrow. This hybridization pattern suggests that cells expressing 608 are associated with areas of remodeling of preexisting bone and are not involved in primary endochondral ossification.
  • At the growth plate level, 608 expressing cells can be seen in the perichondral fibrous ring of LaCroix. Some investigators regard this fibrous tissue as the aggregation of residual mesenchymal cells able to differentiate into both osteoblasts and chondrocytes. In this respect it is noteworthy that single cells expressing 608 can be seen in epiphyseal cartilage. These 608-expressing cells are rounded cells within the lateral segment of epiphysis (sometimes in close vicinity to the LaCroix ring) and flattened cells covering the articulate surface. Most cells in articulate cartilage and all chondrocytes on the growth plate do not show 608 expression. Ovariectomy did not alter the intensity and pattern of 608 expression in bone tissue.
  • In ectopic bone sections, 608 hybridization signal can be seen in some fibroblast-like cells either scattered within unmineralized connective tissue matrix or concentrated at the boundary between this tissue and osteoblasts of immature bone. 608 gene expression patterns revealed by in situ hybridization in bone and cartilage indicate that its expression marks some skeletal tissue elements able to differentiate into two skeletal cell types—osteoblasts and chondrocytes. The terminal differentiation of these cells appears to be accompanied by down-regulation of 608 expression. The latter observation is supported by peculiar temporal pattern of 608 expression in primary cultures of osteogenic cells isolated from calvaria bones of rat fetuses. In these cultures, expression was revealed by in situ hybridization in the vast majority of cells after one and two weeks of incubation in vitro. Three and four week old cultures showing signs of ossification contained no 608 expressing cells. Significantly, no hybridization signal was found on sections of multitissue block hybridized to 608 probe suggesting high specificity of this gene expression for the skeletal tissue in adult organisms.
  • An in situ hybridization study of embryonic sections demonstrated that, at 12.5 dpc, a weak hybridization signal can be discerned in some mesenchymal cells in several locations throughout the embryonic body. The most prominent signal is found in the head in loose mesenchymal tissue surrounding the olfactory epithelium and underlying the surface epithelium of nose tip. Other mesenchymal cells in the head also show a hybridization signal: non-cartilaginous part of basisphenoid bone primordium and mesenchyme surrounding the dental laminae (tooth primordia) in the mandible.
  • In the trunk, expression can be detected in less developed vertebrae primordia in the thoraco-lumbar region. The hybridization signal here marks the condensed portion of sclerotomes. Another area of the trunk showing a hybridization signal is comprised of a thin layer of mesenchymal cells in the anterior part of the thoracic body wall.
  • At later stages of development, 14.5 and 16.5 dpc, probe 608 gave no hybridization signal. Thus, it appears that during embryonic development the 608 gene is transiently expressed by at least some mesenchymal and skeleton-forming cells. This expression is down-regulated at later stages of development. More detailed study of late embryonic and postnatal stages of development reveals the timing of appearance of cells expressing 608 in bone tissue.
  • Further experiments to study the expression of the OCP gene in embryonic development were performed as follows. The targeting vector used to produce OCP knockout mice included a “knock-in” of the β-galactosidase (LacZ) reporter gene into the OCP gene. The LacZ gene was fused to the first exon of the OCP gene—a non-coding exon. Thus, expression of LacZ is expected to depend on the OCP regulatory elements and to mark the cells expressing OCP.
  • Analysis of LacZ staining was performed during embryonic development on OCP knockout mice. The expression pattern revealed by this analysis reflects the activation of the OCP gene promoter, which results in expression of the knocked-in LacZ gene. This data in general supports the pattern detected by the in-situ hybridization described above.
  • At 10.5 dpc expression is seen in the apical ectodermal ridge (AER), in the forelimbs only. This specialized region, together with the zone of polarizing activity (ZPA), directs and coordinates the development of the limb bud.
  • At 12.5 dpc expression in AER is maintained in the forelimbs and appears also in hindlimbs. In addition, it appears in precartilagenous condensations of the ribs, in mesenchymal tissue in the face, in mesenchymal tissue rostral to the forelimb, a region of future muscle development, and in the tip of the genital tubercle.
  • At 14.5 dpc there is broader expression, in the head region, limbs, ribs, and back. Although no expression at 14.5 dpc was detected in the in situ RNA hybridization experiments described above, expression was detected in this experiment, probably because the lacZ staining is a more sensitive detection method.
  • To summarize:
      • 1. There is an interesting pattern of expression of gene OCP in embryonic development: in tissues originating from different germ layers (ectoderm and mesoderm), in critical regions of pattern formation (AER), and an apparently regulated pattern during cartilage and bone development.
      • 2. In mesodermal tissues, the gene is expressed mainly in skeletal lineages, but also in some myoblasts and some dermal cells as well.
      • 3. Ectodermal expression appears in the head mesenchyme, derived from neural crest cells, and cells in the apical ectodermal ridge.
  • The 608 expression pattern during embryonic development is closely coupled with regions of bone and cartilage development. This expression pattern strongly suggests a role for 608 in bone metabolism.
  • Example 2 Isolation of Rat OCP
  • Primary rat calvaria cells grown on elastic membranes that were stretched for 20 minutes provided a model system for a stimulator of bone formation following mechanical force. Gene expression patterns were compared before and after the application of mechanical force.
  • OCP expression was upregulated approximately 3-fold by mechanical force. This was detected both by microarray analysis and by Northern blot analysis using poly (A)+ RNA from rat calvaria cells before and after the mechanical stress. In rat calvaria primary cells and in rat bone extract this gene was expressed as a main RNA species of approximately 8.9 kb and a minor RNA transcript of approximately 9 kb. The hybridization signal was not detected in any other rat RNA from various tissue sources, including testis, colon, intestine, kidney, stomach, thymus, lung, uterus, heart, brain, liver, eye, and lymph node.
  • The partial OCP rat cDNA clone (4007 bp long) isolated from a rat calvaria cDNA phage library was found to contain a 3356 bp open reading frame closed at the 3′ end. Comparison to public mouse databases revealed no sequence homologues. A complete OCP rat cDNA clone was isolated from the rat calvaria cDNA library by a combination of 5′ RACE technique (Clontech), RT-PCR of 5′ cDNA fragments, and ligation of the latter products to the original 3′ clone. The full rat cDNA clone that was generated (shown in FIG. 1-SEQ ID NO:1) was sequenced, and no mutations were found. The full sequence stretch is 8883 bp long and contains an ORF (nt 575-8366) for a 2597 amino acid residue protein. FIG. 3-SEQ ID NO:2. The cDNA does not contain a polyadenylation site, but contains a 3′ poly A stretch.
  • 608 encodes a large protein that appears to be a part of the extra-cellular matrix. The gene may be actively involved in supporting osteoblast differentiation. Another option is that it is expressed in regions were remodeling takes place. Such an hypothesis is also compatible with a role in directing osteoclast action and thus it may be a target for inhibition by small molecules.
  • In normal bone formation, activation of osteoblasts leads to secretion of various factors that attract osteoclast precursors or mature osteoclasts to the sites of bone formation to initiate the process of bone resorption. In normal bone formation both functions are balanced. Imbalance to any side causes either osteitis deformans (osteoblast function overwhelms) or osteoporosis (osteoclast function overwhelms).
  • Among known osteoblast activators—mechanical force stimulation—is actually applied in the present model. As proof of principle, increased expression of several genes known to respond to mechanical stress by transcriptional upregulation were found. They include tenascin, endothelin and possibly trombospondin.
  • Example 3 Full-Length OCP cDNA Construction and Expression
  • TNT (transcription-translation) assays were performed according to the manufacturer's instructions (Promega—TNT coupled reticulocyte lysate systems), using specific fragments taken from various regions of the gene. In all assays a clear translation product was observed. The following fragments were tested:
  • TNT products
    Translation
    Frag. Location Fragment size (bp) product size (kD) Promoter
    1 134-2147 2013 73 T7
    2 3912-5014  1102 40
    3 574-1513 939 34
  • Example 4 The Mouse OCP Gene
  • Two mouse genomic Bac clones containing the mouse OCP gene promoter region and part of the coding region were identified, based on their partial homology to the 5′UTR region of the rat-608 cDNA. These clones (23-261L4 and 23-241H7 with ˜200 Kb average insert length) were bought from TIGR.
  • Specific primers for the amplification of a part of the mouse-OCP promoter region were designed and used for PCR screening of a mouse genomic phage library (performed by Lexicon Genetics Inc. for the Applicants). One phage clone containing part of the genomic region of the mouse 608 gene was detected and completely sequenced. The length of this clone was reported to be 11,963 bp. Parts of the physical “Lexicon” clone were re-sequenced by the inventors and corrections were made. The resequenced clone is 11,967 bp long. Exon-location prediction (FIG. 4) was performed based on the alignment of the mouse genomic and the rat cDNA sequences.
  • Example 5 The Human OCP Gene
