AU758672B2 - Osteoprotegerin - Google Patents

Osteoprotegerin Download PDF

Info

Publication number
AU758672B2
AU758672B2 AU65400/99A AU6540099A AU758672B2 AU 758672 B2 AU758672 B2 AU 758672B2 AU 65400/99 A AU65400/99 A AU 65400/99A AU 6540099 A AU6540099 A AU 6540099A AU 758672 B2 AU758672 B2 AU 758672B2
Authority
AU
Australia
Prior art keywords
opg
seq
polypeptide
nucleic acid
cdna
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.)
Ceased
Application number
AU65400/99A
Other versions
AU6540099A (en
Inventor
William J Boyle
Frank J. Calzone
Ming-Shi Chang
David L. Lacey
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.)
Amgen Inc
Original Assignee
Amgen 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 AU14686/97A external-priority patent/AU710587B2/en
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to AU65400/99A priority Critical patent/AU758672B2/en
Publication of AU6540099A publication Critical patent/AU6540099A/en
Application granted granted Critical
Publication of AU758672B2 publication Critical patent/AU758672B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

-1-
AUSTRALIA
PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT
ORIGINAL
Name of Applicant/s: Amgen Inc.
n.
Actual inventor/s: William J. Boyle and Uavid L. Lacey and Frank J. Calzone and Ming- Shi Chang S. S Address for Service: BALDWIN SHELSTON WATERS MARGARET STREET SYDNEY NSW 2000 'OSTEOPROTEGERIN' Invention Title: Details of Original Application No. 14686/97 dated 20 DEC 1996 The following statement is a full description of this invention, including the best method of performing it known to me/us:- File: 26388AUP00 -la-
OSTEOPROTEGERIN
Field of the Invention The invention relates generally to polypeptides involved in the regulation of bone metabolism. More particularly, the invention relates to a novel polypeptide, termed osteoprotegerin, which is a member of the tumor necrosis factor receptor superfamily. The polypeptide is used to treat bone diseases characterized by increased bone loss such as osteoporosis.
Background of the Invention Any discussion of the prior art throughout the specification should in no way be considered as an admission that such prior art is widely known or forms part of common general knowledge in the field.
Polypeptide growth factors and cytokines are secreted factors which signal a wide variety 15 of changes in cell growth, differentiation, and metabolism, by specifically binding to discrete, surface bound receptors. As a class of proteins, receptors vary in their structure and mode of signal transduction. They are characterized by having an extracellular domain that is involved in ligand binding, and cytoplasmic domain which transmits an appropriate intracellular signal.
Receptor expression patterns ultimately determine which cells will respond to a given ligand, while the structure of a given receptor dictates the cellular response induced by ligand binding.
Receptors have been shown to transmit intracellular signals via their cytoplasmic domains by activating protein tyrosine, or protein serine/threonine phosphorylation platelet derived growth factor receptor (PDGFR) or transforming growth factor-b receptor-I (TGFbR-I), by stimulating G-protein activation b-adrenergic receptor), and by modulating associations with cytoplasmic signal transducing proteins TNFR-1 and Fas/APO) (Heldin, Cell 80, 213- 223 (1995)).
The tumor necrosis factor receptor (TNFR) superfamily is a group of type I transmembrane proteins which share a conserved cysteine-rich motif which is repeated three to six times in the extracellular domain (Smith, et al. Cell 76, 953-962 (1994)). Collectively, these repeat units form the ligand binding domains of these receptors (Chen et al., Chemistry 270, 2874-2878 (1995)). The ligands for these receptors are a structurally related group of proteins homologous to TNFa. (Goeddel et al. Cold Spring Harbor Symp. Quart. Biol. 51, 597-609 (1986); Nagata et al. Science 267, 1449-1456 (1995)). TNFa binds to distinct, but closely related receptors, TNFR-1 and TNFR-2. TNFa produces a variety of biological responses in z5TR~^>35 receptor bearing cells, including, TNFa produces a variety of biological responses in receptor bearing cells, including, proliferation, differentiation, and cytotoxicity and apoptosis (Beutler et al. Ann. Rev.
Biochem. 57, 505-518 (1988)).