  • On the nucleotide level, the rat OCP cDNA sequence is homologous to the human genomic DNA sequence located on chromosome 3. Based on the homology and bioinformatic analysis (FIG. 6), a putative cDNA sequence was generated. FIG. 7. The highest similarity is evident between nt 1-1965 (1-655 a.a); 2179-2337 (727-779 a.a); and 4894-7833 (1635 a.a.-end) as presented in the table shown in FIG. 8. On the protein level, no homologues were found in the data bank.
  • Example 6 The Deduced OCP Protein
  • The deduced OCP protein was generated following the alignment of the rat, mouse and human cDNA sequences and the equivalent rat, mouse and human amino acid sequences, respectively. The following alignments were made: (a) alignment of rat, human, and mouse OCP cDNA coding regions (rat cDNA: SEQ ID NO:7; human 5+3 corrected: SEQ ID NO:8; and mus cDNA 5: SEQ ID NO:9)
  • (b) alignment of rat, human and mouse OCP proteins (rat: SEQ ID NO:10; human 5+3 corrected: SEQ ID NO:11; and mouse 5 corrected: SEQ ID NO: 12) and
    (c) alignment of rat and human OCP proteins (rat: SEQ ID NO:13; and human 5+3 corrected: SEQ ID NO:14).
  • The deduced OCP protein(FIG. 10): contains the following features
  • a. a cleavable, well-defined N-terminal signal peptide (aa 1-28);
  • b. a leucine-rich repeat region (aa 28-280). This region can be divided into N-terminal and C-terminal domains of leucine-rich repeats (aa 28-61 and 219-280, respectively). Between them, there are six leucine-rich repeat outliers (aa 74-96, 98-120, 122-144, 146-168, 178-200, 202-224). Leucine rich repeats are usually found in extracellular portions of a number of proteins with diverse functions. These repeats are thought to be involved in protein-protein interactions. Each leucine-rich repeat is composed of β-sheet and α-helix. Such units form elongated non-globular structures;
  • c. twelve immunoglobulin C-2 type repeats at amino acid positions 488-558, 586-652, 1635-1704, 1732-1801, 1829-1898, 1928-1997, 2025-2100, 2128-2194, 2233-2294, 2324-2392, 2419-2487, 2515-2586. Thus, two Ig-like repeats are found immediately downstream of a leucine-rich region, while the remaining 10 repeats are clustered at the protein's C-terminus. Immunoglobulin C-2 type repeats are involved in protein-protein interaction and are usually found in extracellular protein portions;
  • d. no transmembrane domain; and
  • 5 nuclear localization domains (NLS) at: 724, 747, 1026, 1346 and 1618.
  • These observations indicate that OCP belongs to the Ig superfamily. OCP is a serine-rich protein (10.3% versus av. 6.3%), with a central nuclear prediction domain and an N-terminal extracellular prediction domain.
  • Example 7 Bone Fracture Healing
  • Expression of 608 RNA is bone-specific. Moreover, it seems to be specific to bone progenitors (as judged by their location in bone and involvement in normal bone modeling and remodeling processes) that do not yet express the known bone-specific markers. To further prove the relevance of 608-expressing cells to osteogenic lineage, the patterns of 608 expression in the animal model of bone fracture healing that imply the activation of bone formation processes were studied.
  • The sequence of physiological events following bone fracture is now relatively well understood. Healing takes place in three phases-inflammatory, reparative and remodeling. In each phase certain cells predominate and specific histological and biochemical events are observed. Although these phases are referred to separately, it is well known that events described in one phase persist into the next and events apparent in a subsequent phase begin before this particular phase predominates. These events have been described over the years in investigative reports and review articles. Ham (1969) In, Histology, 6th ed. Philadelphia, Lippincott, p. 441; and Urist and Johnson (1943) J. Bone Joint Surg. 25:375.
  • During the first phase immediately following fracture (the inflammatory phase), wide-spread vasodilatation and exudation of plasma lead to the acute edema visible in the region of a fresh fracture. Acute inflammatory cells migrate to the region, as do polymorphonuclear leukocytes and then macrophages. The cells that participate directly in fracture repair during the second phase (the reparative phase), are of mesenchymal origin and are pluripotent. These cells form collagen, cartilage and bone. Some cells are derived from the cambium layer of the periosteum and form the earliest bone. Endosteal cells also participate. However, the majority of cells directly taking part in fracture healing enter the fracture site with the granulation tissue that invades the region from surrounding vessels. Trueta (1963) J. Bone Joint Surg. 45:402. Note that the entire vascular bed of an extremity enlarges shortly after the fracture has occurred but the osteogenic response is limited largely to the zones surrounding the fracture itself. Wray (1963) Angiol. 14:134.
  • The invading cells produce tissue known as “callus” (made up of fibrous tissue, cartilage, and young, immature fibrous bone), rapidly enveloping the ends of the bone, with a resulting gradual increase in stability of the fracture fragments. Cartilage thus formed will eventually be resorbed by a process that is indistinguishable except for its lack of organization from endochondral bone formation. Bone will be formed by those cells having an adequate oxygen supply and subjected to the relevant mechanical stimuli.
  • Early in the repair process, cartilage formation predominates and glycosaminoglycans are found in high concentrations. Later, bone formation is more obvious. As this phase of repair takes place, the bone ends gradually become enveloped in a mass of callus containing increasing amounts of bone. In the middle of the reparative phase the remodeling phase begins, with resorption of portions of the callus and the laying down of trabecular bone along lines of stress. Finally, exercise increases the rate of bone repair. Heikkinen et al. Scand J. Clin. Lab. Invest. 25(suppl 113):32. In situ hybridization results have shown that OCP expression is confined to very specific regions where bone and cartilage formation is initiated.
  • In order to find out if OCP expression is induced in an animal model of bone fracture healing, a standard midshaft fracture was created in rat femur by means of a blunt guillotine, driven by a dropped weight. Bonnarens et al. (1984) Orthop. Res. 2:97-101. One, 2, 3 and 4 week-fractured bones were excised, fixed in buffered formalin, decalcified in EDTA solution and embedded in paraffin. All sections were hybridized with the OCP probe. The in-situ hybridization results show that a strong hybridization signal was apparent during the first and second weeks of fracture healing in the highly vascularized areas of the connective tissue within the callus (FIGS. 16-18), the endosteum (FIG. 19), the woven bone (FIG. 20) and the periosteum FIG. 21). The periosteum is regarded as a source of undifferentiated progenitors participating in callus formation at the site of bone fracture. The hybridization signal disappeared slowly during further differentiation stages of fracture healing (three and four weeks) and was retained only in the vascularized connective tissue. 22 displays brightfield (left) and darkfield (right) photomicrographs of a section of fractured bone healed for 4 weeks. In these later healing stages, the mature callus tissue was found to be comprised mainly by cancellous bone undergoing remodeling into compact bone, with little if any cartilage or woven bone present. The volume of the vascularized periosteal tissue is decreased but multiple cells in the periosteal tissue area of active remodeling of the cancellous bone covering the callus, show hybridization signal. This tissue covers the center of the callus and is also entrapped within the bone. See FIGS. 22 and 23. The box in FIG. 22 is enlarged in FIG. 23. As in the earlier stages, no hybridization signal was found in chondrocytes and osteoblasts. FIGS. 17 and 23. Several OCP expressing cells are concentrated in the vascular tissue that fills the cavities resulting from osteoclast activity (marked by asterisks).
  • Fractures in the young heal rapidly, while adult bone fractures heal slowly. The cause is a slower recruitment of specific chondro-/osteo-progenitors for the reparative process in the adult bone. Denervation retards fracture healing by diminishing the stress across the fracture site, while mechanical stress increases the rate of repair probably by increasing the proliferation and differentiation of specific bone progenitor cells and as a result, accelerates the rate of bone formation. The above results confirm our conclusions (see also hereunder) that OCP is most probably involved in induction of cortical and trabecular bone formation and remodeling, endochondral bone growth during development, and bone repair processes. In addition, there is strong evidence that OCP expression is tightly regulated, and induced during the earliest stages of bone fracture repair when osteo-/chondro-progenitor cells are recruited. This observation suggests that OCP plays a role in this process.
  • Taking into account the pattern of 608 expression during the process of bone fracture healing, it seems a reasonable hypothesis that 608-positive precursor cells are involved not only in remodeling of intact bone but also in the repair processes of the fractured bone as well.
  • Example 8 OCP Transcriptional Regulation
  • In order to clone the longest possible fragment which will contain the OCP regulatory region/s, bacs L4 and H7 were restricted with three different enzymes: BamHI, Bgl II and SauIIIA. The resulting fragments were cloned into the BamHI site of pKS. Ligation mixes were transformed into bacteria (E. coli-DH5α) and 1720 colonies were plated onto nitrocellulose filters which were screened with 32P-labeled PCR fragment spanning the mouse-OCP-exonl. Positive colonies were isolated.
  • Two identical clones, 14C10 and 15E11, contained the largest inserts (BamHI-derived ˜13 Kb inserts). The 14C10 clone is longer than the OCP “Lexicon” clone by ˜8 Kb at the 5′ end.
  • a. Cloning of Mouse OCP Promoter and UTR Upstream to the Reporter Gene-EGFP
  • The 1.4 Kb genomic region of the mouse OCP gene, flanked by BamHI site (nuc 5098 of the “Lexicon” clone which is the start site of clone p14C10) and the first ATG codon (first nucleotide of exon 2), was synthesized by genomic PCR using the “Lexicon” clone as template and pre-designed primers: 5′primer (For1) located upstream to the BamHI site (nucleotides 4587-4611 of the Lexicon clone) and 3′ primer (Rev 2) located immediately upstream to the first ATG (nucleotides 6560-6540 of the Lexicon clone) and tailed by a NotI site. The PCR product was cut by BamHI and NotI and the resulting 1.4 Kb fragment was ligated to pMCSIE into BamHI/NotI sites upstream to the EGFP reporter gene. The resulting clone was designated pMCSIEm608prm1.4.