TNFa is believed to mediate acute and chronic inflammatory responses (Beutler et al. Ann. Rev. Biochem. 57, 505-508 (1988)). Systemic delivery of TNFa induces toxic shock and widespread tissue necrosis. Because of this, TNFa may be responsible for the severe morbidity and mortality associated with a variety of infectious diseases, including sepsis. Mutations in FasL, the ligand for the TNFR-related receptor Fas/APO (Suda et al. Cell 75, 1169-1178 (1993)), is associated with autoimmunity (Fisher et al. Cell 81, 935-946 (1995)), while overproduction of FasL may be implicated in drug-induced hepatitis. Thus, ligands to the various TNFR-related proteins often mediate the serious effects of many disease states, which suggests that agents that neutralize the activity of these ligands would have therapeutic value. Soluble TNFR-1 receptors, and antibodies that bind TNFa, have been tested for their ability to neutralize systemic TNFa (Loetscher et al. Cancer Cells 221-226 (1991)). A naturally occurring form of a secreted TNFR-1 mRNA was recently cloned, and its product tested for its ability to neutralize TNFa activity i vitro and in vivo (Kohno et al. PNAS USA 82, 8331-8335 (1990)). The ability of this protein to neutralize TNFa suggests that soluble TNF receptors function to bind and clear TNF thereby blocking the cytotoxic effects on TNFR- bearing cells.
20 An object of the invention to identify new members of the TNFR super family. It is anticipated that new family members may be transmembrane proteins or soluble forms thereof comprising extracellular domains and lacking transmembrane and cytoplasmic domains. We have identified a new member of the TNFR superfamily which encodes a secreted protein that is closely related to TNFR-2. By analogy to soluble TNFR-1, the TNFR-2 related protein may negatively regulate the activity of its ligand, and thus may be useful in the treatment of certain human diseases.
Summary of the Invention A novel member of the tumor necrosis factor receptor (TNFR) superfamily has been identified from a fetal rat intestinal cDNA library. A full-length cDNA clone was obtained and sequenced. Expression of the rat cDNA in a transgenic mouse revealed a marked increase in bones density, particularly in long bones, pelvic bone and vertebrae.
-3- The invention provides for nucleic acids encoding a polypeptide having at least one of the biological activities of OPG. Nucleic acids which hybridize to nucleic acids encoding mouse, rat or human OPG as shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO: 122), and 9C-9D (SEQ ID NO: 124) are also provided. Preferably, OPG is mammalian OPG and more preferably is human OPG. Recombinant vectors and host cells expressing OPG are also encompassed as are methods of producing recombinant OPG. Antibodies or fragments thereof which specifically bind the polypeptide are also disclosed.
Methods of treating bone diseases are also provided by the invention. The polypeptides are useful for preventing bone resorption and may be used to treat any condition resulting in bone loss such as osteoporosis, hypercalcemia, Paget's disease of bone, and bone loss due to rheumatoid arthritis or osteomyelitis, and the like. Bone disease may also be treated with antisense or gene therapy using nucleic acids of the invention. Pharmaceutical compositions comprising OPG nucleic acids and polypeptides are also encompassed.
SAcording to a firt aspect, the present invention consists in an isolated nucleic acid encoding a polypeptide comprising at least one of the biological activities of OPG wherein the nucleic acid hybridises under stringent conditions with the complement of the polypeptideencoding regions as shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124); with the proviso that the nucleic acid does not have the sequence as set forth in SEQ ID NO:6 ofWO96/26217.
According to a second aspect, the present invention consists in a polypeptide encoded by the nucleic acid as described in the first aspect.
According to a third aspect, the invention provides a purified and isolated polypeptide encoded by the nucleic acid of the first aspect with the proviso that the polypeptide does not have the sequence as set forth in SEQ ID NO: 5 of WO96/26217.
According to a fourth aspect, the present invention provides an expression vector comprising the nucleic acid as described in the first aspect.
According to a fifth aspect, the present invention provides a transgenic mammal comprising the expression vector according to the fourth aspect.
According to the sixth aspect, the present invention provides a host cell transformed or transfected with the expression according to the fourth aspect.
According to the seventh aspect, the present invention provides a process for the production of the polypeptide according to the third aspect comprising: growing under suitable nutrient conditions host cells transformed or transfected with the Snucleic acid of the first aspect; and isolating the polypeptide products of the expression of the nucleic acids.
-3a- According to an eighth aspect, the present invention provides an antibody or fragment thereof which specifically binds to the polypeptide according to the second or third aspect.
According to a ninth aspect, the present invention provides a method for detecting the presence of the polypeptide of the second aspect in a biological sample comprising: incubating the sample with the antibody according to the eighth aspect under conditions that allow binding of the antibody to the polypeptide of the second aspect; and detecting the bound antibody.
According to a tenth aspect, the invention provides a method to assess the ability of a candidate substance to bind to the polypeptide of the second or third aspect comprising; incubating the polypeptide of the second or third aspect with the candidate substance under conditions that allow binding; and measuring the bound substances.
According to an eleventh aspect, the invention provides a method of regulating the levels of the polypeptide of the second aspect in an animal comprising modifying the animal with the nucleic acid according to the first aspect.
According to the twelfth aspect, the invention provides a pharmaceutical composition comprising a therapeutically effective amount of the polypeptide of the third aspect in a pharmaceutically acceptable carrier, adjuvant, solubilizer, stabiliser and/or anti-oxidant.
According to a thirteenth aspect, the invention provides a method of treating a bone disorder comprising administering a therapeutically effective amount of the polypeptide according to the third aspect.
According to a fourteenth aspect, the invention provides the use of a polypeptide of the third aspect for the preparation of a medicament for the treatment of a bone disorder.
Unless the context clearly requires otherwise, throughout the description and the claims, the words 'comprise', 'comprising', and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of "including, but not S. limited to".
Description of the Figures Figure 1. A. FASTA analysis of novel EST LORF. Shown is the deduced FRI-1 amino acid sequence aligned to the human TNFR-2 sequence. B. Profile analysis of the novel EST LORF shown is the deduced FRI-1 amino acid sequence aligned to the TNFR-profile. C.
Structural view of TNFR superfamily indicating region which is homologous to the novel FRI-1.
Figure 2. Structure and sequence of full length rat OPG gene, a novel member of the Ir -f R ,Z TNFR superfamily. A. Map of pMOB-B1.1 insert. Box indicates position of LORF within the cDNA sequence (bold line). Black box indicates signal peptide, and gray ellipses indicate -3b position of cysteine-rich repeat sequences. B, C. Nucleic acid and protein sequence of the Rat OPG cDNA. The predicted signal peptide is underlined, and potential sites of N-linked glycosylation are indicated in bold, underlined letters. D,E. Pileup sequence comparison (Wisconsin GCG Package, Version 8.1) of OPG with other members of the TNFR superfamily, fas (SEQ ID NO:128); tnfrl (SEQ ID NO: 129); sfu- *oo 4 t2 (SEQ ID NO:130); tnfr2 (SEQ ID NO:131); cd40 (SEQ ID NO:132); osteo (SEQ ID NO:133); ngfr (SEQ ID NO:134); ox40 (SEQ ID NO: 135); 41bb (SEQ ID NO:136).
Figure 3. PepPlot analysis (Wisconsin GCG Package, Version 8.1) of the predicted rat OPG protein sequence. A. Schematic representation of rat OPG showing hydrophobic (up) and hydrophilic (down) amino acids. Also shown are basic (up) and acidic (down) amino acids. B. Display of amino acid residues that are beta-sheet forming (up) and beta-sheet breaking down) as defined by Chou and Fasman (Adv. Enz.
47, 45-147 (1948)). C. Display of propensity measures for alpha-helix and beta-sheet (Chou and Fasman, ibid). Curves above 1.00 show propensity for alpha-helix or betasheet structure. Structure may terminate in regions of protein where curves drop below 1.00. D. Display of residues that are alpha-forming (up) or alpha-breaking (down). E.