  • Clone p14C10 was cut by XbaI and BamHI and the excised 4.088 Kb fragment was ligated into the BamHI and XbaI sites of pMCSIEm608prm1.4, upstream to the 1.4 Kb insert. The resulting clone was designated pMCSIEm608prm5.5 and contains 5552 nucleotides of the mouse 608 promoter and UTR upstream to EGFP. The insert of pMCSIEm608prm5.5 clone was completely sequenced.
  • The whole 13 Kb insert of p14C10 was excised by BamHI and ligated upstream to the 1.4 Kb insert of pMCSIEm608prm1.4 into the BamHI site. The resulting construct, pMCSIEm608prm14.5 contains a 14.5 Kb fragment of the mouse-OCP promoter and UTR upstream to EGFP.
  • b. Cloning Mouse OCP Promoter and UTR Upstream to the Reporter Gene-Luciferase
  • Both inserts of pMCSIEm608prm5.5 and of pMCSIEm608prm14.5 were also cloned upstream to luciferase, in Promega's pGL3-Basic vector. The 5.5 Kb insert of pMCSIEm608prm5.5 was excised by EcoRV and XbaI and ligated to SmaI and NheI sites of pGL3-Basic vector. The resulting clone is designated pGL3basicm608prm5.5.
  • Plasmid pMCSIEm608prm14.5 was restricted by NotI and the cohesive ends of the linearized plasmid were filled and turned into blunt ends. The 14.5 Kb insert was then excised by cutting the linear plasmid by SalI. The purified 14.5 Kb fragment was ligated to the XhoI and HindIII (filled in) sites of pGL3-basic upstream to the luciferase gene to create the construct designated pGL3basicm608prm14.5. SEQ ID NO:18 depicts 4610 bp that have been sequenced.
  • c. Analysis of TF Binding DNA Elements Common to Mouse and Human OCP
  • Known transcription factor (TF) binding DNA elements were analyzed for similarity upstream of human and mouse OCP ATG using the DiAlign program of Genomatix GmbH. The genomic pieces used are the proprietary mouse genomic OCP and reverse complement of AC024886 92001 to 11 1090. The locations of the ATG in these DNA pieces are:
      • 575 on rat cDNA
      • *6521 on mouse genomic
      • *3381 on the piece extracted from human genomic DNA AC0024886
        14 elements were extracted in this procedure and analyzed for transcription binding motifs using the MatInspector.
  • Some of the main “master gene” binding sites are the osteoblast-/chondrocyte-specific Cbfa1 factor; the chondrocyte-specific SOX 9 factor; the myoblast-specific Myo-D and Myo-F factors; the brain- and bone-specific WT1; Egr 3 and Egr 2 factors (Egr superfamily); the vitamin D-responsive (VDR) factor; the adipocyte-specific PPAR factor; and the ubiquitous activator SP1.
  • Example 9 Expression Pattern and Regulation of Gene 608: Expression of Gene 608 in Regard to Other Osteogenic Lineage Markers
  • Expression of gene 608 was tested in primary cells and in cell lines with regard to expression of various markers of osteogenic and chondrogenic lineages. The results of this analysis are summarized in the following table and showed that expression of 608 is restricted to committed early osteoprogenitor cells.
  • Cells 608 Collagen I Collagen II Alk. Phos. Osteocalcin Cbfa1 Osteopontin
    STO + + + +
    (fibroblasts)
    ROS + + +/− +
    (osteosarcoma)
    MC3T3 (pre- + + + + +
    osteoblasts)
    C2C12(pre- + +
    myoblasts)
    C6 (glioma)
    Calvaria mouse + +
    Calvaria rat + +
    C3H10T1/2 + + +
    (mesenchymal
    stem cells)
  • Example 10 OCP Expression is Mechanically Induced in MC3T3 E1 Cells
  • OCP transcription was detected by RT-PCR in mouse calvaria cells, U20S cells (human osteosarcoma cell line), and human embryonal bone. FIG. 14. OCP was initially discovered as being upregulated during mechanical stress in calvaria cells. In the present invention, we demonstrate that the influence of mechanical stress on OCP expression can be reproduced in another cell system using a different type of mechanical stimulation. In serum-deprived MC3T3-E1 pre-osteoblastic cells, mechanical stimulation caused by mild (287×g) centrifugation markedly induced OCP mRNA accumulation. FIG. 15. Other osteoblastic marker genes (osteopontin, ALP (staining—not shown) and Cbfa1) were transcriptionally augmented by this procedure. FIG. 15. The RT-PCR product of a non-osteoblastic marker gene (GAP-DH) was used as a control to compare RNA levels between samples. No increased expression was noticed when the latter primers were used. No expression was detected in non-osteoblastic cells (FIG. 14), suggesting that OCP expression is specifically induced in osteogenesis. Responsiveness of CMF608 expression to mechanical stimulation was confirmed by Northern blot analysis using polyA RNA from primary rat calvaria cells before and after mechanical stress (FIG. 12.).
  • Example 11 OCP Induction During Endochondral Growth—In Situ Hybridization Analysis
  • Our previous results demonstrated that OCP is expressed during adult mice bone modeling and remodeling. The expression was restricted to the following regions:
    • 1 perichondrium
    • 2 periosteum
    • 3 active remodeling and modeling regions
    • 4 perivascular connective tissue
    • 5 articular cartilage covering cells
    • 6 embryo-condensed mesenchymal cells-head, vertebrae and trunk
    • 7 ectopic bone formation
  • No previous observations suggest any role for OCP in bone development or initiation of endochondral ossification (longitudinal growth of long bones). Thus, the expression pattern of OCP by in situ hybridization on sections of bones from 1 week old mice was analyzed. At this stage of bone development, osteogenesis starts within the epiphysis (secondary ossification center). The hind limb skeleton of 1 week old rat pups (femur together with tibia) was fixed in buffered formalin and longitudinal sections of decalcified tissue were processed for in situ hybridization according to standard in-house protocol. Autoradiographs were developed, stained with hematoxylin-eosin and studied under microscope using brightfield and darkfield illumination.
  • A strong fluorescence signal was observed all over the second ossification center using OCP probes. FIG. 17 In addition, the hybridization signal delineates periosteal and perichondrial tissue in a way similar to that found earlier in adult bones. Surrounding mature chondrocytes displayed no signal. A very faint signal was observed using the osteocalcin probe which is a marker of mature osteoblasts.
  • In conclusion, OCP is expressed in osteoprogenitor cells that initiate endochondral ossification during bone development.
  • Example 12 In vivo Regulation by Stimuli Either Promoting or Suppressing Bone Formation Estrogen Administration, Blood Loss and Sciatic Neurotomy
  • Osteogenic cells are believed to derive from precursor cells present in the marrow stroma and along the bone surface. Blood loss, a condition that stimulates hemopoietic stem cells, activates osteoprogenitor cells in the bone marrow and initiates a systemic osteogenic response. High-dose estrogen administration also increases de novo medullary bone formation possibly via stimulation of generation of osteoblasts from bone marrow osteoprogenitor cells. In contrast, skeletal unweighting, whether due to space-flight, prolonged bed-rest, paralysis or cast immobilization leads to bone loss in humans and laboratory animal models. To detect alteration in OCP expression pattern following the above procedures, the following experiments were performed on two month old mice:
      • estrogen administration (500 μg/animal/week),
      • bleeding (withdrawing approximately 1.6% body weight),
      • unilateral (right limb) sciatic neurotomy,
      • control groups for each treatment
  • Total RNA was extracted from long bones after two-day treatment and RT-PCR using OCP-specific primers was performed. The results demonstrate that OCP expression was highly enhanced following blood loss and estro gen administration, while down-regulation was observed following sciatic neurotomy. FIG. 19.
  • By having a unique cell marker (OCP) we can show that the above procedures induce or reduce bone formation by increasing ordecreasing the number of osteoprogenitor cells. The above results suggest once more that OCP is a major member of a group of “bone specific genes” that regulate the accumulation of bone specific precursor cells.
  • Example 13 OCP Induction During Osteoblastic Differentiation of Bone Marrow Stroma Cells
  • Bone formation should be augmented in trabecular bone and cortical bone in osteoporotic patients. We have previously detected OCP expression in periosteum and endosteum (surrounding the cortical bone) but no signal was apparent in bone marrow cells. The latter cells normally differentiate to mature osteoblasts embedded in the trabecular and cortical bone matrix.
  • To further assess OCP expression in bone marrow progenitor cells, the inventors extracted total RNA from mouse and rat bone marrow immediately after obtaining it and after cultivation for up to 15 days in culture. No OCP-specific RT-PCR product was detected with RNA from freshly obtained bone marrow (both in adherent and non-adherent) cells. However, a faint signal was found after 5 days in culture, and it was further enhanced when RNA from cells grown for 15 days in culture was used. ALP (alkaline phosphatase) expression (an osteoblastic marker) was also found to be enhanced after 15 days. At both time points, adherent and non-adherent cells were reseeded, and RNA extractions were prepared 5 and 15 days later. A stronger RT-PCR product was observed with RNA extracted from originally adherent cells, suggesting the existence of less mature progenitors in the non-adherent population of bone marrow cells. The RT-PCR product of a non-osteoblastic marker gene (GAP-DH) was used as a control to compare RNA levels between samples.
  • In conclusion, bone marrow progenitor cells do not express OCP, but differentiate to more committed cells that do express this gene.
  • Example 14 OCP Role in Osteogenesis In Vitro
  • The ultimate test for the role of OCP as a crucial factor that induces osteoblast-related genes is its ability to up-regulate these genes in pre-osteoblastic and osteoblastic cells. Stable transfection of OCP to ROS 17/2.8 (differentiating osteoblast cell line) cells upregulated ALP and BSP expression. FIG. 24 In addition, marked increase in osteoblastic proliferation was observed; see FIG. 25