S Display of portions of the protein sequence that resemble sequences typically found at the amino end of alpha and beta structures (Chou and Fasman, ibid). F. Display of portions of the protein sequence that resemble sequences typically found at the carboxyl end of alpha and beta structures (Chou and Fasman, ibid). G. Display of portions of the proteins sequence typically found in turns (Chou and Fasman, ibid) H. Display of the helical hydrophobic moment (Eisenberg et al. Proc. Natl. Acad. Sci. USA 81, 140-144 (1984)) at each position in the sequence. I. Display of average hydrophathy based upon Kyte and Doolittle Mol. Biol. 157, 105-132 (1982)) and Goldman et al. (reviewed in 20 Ann. Rev. Biophys. Biophys. Chem. 15, 321-353 (1986)).
SFigure 4. mRNA expression patterns for the OPG cDNA in human tissues.
Northern blots were probed with a 32P-labeled rat cDNA insert left two panels), or with the human cDNA insert right panel).
Figure 5. Creation oftransgenic mice expressing the OPG cDNA in hepatocytes.
Northern blot expression of HE-OPG transgene in mouse liver.
Figure 6. Increase in bone density in OPG transgenic mice. Panel A-F. Control Mice. G-J, OPG expressing mice. At necropsy, all animals were radiographed and photographs prepared. In A-F, the radiographs of the control animals and the one transgenic non-expressor are shown. Note that the bones have a clearly defined cortex and a lucent central marrow cavity. In contrast, the OPG animals have a poorly defined cortex and increased density in the marrow zone.
Figure 7. Increase in trabecular bone in OPG transgenic mice. A-D.
Representative photomicrographs of bones from control animals. In A and B, low (4X, power images of the femurs are shown (Masson Trichrome stain). Stains for tartrate resistant acid phosphatase (TRAP) demonstrate osteoclasts (see arrows) both resorbing cartilage and trabecular bone Note the flattened appearance of osteoclasts on trabecular bone. E-H. Representative photomicrographs of bones from OPG-expressing animals. In E and F, low (4X, 10X) power images of the femurs are shown (Masson Trichrome stain). The clear region is the growth plate cartilage, blue stained area is bone, and the red area is marrow. Note that in contrast to the controls, the trabecular bone has not been resorbed resulting in the absence of the usual marrow cavity. Also, the resulting trabeculae have a variegated appearance with blue and clear areas. The clear areas are remnants of growth plate cartilage that have never been remodelled. Based on TRAP stains, these animals do have osteoclasts (see arrows) at the growth plate which may be reduced in number. However, the surfaces of the trabeculae away from the growth plate are virtually devoid of osteoclasts a finding that stands in direct contrast with the control animals (see D).
Figure 8. HE-OPG expressors donot have a defect in monocyte-macrophage development. One cause for osteopetrosis in mice is defective M-CSF production due to a point mutation in the M-CSF gene. This results in a marked deficit of circulating and 2 0 tissue based macrophages. The peripheral blood of OPG expressors contained monocytes as assessed by HIE analysis. To affirm the presence of tissue macrophages, immnohistochemistry was performed using F480 antibodies, which recognize a cell surface antigen on murine macrophages. A and C show low power (4X) photomicrographs of the spleens from normal and CR1 overexpressors. Note that both animals have numerous F480 positive cells. Monocyte-macrophages were also present in the marrow of normal and HE-OPG overexpressors Figure 9. Structure and sequence of mouse and human OPG cDNA clones. A, B.
Mouse cDNA and protein sequence. C, D. Human cDNA and protein sequence. The predicted signal peptides are underlined, and potential sites of N-linked glycosylation are indicated in bold. E, F. Sequence alignment and comparison of rat, mouse and human OPG amino acid sequences.
Figure 10. Comparison of conserved sequences in extracellular domain of TNFR-1 and human OPG. PrettyPlot (Wisconsin GCG Package, Version 8.1) of the TNFR1 and OPG alignment described in example 6. Top line, human TNFR1 sequences encoding domains 1-4. Bottom line, human OPG sequences encoding domains 1-4.
Conserved residues are highlighted by rectangular boxes.
Figure 11. Three-dimensional representation of human OPG. Side-view of the Molescript display of the predicted 3-dimensional structure of human OPG residues through 163, (wide line), co-crystallized with human TNFb (thin line). As a reference for orientation, the bold arrows along the OPG polypeptide backbone are pointing in the N-terminal to C-terminal direction. The location of individual cysteine residue side chains are inserted along the polypeptide backbone to help demonstrate the separate cysteine-rich domains. The TNFb molecule is aligned as described by Banner et al.
(1993).
Figure 12. Structure of OPG cysteine-rich domains. Alignment of the human (top line SEQ ID NO:136)and mouse (bottom line) OPG amino acid sequences highlighting the predicted domain structure of OPG. The polypeptide is divided into two halves; the N-terminus and C-terminus The N-terminal half is predicted to contain four cysteine rich domains (labeled The predicted intrachain disulfide bonds are indicated by bold lines, labeled "SS1", "SS2", or "SS3". Tyrosine 28 and histidine S* 20 (underlined) are predicted to form an ionic interaction. Those amino acids predicted to interact with an OPG ligand are indicated by bold dots above the appropriate residue.
The cysteine residues located in the C-terminal half of OPG are indicated by rectangular boxes.
Figure 13. Expression and secretion of full length and truncated mouse OPG-Fc fusion proteins. A. Map indicating points of fusion to the human IgG1 Fc domain are indicated by arrowheads. B. Silver stain of a SDS-polyacrylamide gel of conditioned media obtained from cells expressing either Fl.Fc (Full length OPG fused to Fc at Leucine 401) or CT.Fc (Carboxy-terminal truncated OPG fused to Fc at threonine 180) fusion protein expression vectors. Lane 1, parent pCEP4 expression vector cell line; Lane 2, Fl.Fc vector cell line; Lane 3, CT.Fc vector cell line. C. Western blot of conditioned media obtained from Fl.Fc and CT.Fc fusion protein expression vectors /4 probed with anti-human IgGI Fc domain (Pierce). Lane 1, parent pCEP4 expression vector cell line; Lane 2, Fl.Fc vector cell line; Lane 3, CT.Fc vector cell line.
Figure 14. Expression of human OPG in E. coli. A. Construction of a bacterial expression vector. The LORF of the human OPG gene was amplified by PCR, then joined to a oligonucleotide linker fragment (top strand is SEQ ID NO:137; bottom strand is SEQ ID NO:127), and ligated into pAMG21 vector DNA. The resulting vector is capable of expressing OPG residues 32-401 linked to a N-terminal methionine residue.
B SDS-PAGE analysis of uninduced and induced bacterial harboring the pAMG21human OPG -32-401 plasmid. Lane 1, MW standards; lane 2, uninduced bacteria; lane 3, 30 0 C induction; lane 4, 37 0 C induction; lane 5, whole cell lysate from 37'C induction; lane 6, soluble fraction of whole cell lysate; lane 7, insoluble fraction of whole cell lysate; lane 8, purified inclusion bodies obtained from whole cell lysate.
Figure 15. Analysis of recombinant murine OPG produced in CHO cells by SDS- SPAGE and western blotting. An equal amount of CHO conditioned media was applied to each lane shown, and was prepared by treatment with either reducing sample buffer *:(left lane), or non-reducing sample buffer (right lane). After electrophoresis, the resolved proteins were transferred to a nylon membrane, then probed with anti-OPG antibodies. The relative positions of the 55 kd monomeric and 100 kd dimeric forms of OPG are indicated by arrowheads.
Figure 16. Pulse-chase analysis of recombinant murine OPG produced in CHO :cells. CHO cells were pulse-labeled with 3 5 S-methionine/cysteine, then chased for the indicated time. Metabolically labeled cultures were separated into both conditioned media and cells, and detergent extracts were prepared from each, clarified, then immunoprecipitated with anti-OPG antibodies. The immunoprecipitates were the resolved by SDS-PAGE, and exposed to film. Top left and right panels; samples analyzed under non-reducing conditions. Lower left and right panels; samples analyzed under reducing conditions. Top and bottom left panels; Cell extracts. Top and bottom right panels; Conditioned media extracts. The relative mobility of the 55 kd monomeric and 100 kd dimeric forms of OPG are indicated by arrowheads.