  • C3H10T1/2 cells were transfected with the following constructs containing the CMV promoter:
  • 1. 608-663 a.a—Construct containing 5′ untranslated region of β-actin, the OCP coding region from ATG at position 1 to the amino acid at position 663 of FIG. 3 (SEQ ID NO:2) and 3′Flag Tag. The functional portion of the mammalian OCP expressed using this construct contains the first 663 amino acids of the OCP polypeptide sequence, plus several additional amino acids of the 3′Flag tag
  • An additional construct was made, designated pCm-H608-663Nterm, which has the 5′ untranslated region of β-actin, the human OCP coding region from which encodes polypeptide from the ATG at position 1 to the amino acid at position 663 of FIG. 30 (SEQ ID NO:24) but no Flag Tag; this construct was deposited in the ATCC on Aug. 14, 2001 under ATCC Number PTA-3638.
  • 2. pCMV-neo—as negative control. This is the empty plasmid into which the 608-663aa was cloned to create vector #1 above. It serves as negative control to show that the effects are not caused by any other part of the #1 construct but by expression of the 608-663aa.
  • Example 15 Creation of a Readout System
  • A readout system is created to identify small molecules that can either activate or inactivate the OCP bone-precursor-specific promoter
  • Example 16 Bioinformatic Analysis of Human 608
  • A DNA sequence encoding a fragment of human OCP named AC024886 is found in htgs database but not in nt. There is no genomic DNA corresponding to the rat cDNA. Alignment of AC024886 against the rat cDNA using BLAST shows two areas of long alignment (and several shorter areas):
  • 1. cDNA: 6462-8186
      • Genomic: 89228-90952
      • plus/plus orientation: 81% identity
  • 2. cDNA: 5581-6451
      • Genomic: 107710-106840
      • Plus/minus orientation: 80% identity
  • Thus AC024886 was wrongly assembled in the region upstream of position 6462 (according to the rat cDNA), it was in the incorrect orientation. Using the incorrect orientation provided incorrect coding sequence and does not yield the human OCP protein.
  • The Genbank report on AC024886 was as follows:
  • LOCUS AC024886 175319 bp DNA HTG 06-SEP-2000
  • DEFINITION Homo sapiens chromosome 3 clone RP11-25K24, WORKING DRAFT
    SEQUENCE, 9 unordered pieces.
  • ACCESSION AC024886 VERSION AC024886.10 GI:9438330 KEYWORDS HTG; HTGS_PHASE1; HTGS_DRAFT.
  • SOURCE human.
    NOTE: This was a ‘working draft’ sequence. It consisted of 9 contigs. The true order of the pieces was not known and their order in this sequence record was arbitrary. Gaps between the contigs are represented as runs of N, but the exact sizes of the gaps was unknown.
      • 1 62523: contig of 62523 bp in length
      • 62524 62623: gap of unknown length
      • 62624 85445: contig of 22822 bp in length
      • 85446 85545: gap of unknown length
      • 85546 106059: contig of 20514 bp in length
      • 106060 106159: gap of unknown length
      • 106160 127908: contig of 21749 bp in length
      • 127909 128008: gap of unknown length
      • 128009 143068: contig of 15060 bp in length
      • 143069 143168: gap of unknown length
      • 143169 158734: contig of 15566 bp in length
      • 158735 158834: gap of unknown length
      • 158835 170042: contig of 11208 bp in length
      • 170043 170142: gap of unknown length
      • 170143 173715: contig of 3573 bp in length
      • 173716 173815: gap of unknown length
      • 173816 175319: contig of 1504 bp in length.
        a. Mapping Human Genomic 608 Exons
  • Ten exons were mapped on the rat cDNA sequence from base 107 to 6451. Thus the first exon on the human genomic piece may be lacking. The human genomic piece (AC024886) upstream (19090 bases) of base 6462 of cDNA (reverse complement from base of AC024886 92001 to 111090) was compared with the rat cDNA using the program ExonMapper of Genomatix. In the Table, base 1 is actually 1131 in the genomic piece used so that the actual genomic location starts at 91870.
  • Two additional exons were mapped on the rat cDNA sequence from base 6462 to 8883. Thus bases 6452-6461 are lacking. The human genomic piece used is from base 165,337 to 17,5667 (10,331 bases). The same type of program was used to compare this sequence to the genomic mouse 608 sequence deduced as described above.
  • Connecting the exon/intron borders from the genomic sequences yielded the predicted human and mouse cDNAs. The mouse and human predicted cDNAs were modified in order to allow frame shifts that allow a good multiple alignment of the human, mouse and rat proteins. Alignment was done using CLUSTALX and Pretty.
  • The cDNA modifications after the alignment of human cDNA to rat cDNA by GeneWise were as follows. In the following two tables,-x indicates a deletion of nucleotide x in the cDNA sequence; +x indicates an insertion of nucleotide x in the cDNA sequence; and all changed positions are in relation to the original sequence
  • Position Change
    1111 −g
    4154 −c
    4538 +g
    4730 −a
    4744-5 aa
    4830 +c
    4852 −g
    4902 +t
    4942 +c
    5370 +t
    5387 −a
    5395 +c
  • The corrections of frame-shifts in mouse 608 were as follows:
  • Position Change
    678 −c
    1106 −a
  • Chromosomal Location on the Human Chromosome:
  • Two different types of data exist.
      • a. Genomic piece AC024886 has identity to the fragment identified as ACCESSION D14436 as described by Fukui et al. (1994) Biochem. Biophys. Res. Commun. 201:894-901.
      • Alignment information:
        • Identities=315/335 (94%),
        • hrh1: 4-338
        • AC024886: 41662-41328
      • Hrh1 is mapped to chromosome 3 and to 3p25; and
      • b. Identity to STS at 3q. STS: 20-432 is identified as ACCESSION G54370 and described by Joensuu et al. (2000) Genomics 63:409-416.
    Example 17 Polyclonal Antibody Preparation
  • Polyclonal antibodies specific to the whole 608 putative protein are prepared by methods well-known in the art (the structure of 608 resembles that of growth factor precursors). Polyclonal antibodies are identified and the recombinant active form of 608 is prepared. The activities of the polyclonal antibodies are tested in vivo in mice. The antibodies can be used for the identification of the active form of this protein which is likely to constitute a fraction of the 608 protein.
  • Example 18 Stretch of Basic Amino Acids Found at the Boundary of the Rat and Human 608 Proteins, and its Implications
  • The homology between the rat and human N-terminal portions of the 608 protein is especially significant within the first 250 amino acids. At the boundary of this conserved region there is a completely conserved stretch of basic amino acids: KCKKDR (aa 242-247 and 240-245, in rat and human proteins, respectively). Stretches of basic amino acids frequently serve as protease cleavage sites. The fact that such a stretch is found on the boundary of more or less conserved sequences and the fact that it occurs within the C-terminal LRR, a generally conserved domain, suggests an underlying biological significance.
  • Accordingly, the 608 protein may undergo post-translational processing through the cleavage of its highly conserved N-terminal portion and this portion may be an active part of the 608 protein or possess at least part of its biological activities. Since the resulting 25 kD protein preserves the signal peptide, it would be secreted.
  • The biologically active 25 kD N-terminal cleavage product of 608 can thus be used for treatment and/or prevention of osteoporosis, fracture healing, bone elongation and periodontosis. As an indirect product (inhibition by either chemicals or by neutralizing mAbs), the fragment can be used for treatment and/or prevention of osteoarthritis, osteopetrosis, and osteosclerosis.
  • Example 19 The Adlican Protein and Gene
  • Adlican is a recently described protein. Crowl and Luk (2000) Arthritis Biol. Res. Adlican, a proteoglycan, was derived from placenta. The full amino acid sequence of Adlican is disclosed and identified as AF245505.1:1.8487, and is hereby incorporated by reference into this application; see FIG. 27.
  • The structure of Adlican was analyzed using methods described herein and found to have leucine-rich repeats and immunoglobulin regions similar to those of the OCP protein. The overall homology found between the amino acid residues of the indicated regions in the two proteins, is as follows:
  • OCP Adlican %
     1-661  1-669 38.4
    662-1629 670-1865 19.7
    1630-2587  1866-2828  46.5
     1-2587  1-2828 33.2
  • The invention therefor encompasses the use of Adlican in any manner described herein for the OCP protein. These functions and uses have not been disclosed previously for Adlican. They include use of Adlican, or a functional portion thereof, for preventing, treating or controlling osteoporosis, or for fracture healing, bone elongation or treatment of osteopenia, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving mechanical stress or lack thereof in a subject.
  • The Adlican gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene. Compositions comprising the Adlican gene, Adlican or antibodies specific for Adlican and physiologically acceptable excipients are likewise encompassed by the invention. Such excipients are known in the art and include saline, phosphate buffered saline and Ringer's solutions.
  • Example 20 Sequencing of the N-Terminal of the OCP Gene
  • Sequencing of the N-terminal fragment of the OCP gene using the 663 amino acid human construct (Example 14) added six additional nucleotides to the DNA sequence as shown in FIG. 29 (SEQ ID NO:23), where these 6 additional nucleotides are underlined.
  • The corresponding amino acid sequence of the encoded OCP protein thus has an additional two amino acids, as shown in FIG. 30,(SEQ ID NO:24) where these 2 additional amino acids are underlined.
  • Example 21 Preparation of a Recombinant Functional Portion of OCP
  • The 663 amino acid construct described in Example 14 was expressed in 293T cells. Western blot analysis of the medium, using antibody to the Flag tag, showed the presence of the 663 amino acid polypeptide. This polypeptide was purified from the medium, using a column of anti-Flag tag antibodies.
  • Our objective was to determine if the 1-663 amino acid polypeptide fragment of the 608 protein could induce proliferation in bone-related cell lines. Proliferation activity was tested by 3[H] thymidine incorporation assay on 4 bone related cell lines, with IGF1 or PTH as standards. (Pre-osteoblastic and osteoblastic proliferation is an activity that characterizes bone formation inducing factors such as IGF1 and PTH.)