Figure 17. Expression of OPG in the CTLL-2 cell line. Serum-free conditioned media from CTLL-2 cells and CHO-mu OPG [1-401] transfected cells was prepared, 8 concentrated, then analyzed by non-reducing SDS-PAGE and western blotting. Left lane; CTLL-2 conditioned media. Right lane; CHO-muOPG conditioned media. The relative mobility of the 55 kd monomeric and 100 kd dimeric forms of OPG are indicated by arrowheads.
Figure 18. Detection of OPG expression in serum samples and liver extracts obtained from control and OPG transgenic mice. Transgenic mice were constructed as described in Example 4. OPG expression was visualized after SDS-PAGE followed by Western blotting using anti-OPG antibodies.
Figure 19. Effects of huOPG [22-401]-Fc fusion protein on osteoclast formation in vitro. The osteoclast forming assay was performed as described in Example 11A in the absence (control) or presence of the indicated amounts of huOPG [22-401]-Fc fusion.
Osteoclast formation was visualized by histochemical staining for tartrate acid phosphatase A. OPG added to 100 ng/ml. D. OPG added to 0. I ng/ml. E.
OPG added to 0.01 ng/ml. F. OPG added to 0.001 ng/ml. G. Control. No OPG added.
15 Figure 20. Decrease in osteoclast culture TRAP activity with increasing amounts of OPG. Indicated concentrations of huOPG [22-401 ]-Fc fusion protein were added to osteoclast forming assay and TRAP activity quantitated as described in Example 11A.
Figure 21. Effect of OPG on a terminal stage of osteoclast differentiation. huOPG .i [22-401]-Fc fusion was added to the osteoclast forming assay during the intermediate 2 0 stage of osteoclast maturation (days 5-6; OPG-CTL) or during the terminal stage of osteoclast maturation (days 7-15; CTL-OPG). TRAP activity was quantitated and compared with the activity observed in the absence of OPG (CTL-CTL) in the presence of OPG throughout (OPG-OPG).
Figure 22. Effects of IL-lb, IL-la and OPG on blood ionized calcium in mice.
Levels of blood ionized calcium were monitored after injection of IL-lb alone, IL-la alone, IL-lb plus muOPG [22-401]-Fc, IL-la plus MuOPG [22-401]-Fc, and muOPG [22-401]-Fc alone. Control mice received injections of phosphate buffered saline (PBS) only. IL-1B experiment shown in A; IL-la experiment shown in B.
Figure 23. Effects of OPG on calvarial osteoclasts in control and ILl-treated mice.
Histological methods for analyzing mice calvarial bone samples are described in Example 11B. Arrows indicate osteoclasts present in day 2-treated mice. Calvarial samples of mice receiving four PBS injections daily one injection of IL-1 and three injections of PBS daily one injection of PBS and three injections of OPG daily one injection of IL-1 and three injections of OPG daily.
Figure 24. Radiographic analysis of bone accumulation in marrow cavity of normal mice. Mice were injected subcutaneously with saline or muOPG [22-401]- Fc fusion (5mg/kg/d) for 14 days and bone density determined as described in Example 11C.
Figure 25. Histomorphometric analysis of bone accumulation in marrow cavity of normal mice. Injection experiments and bone histology performed as described in Example 11C.
Figure 26. Histology analysis of bone accumulation in marrow cavity of normal mice. Injection experiments and bone histology performed as described in Example 11C. A. Saline injection B. Injection of muOPG [22-401]-Fc fusion.
Figure 27. Activity of OPG administered to ovariectomized rats. In this two week experiment the trend to reduced bone density appears to be blocked by OPG or other anti-resorptive therapies. DEXA measurements were taken at time of ovariectomy and at week 1 and week 2 of treatment. The results are expressed as change from the initial bone density (Mean SEM).
Figure 28. Bone density in the femoral metaphysis, measured by S2 0 histomorphometric methods, tends to be lower in ovariectomized rats (OVX) than sham S• •operated animals (SHAM) 17 days following ovariectomy. This effect was blocked by OPG-Fc, with OPG-Fc treated ovariectomized rats (OVX+OPG) having significantly higher bone density than vehicle treated ovariectomized rats (OVX). (Mean SEM).