  • In this key series of experiments, the purified 663 polypeptide showed a proliferative effect on W-20-17, a mouse bone marrow stromal cell line. This effect was reproduced with two 663-polypeptide batches in 5 independent experiments.
  • The activity of proliferation of bone marrow stromal cells demonstrated in the above experiments could be indicative of pre-osteoblastic proliferation activity induced by the 663 amino acid polypeptide. The 663 polypeptide activity could be mimicking the complete 608 protein in vivo activity. Alternatively, the 663 polypeptide activity could have a dominant negative effect, i.e. an effect that inhibits the whole 608 protein in vivo activity. Regardless of the mechanism, the 663 polypeptide could be used to induce proliferation of pre-osteoblastic stromal cells. This activity could help restore the pre-osteoblastic cell population that is known to be depleted in old-age or senile osteoporosis.
  • Example 22 Identification of RGD and Subtilisin-Like Proprotein Convertase (SPC) Motifs in Rat OCP
  • SEQ ID NO: 2 and SEQ ID NO: 34 depict the amino acid sequence of the rat 608 polypeptide. There is an RGD sequence at positions 729-731, and there is a putative cleavage motif subtilisin-like proprotein convertase (SPC) consensus sequence at positions 735-741.
  • The 608 protein was partially cleaved by SPC, in 293HEK cells. This putative peptide also contained the RGD sequence. Many adhesive proteins, present in extracellular matrices and in the blood, contain this tripeptide as their cell recognition site. Therefore, the 608 peptide comprising 1-741 amino acids, or a shorter fragment of the 608 protein containing the RGD sequence, may be a much more effective drug than the 663 amino acid fragment. The RGD and RxxRxxR (viz. R-aa1-aa2-R-aa3-aa4-R, i.e., SPC cleavage site) sequences are present in the human 608 protein sequence but are not present in Adlican or in Adlican-2.
  • Example 23 Natural Cleavage of Rat OCP
  • A polyclonal antibody against the rat 608 fragment comprising amino acid residues 1-312 was prepared by methods well-known in the art. This antibody was used to identify 608 peptides on Western blots. Several 608 sequences were expressed in cells derived from the transiently transfected 293T kidney cell line. The sequences were rat full length 608 polypeptide, rat 608 polypeptide fragment comprising amino acid residues 1-1634, and rat 608 polypeptide fragment comprising amino acid residues 1-663. The antibody identified a peptide of about 90 kDa in all three constructs produced. This peptide was detected by the anti-608 antibody in the conditioned medium of the cells, and not in cell extracts.
  • Table: Western blot analysis using polyclonal antibody to rat 608 fragment comprising 1-312 amino acid residues:
  • 608 amino acid sequence Expected size Detected size
    1-663  80 kDa 75-100 kDa
    1-1634 196 kDa 75-100 kDa
    (larger than 1-663 aa peptide)
    1-2597 (full length) 311 kDa 75-100 kDa
    (larger than 1-663 aa peptide)
    1-741  89 kDa
  • The 608 full length and the 608 1-1634 aa proteins produced in 293T cells were cleaved and secreted into the medium. The cleaved products appeared to be of identical size. The 608 1-663 aa protein was also secreted into the medium, but appeared to be slightly smaller than the cleaved full length and 1-1634 aa proteins. The expected size of the 608 fragment from 1-741 aa, that is, the putative SPC cleavage product, was approximately 89 kDa.
  • In further experiments, mouse calvaria cells cultured in vitro were analyzed by western blotting with the antibody to the 608 fragment 1-312aa. No 608 specific band was detected in cell extracts. In the conditioned medium from the cells a band of approximately 350 kDa was detected by the anti 608 1-312aa. The size of this band correlates with the protein size expected from the full length 608 protein. This analysis probably indicates that the 608 full-length protein is secreted.
  • To summarize, in human embryonic kidney cells, which do not normally express the 608 gene, overexpression of 608 protein results in secretion of a cleaved part of the 608 protein. In mouse calvaria cells, which normally express the 608 gene, the naturally expressed 608 protein is probably secreted uncleaved. One possible explanation of this data is that 608 activity is regulated by proteases that are selectively expressed.
  • Example 24 Human Adlican as a Candidate for Osteoblast Proliferation and Differentiation
  • As discussed in Example 19, Adlican is a recently described protein. The Adlican protein has LRR (Leucine-rich-repeats) and immunoglobulin regions highly similar to those of the OCP protein. The overall homology found between the amino acid residues of the indicated regions in the two human proteins is as shown in Example 19.
  • The deduced Adlican protein comprises the following features:
  • a. A cleavable, well-defined N-terminal signal peptide at 1-26 aa,
  • b. A LRR region (26-205 aa). This region can be divided into N-terminal and C-terminal domains of LRR (aa 26-59 and 217-276, respectively). Between them, there are six LRR (aa 55-77, 78-101, 102-125, 126-149, 150-173, 182-205).
  • c. Twelve immunoglobulin C-2 type repeats at amino acid positions 492-562, 590 658, 1866-1935, 1963-2032, 2060-2129, 2159-2228, 2256-2331, 2359-2425, 2457-2525, 2555-2623, 2650-2718, 2746-2817. Thus, two Ig-like repeats are found immediately downstream of a LRR region, while the remaining 10 repeats are clustered at the protein's C-terminus, as in OCP.
  • d. 4 nuclear putative localization domains (NLS) at amino acids: 676-682, 1146-1165, 1230-1236, and 1747-1763.
  • Therefore, we have determined that Adlican is a good candidate as an inducer of osteoblast proliferation and differentiation.
  • In order to determine if human Adlican expression causes proliferation and differentation of osteoblasts and chondrocytes, the expression product of Adlican, or cells or vectors expressing Adlican are monitored to determine if they cause cells to selectively proliferate and differentiate and thereby increase or alter bone density. Detecting levels of Adlican mRNA or expression and comparing it to “normal” non-osteopathic levels will allow screening amd detection of individuals who may be at risk for developing osteoporosis or lower levels of osteoblasts and chondrocytes.
  • Example 25 The Deduced Adlican-2 Protein
  • The deduced Adlican-2 protein (Genomic location: Yq11.21) was generated following the alignment (shown in FIG. 41) comparing Adlican-2 predicted sequences (FIGS. 39 and 40) and the equivalent human Adlican amino acid sequences (FIG. 27). This DNA molecule and the encoded polypeptide are novel molecules and constitute an integral part of this invention.
  • A Y chromosome BAC clone (gi 8748884) shows 93% homology to Human Adlican. Two mRNA sequences 100% homologous to this BAC clone were submitted to the gene bank (gi 14719942, and gi 14719940). However, the sequence of these clones is not based on cDNA sequences, but on human genomic data and they cover a short stretch of the nucleotide sequence in the C-terminal Ig region. We performed upstream nucleotide and deduced amino acid sequencing. The sequence alignment of Adlican and Adlican-2 exists along the entire Adlican sequence with one possible exception. Alignment along aa 66-215 of Adlican may be missing from the Adlican-2 molecule. This is the area of the 6 LRR (leucine-rich repeats). Although the encoding nt's for the 6 LRR region have not yet been observed, their existence has not been definitely ruled out.
  • The invention therefore encompasses the use of Adlican-2 in any manner described herein for the OCP protein. No functions or uses have been disclosed previously for Adlican-2. The proposed uses include use of Adlican-2, or a functional portion thereof, for preventing, treating or controlling osteoporosis, or of fracture healing, bone elongation or treatment of osteopenia, periodontosis, bone fractures or low bone density or other factors causing or contributing to osteoporosis or symptoms thereof or other conditions involving maechanical stress or lack thereof in a subject. As an indirect product (inhibition by either chemicals or by neutralizing mAbs), Adlican-2 can be used for treatment and/or prevention of osteoarthritis, osteopetrosis, and osteosclerosis. The Adlican-2 gene, or functional portions thereof, can likewise be used for any purpose described herein for an OCP gene. Compositions comprising the Adlican-2 gene, Adlican-2 or antibodies specific for Adlican-2 and physiologically acceptable excipients are likewise encompassed by the invention. Such excipients are known in the art and include saline, phosphate buffered saline and Ringer's solutions.
  • Example 26 The Physical Sequence of the Human OCP
  • Obtaining the sequence of human OCP was a difficult task. Initially several attempts were made to do amplification via RT-PCR using rat primers from the rat OCP coding sequence, in order to obtain human OCP cDNA, but these efforts failed. Thereafter, a predicted sequence was created as described in Examples 5 and 16 by bioinformatic analysis of the human genome. Then primers specifically designed according to the predicted sequence were used to amplify human cDNA. This proved difficult, due to the large size of the gene and also to the problem of the low abundancy of OCP mRNA in human tissue and the unavailabilty of such tissue. Eventually, cell line U20S (human osteosarcoma cell line) was found to be a suitable source for OCP mRNA. It was also decide to clone the DNA in fragments.
  • The first section of the gene to be cloned was a small fragment corresponding to the first 663 amino acids, creating the plasmid described in Example 14 (ATCC Number PTA-3638), and giving a corrected predicted sequence.
  • Since the complete human OCP gene could not be amplified and cloned as one entity, three large overlapping fragments were amplified, spanning the complete ORF. These PCR fragments were sequenced and the physical sequence of the human OCP was determined accordingly. The physical sequence was found to contain inserts relative to the predicted sequence. The overlapping PCR fragments were subsequently cloned in three separate plasmids (described below) as continous clones (overlapping regions were removed).
  • FIG. 42 shows the physical DNA sequence of the coding region-ORF of human OCP (SEQ ID NO: 31) having 7872 base pairs, including the stop codon. The sequence contains a silent mutation (C>T transition) at position 6729 compared to the predicted sequence of human OCP ORF. This transition does not change the identity of the encoded amino acid residue.
  • FIG. 43 shows the predicted amino acid sequence corresponding to the physical DNA sequence of the coding region-ORF of human OCP (SEQ ID NO:32), having 2623 amino acids.
  • The three plasmids harboring the 5′ fragment (A), middle fragment (B) and 3′ fragment (C) are depicted in FIGS. 34, 36 and 38 respectively, and were deposited on Nov. 21, 2001 under the terms of the Budapest Treaty with the American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, Va. 20108, USA, under ATCC accession numbers PTA-3878, PTA-3876 and PTA-3877 respectively.
  • FIG. 34 shows the physical sequence of the 5′ fragment (A) cloned into pBluescript KS to NotI (5′) and HindIII (3′) sites. Fragment A is comprised of the 5′ region (2440 bp) of the complete human OCP sequence and includes, in addition, at the 5′ end, 21 nucleotides of the β-actin “Kozak” region (nucleotides 9-29) followed by the ATG initiation coNotI (5′) and HindIII (3′) sites are located at nucleotides 1-8 and 2464-2469 respectively (SEQ ID NO:26).
  • FIG. 36 shows the physical sequence of the middle fragment (B) cloned into pBluescript KS to HindIII (5′) and SalI (3′) sites. Fragment B is comprised of the central region (3518 bp) of the complete human OCP sequence; the HindIII (5′) and SalI (3′) sites are located at nucleotides 1-6 and 3513-3518 respectively (SEQ ID NO:27).
  • FIG. 38 shows the physical sequence of the 3′ fragment (C) cloned into pMCS SV(A) to SalI (5′) and SpeI (3′) sites. Fragment C is comprised of the 3′ region (1923 bp, not including the 3 bp stop codon) of the complete human OCP sequence and includes at the 3′ end, 18 nucleotides coding for 6 Histidine residues, nucleotides 1924-1941, followed by the TGA stop codon.; the SalI (5′) and SpeI (3′) sites are located at nucleotides 1-6 and 1945-1950 respectively (SEQ ID NO:28).
  • Additionally, as discussed above, cloned fragment C contains a silent mutation (C>T transition) at nucleotide 783 compared to the predicted sequence of human OCP ORF; this transition does not change the identity of the encoded amino acid residue.
  • Note that if the number of OCP—encoding nucleotides in the three separate clones is added (viz., 2440+3518+1923), nine (9) more nucleotides are obtained than in the single complete sequence (7881 nucleotides vs 7872 nucleotides). This discrepancy is due to three reasons:
  • 1. The restriction site that appears at the 3′ end of fragment A and at the 5′ end of fragment B is counted twice, once in each fragment, giving an extra 6 nucleotides
  • 2. The restriction site that appears at the 3′ end of fragment B and at the 5′ end of fragment C is counted twice, once in each fragment, giving an extra 6 nucleotides
  • 3. The sequence of fragment C does not include the 3 nucleotide stop codon at the 3′ end, since it is interrupted by 18 nucleotides coding for 6 Histidine residues.
  • Therefore the difference is 6+6−3=9, which exactly explains the discrepancy mentioned above.
  • Example 27 608 Knockout Bone Phenotypes in Females with and without Ovariectomy
  • Introduction
  • Knockout (KO) mice deleted of the 608 gene were prepared by the method of Wattler et. al. BioTechniques 26:1150-1160, 1999. Comparison of 608 knockout (KO) mice to age, sex, and treatment matched wild type (WT) mice was performed to test the effect of 608 absence on bone parameters. Bone parameters of KO and WT were compared in untreated 3 and 4 months old females. KO and WT bone parameters were also compared in 3 months old female mice 5 weeks post ovariectomy (post-menopausal osteoporosis model).
  • The bone-related phenotypes were evaluated using two analyses: Peripheral Quantitative Computed Tomography (PQCT) of femur and tibia Rosen H N et. Al. Calcif. Tissue Int. 57:35-39, 1995) and serum Alkaline phosphatase (ALP) (Farley J R et. al. J Bone Miner Res 9:497-508, 1994. pQCT scanning is a 2D X-ray analysis that measures bone mineral density (BMD), bone mineral content (BMC), and bone geometric parameters. Serum ALP is a biochemical marker of bone formation.
  • Results
  • A. Untreated KO Females
  • It was found that the serum marker (serum ALP) of bone formation was significantly increased in 3 month old 608 KO mice. These results are depicted in FIG. 46.
  • pQCT scanning was performed for two groups of mice. pQCT of 3 months old untreated (sham operated) female mice gave parameters that were significantly different between WT and KO by two-way ANOVA analysis (pvalue<0.05), as shown in the Table below.
  • pQCT WT KO %
    Parameter Average Average P Value Increase
    Femur 3.572 4.070 P < 0.05 14
    Metaphysis
    Total Area
    Tibia 0.296 0.314 P < 0.05 6.1
    Diaphysis
    Cortical
    Thickness
    Femur 0.990 1.114 P < 0.05 12.5
    Diaphysis
    Cortical Area
  • Similarly, pQCT of 4 months old untreated female mice also gave parameters that were significantly different between WT and KO by one-way ANOVA analysis (pvalue<0.05), as shown in the following Table.
  • pQCT WT KO %
    Parameter Average Average P Value Increase
    Tibia 833.4 911.6 0.001803 9.4
    metaphysis
    cortical BMD
    Femur 3.38 3.8 0.004296 12.4
    Metaphysis
    Total Area
    Tibia 1150.4 1192 0.015211 3.6
    Diaphysis
    Cortical BMD
    Femur 1202.4 1241.5 0.015951 3.3
    Diaphysis
    Cortical BMD
  • In summary, the bone related phenotype of untreated 608 KO females is as follows:
  • At 3 months old, serum ALP is significantly increased in 608 KO mice. This may indicate that bone metabolism is different due to lack of the 608 gene. At this age, the significant increases are in bone geometric parameters. Slightly larger bone diameter and increased cortical thickness could affect bone strength.
  • At 4 months old, there is also a significant increase in cortical BMD of both femur and tibia. The incidence of fracture is closely related to BMD. Patients who sustain fractures have significantly decreased BMD.
  • In all parameters that showed a significant difference, the 608KO values were higher compared to WT. This may implicate an inhibitory role for the 608 gene in bone metabolism.
  • B. Ovariectomized KO Females
  • None of the parameters that were significantly increased in untreated females showed differences in ovariectomized females. Loss of tibia metaphysis total BMD due to ovariectomy may be smaller in 608KO mice. However, this difference was not found significant. Increasing the number of animals in each group could improve the statistical results.
  • CONCLUSIONS
  • The bones of KO mice appeared to have some basic anatomical differences compared to the bones of WT mice. This observation is based on trends seen in parameters reflecting bone geometry, such as total slice area, periosteal circumference, cortical area and thickness. Compared to untreated WT mice, an increase in the femur metaphysis area was observed in KO animals, both in 4 month-old and 3 month-old mice. Distal femur total BMD was notably unaffected by genotype despite the differences in bone size. Similar increases in geometric parameters were noted in KO mice compared to WT at the femur diaphysis (cortical area) and at the tibia diaphysis (cortical thickness) in 3 month-old mice. At 4 months old there is also a significant increase in cortical BMD of both femur and tibia.
  • Consistent with these effects on bone mass, biochemical markers of bone turnover were increased in KO mice, relative to the WT controls, suggesting that bone metabolism is different. Parameters that could affect bone strength, such as a slightly larger bone diameter and increased cortical thickness, could contribute to bone strength.
  • The effects on bone mass and biochemical markers of bone turnover noted in the KO mice appear to be indicative of a protective effect of the KO phenotype on bone loss following ovariectomy, although the effects were small. A trend to a genotype-related prevention of bone loss in the distal femur metaphysis relative to the ovariectomized controls was observed in KO animals. A slight partial prevention of bone loss relative to the ovariectomized control group was observed in KO mice at the endocortical surface of femur and tibia metaphysis, although the effects were not marked.
  • In conclusion, the effects on bone mass and biochemical markers of bone turnover noted in the KO mice appear to be indicative of a protective effect of the KO genotype on bone loss following ovariectomy. Bone metabolism and bone geometry could be different in KO mice.
  • The following hypothesis may correlate the in vitro data in the previous Examples with this in vivo data from the KO mice analyses:
  • The function of the 608 protein could be to promote proliferation of the undifferentiated osteoprogenitor cell population. This hypothesis is based on the proliferative effect of the 608 1-663aa polypeptide on mouse bone marrow cell line, as shown in Example 21. In the absence of this protein the balance between proliferation and differentiation of osteoprogenitors is changed in favor of differentiation and therefore the increased bone parameters are obtained at a young age. It could be that in aged mice this change in balance causes a decrease in bone parameters due to the normal decrease in osteoprogenitors that occurres with aging. If this hypothesis is correct an intermittent administration of the 608 protein or fragments of it could be used as a treatment for osteoporosis. Administration of the 608 polypeptide would cause proliferation of osteoprogenitors. When 608 level is allowed to decrease to normal levels, differentiation could take place.
  • An example of timing of intermittent treatment may be daily e.g. daily administration, preferably by injection, preferably subcutaneous, as opposed to continuous administration e.g. by infusion. Other examples may be administration every other day, or every few days, or even once a week or once a month. In the case of parathyroid hormone (1-34 amino acid), daily subcutaneous injections of 20-40 μg were considered intermittent administration as opposed to continuous infusions; see Neer R. M. et. al. 2001, The New England Journal of Medicine. 344: 1434-1441, Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis.
  • Alternatively the 663aa fragment may act as an inhibitor of 608 function, as discussed in Example 21.