Detailed Description of the Invention A novel member of the tumor necrosis factor receptor (TNFR) superfamily was identified as an expressed sequence tag (EST) isolated from a fetal rat intestinal cDNA library The structures of the full-length rat cDNA clones and the corresponding mouse and human cDNA clones were determined as described in Examples 1 and 6. The rat, mouse and human genes are shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO: 122), and 9C-9D (SEQ ID NO: 124), respectively. All three sequences showed strong similarity to the extracellular domains of TNFR family members. None of the full-length cDNA clones isolated encoded transmembrane and cytoplasmic domains that would be expected for membrane-bound receptors, suggesting that these cDNAs encode soluble, secreted proteins rather than cell surface receptors. A portion of the human gene spanning nucleotides 1200-1353 shown in Figure 9D was deposited in the Genebank database on November 22, 1995 under accession no. 17188769.
The tissue distribution of the rat and human mRNA was determined as described in Example 2. In rat, mRNA expression was detected in kidney, liver, placenta and heart with the highest expression in the kidney. Expression in skeletal muscle and pancreas was also detected. In humans, expression was detected in the same tissues along with lymph node, thymus, spleen and appendix.
The rat cDNA was expressed in transgenic mice (Example 3) using the liver- Sspecific ApoE promoter expression system. Analysis of expressors showed a marked increase in bone density, particularly in long bones (femurs), vertebrae and flat bones (pelvis). Histological analysis of stained sections of bone showed severe osteopetrosis 15 (see Example 4) indicating a marked imbalance between bone formation and resorption which has led to a marked accumulation of bone and cartilage. A decrease in the number o°of trabecular osteoclasts in the bones of OPG expressor animals indicate that a significant portion of the activity of the TNFR-related protein may be to prevent bone resorption, a process mediated by osteoclasts. In view of the activity in transgenic 20 expressors, the TNFR-related proteins described herein are termed OPGs.
Using the rat cDNA sequence, mouse and human cDNA clones were isolated (Example Expression of mouse OPG in 293 cells and human OPG in E. coli is described in Examples 7 and 8. Mouse OPG was produced as an Fc fusion which was purified by Protein A affinity chromatography. Also described in Example 7 is the expression of full-length and truncated human and mouse OPG polypeptides in CHO and 293 cells either as fusion polypeptides to the Fc region of human IgG1 or as unfused polypeptides. The expression of full-length and truncated human and mouse OPGs in E.
coli either as Fc fusion polypeptides or as unfused polypeptides is described in Example 8. Purification of recombinantly produced mammalian and bacterial OPG is described in Example 11 The biological activity of OPG was determined using an in vitro osteoclast maturation assay, an in vivo model of interleukin-1 (IL-1) induced hypercalcemia, and injection studies of bone density in normal mice (see Example 11). The following OPG recombinant proteins produced in CHO or 293 cells demonstrated activity in the in E.
coli osteoclast maturation assay: muOPG [22-185]-Fc, muOPG [22-194]-Fc, muOPG [22-401]Fc, muOPG [22-401], huOPG [22-201]-Fc, huOPG [22-401]-Fc. muOPG [22- 180]-Fc produced in CHO cells and huOPG met[32-401] produced in E. coli did not demonstrate activity in the in vitro assay.
OPG from several sources was produced as a dimer and to some extent as a higher multimer. Rat OPG [22-401] produced in transgenic mice, muOPG [22-401] and huOPG [22-401] produced as a recombinant polypeptide in CHO cells, and OPG expressed as a naturally occurring product from a cytotoxic T cell line were predominantly dimers and trimers when analyzed on nonreducing SDS gels (see Example Truncated OPG polypeptides having deletions in the region of amino acids 15 186-401 OPG [1-185] and OPG [1-194]) were predominantly monomeric suggesting that the region 186-401 may be involved in self-association of OPG polypeptides. However, huOPG met[32-401] produced in E. coli was largely monomeric.
OPG may be important in regulating bone resorption. The protein appears to act as 20 a soluble receptor of the TNF family and may prevent a receptor-ligand interaction involved in the osteolytic pathway. One aspect of the regulation appears to be a 4* reduction in the number of osteoclasts.