Claims (16)

1. An isolated polypeptide encoded by a nucleic acid molecule comprising consecutive nucleotides having a sequence set forth in SEQ ID NO:6, SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29 or SEQ ID NO:31 or comprising nucleotides having a sequence incorporated in plasmids deposited under ATCC Accession Nos. PTA-3638, PTA-3876, PTA-3877, PTA-3878 complements thereof or a polynucleotide having a sequence that differs due to the degeneracy of the genetic code from SEQ ID NO:6, SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:31 or a sequence incorporated in plasmids deposited under ATCC Accession Nos. PTA-3638, PTA-3876, PTA-3877 or PTA-3878, or a sequence which hybridizes under stringent conditions to SEQ ID NO:6, SEQ ID NO: 20, SEQ ID NO: 23, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29 or SEQ ID NO:31 or to a sequence incorporated in plasmids deposited under ATCC Accession Nos. PTA-3638, PTA-3876, PTA-3877 or PTA-3878 or a functional portion thereof or a polynucleotide which is at least substantially homologous thereto.
2. The isolated polypeptide of claim 1, wherein the polypeptide is identified as human protein 608, or a functional portion of protein 608 or Adlican-2, or a polypeptide which is at least substantially homologous thereto.
3. The isolated polypeptide of claim 2, wherein the polypeptide comprises about 663 to about 1634 amino acids.
4. An antibody which specifically binds to a polypeptide of claim 2 or a functional portion thereof.
5. An isolated polypeptide wherein the functional portion comprises consecutive amino acids having a sequence set forth in SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 30 or SEQ ID NO: 32.
6. The isolated polypeptide of claim 5 wherein the sequence comprises about the first 663 amino acids of the sequence set forth in SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 30 or SEQ ID NO: 32.
7. The isolated polypeptide of claim 6 wherein the sequence comprises about the first 741 amino acids of the sequence set forth in SEQ ID NO:16, SEQ ID NO: 24, SEQ ID NO: 30 or SEQ ID NO: 32.
8. The isolated polypeptide of claim 5, wherein the polypeptide is identified as human 608 protein or human Adlican-2 protein or a functional portion thereof or a polypeptide which is at least substantially homologous thereto.
9. An isolated polypeptide of claim 5 comprising consecutive amino acids having a sequence set forth in SEQ ID NO:32, designated human 608 protein.
10. An isolated polypeptide of claim 5 comprising consecutive amino acids having a sequence set forth in SEQ ID NO: 30 designated Adlican-2 polypeptide.
11. An isolated polypeptide of claim 10 comprising consecutive amino acids having a sequence set forth in SEQ ID NO: 30 deleted of amino acids 6-215.
12. An antibody which binds specifically to a polypeptide of claim 9.
13. An antibody which binds specifically to a polypeptide of claim 10.
14. An antibody of claim 12, which does not bind specifically to a rat 608 polypeptide.
15. An antibody of claim 14, wherein the rat polypeptide has a sequence set forth in SEQ ID NO: 34.
16. A composition comprising the antibody of claim 14
US11/841,827 1999-05-14 2007-08-20 Genes associated with mechanical stress, expression products therefrom, and uses thereof Abandoned US20090258021A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/841,827 US20090258021A1 (en) 1999-05-14 2007-08-20 Genes associated with mechanical stress, expression products therefrom, and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US31221699A 1999-05-14 1999-05-14
PCT/US2001/046400 WO2002046364A2 (en) 2000-12-04 2001-12-04 Genes associated with mechanical stress, expression products therefrom, and uses thereof
US10/454,351 US7259253B2 (en) 1999-05-14 2003-06-04 Genes associated with mechanical stress, expression products therefrom, and uses thereof
US11/841,827 US20090258021A1 (en) 1999-05-14 2007-08-20 Genes associated with mechanical stress, expression products therefrom, and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/454,351 Division US7259253B2 (en) 1999-05-14 2003-06-04 Genes associated with mechanical stress, expression products therefrom, and uses thereof

Publications (1)

Publication Number Publication Date
US20090258021A1 true US20090258021A1 (en) 2009-10-15

Family

ID=46299369

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/454,351 Expired - Fee Related US7259253B2 (en) 1999-05-14 2003-06-04 Genes associated with mechanical stress, expression products therefrom, and uses thereof
US11/841,827 Abandoned US20090258021A1 (en) 1999-05-14 2007-08-20 Genes associated with mechanical stress, expression products therefrom, and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/454,351 Expired - Fee Related US7259253B2 (en) 1999-05-14 2003-06-04 Genes associated with mechanical stress, expression products therefrom, and uses thereof

Country Status (1)

Country Link
US (2) US7259253B2 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3388075T3 (en) 2015-03-27 2023-12-11 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against various tumors (seq id 25 - mrax5-003)
GB201505305D0 (en) 2015-03-27 2015-05-13 Immatics Biotechnologies Gmbh Novel Peptides and combination of peptides for use in immunotherapy against various tumors

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4912207A (en) * 1987-05-06 1990-03-27 Washington University DNA clone of human thrombomodulin and portions thereof
US5002876A (en) * 1986-09-22 1991-03-26 Phillips Petroleum Company Yeast production of human tumor necrosis factor
US5445941A (en) * 1993-06-21 1995-08-29 Eli Lilly And Company Method for screening anti-osteoporosis agents
US5599708A (en) * 1991-07-23 1997-02-04 Osteosa Liquidation Trust Osteoclast growth regulating factors and antibodies
US5759781A (en) * 1995-12-22 1998-06-02 Yale University Multiparametric fluorescence in situ hybridization
US5763416A (en) * 1994-02-18 1998-06-09 The Regent Of The University Of Michigan Gene transfer into bone cells and tissues
US5861249A (en) * 1996-04-23 1999-01-19 Cold Spring Harbor Laboratory Assays and reagents for identifying modulators of cdc25-mediated mitotic activation
US5882925A (en) * 1995-02-10 1999-03-16 Millennium Pharmaceuticals, Inc. Compositions and method for the treatment and diagnosis of cardiovascular disease using rchd502 as a target
US6369027B1 (en) * 1995-12-22 2002-04-09 Amgen Inc. Osteoprotegerin

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU4659893A (en) * 1992-06-30 1994-01-24 Research Corporation Technologies, Inc. Trivalent synthesis of oligonucleotides containing stereospecific alkylphosphonates and arylphosphonates
EP1082463B1 (en) 1998-05-15 2007-01-03 Quark Biotech, Inc. Mechanical stress induced genes, expression products therefrom, and uses thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5002876A (en) * 1986-09-22 1991-03-26 Phillips Petroleum Company Yeast production of human tumor necrosis factor
US4912207A (en) * 1987-05-06 1990-03-27 Washington University DNA clone of human thrombomodulin and portions thereof
US5599708A (en) * 1991-07-23 1997-02-04 Osteosa Liquidation Trust Osteoclast growth regulating factors and antibodies
US5445941A (en) * 1993-06-21 1995-08-29 Eli Lilly And Company Method for screening anti-osteoporosis agents
US5763416A (en) * 1994-02-18 1998-06-09 The Regent Of The University Of Michigan Gene transfer into bone cells and tissues
US5882925A (en) * 1995-02-10 1999-03-16 Millennium Pharmaceuticals, Inc. Compositions and method for the treatment and diagnosis of cardiovascular disease using rchd502 as a target
US5759781A (en) * 1995-12-22 1998-06-02 Yale University Multiparametric fluorescence in situ hybridization
US6369027B1 (en) * 1995-12-22 2002-04-09 Amgen Inc. Osteoprotegerin
US5861249A (en) * 1996-04-23 1999-01-19 Cold Spring Harbor Laboratory Assays and reagents for identifying modulators of cdc25-mediated mitotic activation

Also Published As

Publication number Publication date
US7259253B2 (en) 2007-08-21
US20040053301A1 (en) 2004-03-18

Similar Documents

Publication Publication Date Title
WO2002046364A9 (en) Genes associated with mechanical stress, expression products therefrom, and uses thereof
AU2020270508B2 (en) C/EBP alpha short activating RNA compositions and methods of use
US6262333B1 (en) Human genes and gene expression products
KR102365486B1 (en) Modulation of prekallikrein (pkk) expression
EP1082463B1 (en) Mechanical stress induced genes, expression products therefrom, and uses thereof
US20020015950A1 (en) Atherosclerosis-associated genes
ES2397441T3 (en) Polynucleotide and polypeptide sequences involved in the bone remodeling process
US20030082233A1 (en) Method and composition for modulating bone growth
EP1370287A2 (en) Method and composition for modulating bone growth
KR20160027968A (en) Compositions and methods for modulating foxp3 expression
Xu et al. Cloning the full‐length cDNA for rat connective tissue growth factor: Implications for skeletal development
KR20060015296A (en) Compositions and methods for the diagnosis and treatment of tumors of glial origin
EP1019502A2 (en) Human orphan receptor ntr-1
KR20220139926A (en) artificial synapse
NZ529560A (en) Pharmaceutical compositions and methods of using secreted frizzled related protein
US20070243186A1 (en) ISLR gene and its association with osteoarthritis and other bone and cartilage disorders, expression products derived therefrom, and uses thereof
AU726918B2 (en) TGFbeta signal transduction proteins, genes, and uses related thereto
US20090258021A1 (en) Genes associated with mechanical stress, expression products therefrom, and uses thereof
US20220265798A1 (en) Cancer vaccine compositions and methods for using same to prevent and/or treat cancer
JP4763282B2 (en) How to control bone development
US20020022026A1 (en) Genes associated with mechanical stress, expression products therefrom, and uses thereof
US20030096272A1 (en) Genes regulated by peroxisome proliferator-activated receptor gamma agonist
US20020137705A1 (en) Genes associated with mechanical stress, expression products therefrom, and uses thereof
US6673570B1 (en) Smad associating polypeptides
US20020086825A1 (en) Genes associated with mechanical stress, expression products therefrom, and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: QUARK PHARMACEUTICALS, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:QUARK BIOTECH, INC.;REEL/FRAME:021630/0770

Effective date: 20070601

STCB Information on status: application discontinuation

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