Nucleic Acids The invention provides for an isolated nucleic acid encoding a polypeptide having at least one of the biological activities of OPG. As described herein, the biological activities of OPG include, but are not limited to, any activity involving bone metabolism and in particular, include increasing bone density. The nucleic acids of the invention are selected from the following: a) the nucleic acid sequences as shown in Figures 2B-2C (SEQ ID NO:120), 9A- 9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124) or complementary strands thereof; b) the nucleic acids which hybridize under stringent conditions with the polypeptide-encoding region in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124); and c) nucleic acids which hybridize under stringent conditions with nucleotides 148 through 337 inclusive as shown in Figure 1A.
d) the nucleic acid sequences which are degenerate to the sequences in and The invention provides for nucleic acids which encode rat, mouse and human OPG as well as nucleic acid sequences hybridizing thereto which encode a polypeptide having at least one of the biological activities of OPG. Also provided for are nucleic acids which hybridize to a rat OPG EST encompassing nucleotides 148-337 as shown in Figure 1A. The conditions for hybridization are generally of high stringency such as 5xSSC, 50% formamide and 42 0 C described in Example 1 of the specification.
E vaienfrstingency to these Uconditionsmay bcreadtrly obtainedubyadjustXg salt and organic solvent concentrations and temperature. The nucleic acids in encompass 15 sequences encoding OPG-related polypeptides which do not undergo detectable hybridization with other known members of the TNF receptor superfamily. In a preferred embodiment, the nucleic acids are as shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124).
The length of hybridizing nucleic acids of the invention may be variable since 20 hybridization may occur in part or all of the polypeptide-encoding regions as shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO: 124), and may also occur in adjacent noncoding regions. Therefore, hybridizing nucleic acids may be truncations or extensions of the sequences shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124). Truncated or extended nucleic acids are encompassed by the invention provided they retain one or more of the biological properties of OPG. The hybridizing nucleic acids may also include adjacent noncoding regions which are 5' and/or 3' to the OPG coding region.
The noncoding regions include regulatory regions involved in OPG expression, such as promoters, enhance, translational initiation sites, transcription termination sites and the like.
13 Hybridization conditions for nucleic acids are described in Sambrook et al.
Molecular Cloning: A Laboratory Manual, 2nd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989) DNA encoding rat OPG was provided in plasmid pMO-B 1.1 deposited with the American Type Culture Collection, Rockville, MD on December 27, 1995 under ATCC accession no. 69970. DNA encoding mouse OPG was provided in plasmid pRcCMVmurine OPG deposited with the American Type Culture Collection, Rockville, MD on December 27, 1995 under accession no. 69971. DNA encoding human OPG was provided in plasmid pRcCMV human OPG deposited with the American Type Culture Collection, Rockville, MD on December 27, 1995 under accession no. 69969. The nucleic acids of the invention will hybridize under stringent conditions to the DNA inserts of ATCC accession nos. 69969, 69970, and 69971 and have at least one of the biological activities of OPG.
Also provided by the invention are derivatives of the nucleic acid sequences as 15 shown in Figures 2B, 9A and 9B. As used herein, derivatives include nucleic acid sequences having addition, substitution, insertion or deletion of one or more residues such that the resulting sequences encode polypeptides having one or more amino acid residues which have been added, deleted, inserted or substituted and the resulting i polypeptide has the activity of OPG. The nucleic acid derivatives may be naturally S 20 occurring, such as by splice variation or polymorphism, or may be constructed using site-directed mutagenesis techniques available to the skilled worker. One example of a naturally occurring variant of OPG is a nucleic acid encoding a lys to asn change at residue 3 within the leader sequence (see Example It is anticipated that nucleic acid derivatives will encode amino acid changes in regions of the molecule which are least likely to disrupt biological activity. Other derivatives include a nucleic acid encoding a membrane-bound form of OPG having an extracellular domain as shown in Figures 2B- 2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO:124) along with transmembrane and cytoplasmic domains.
In one embodiment, derivatives of OPG include nucleic acids encoding truncated forms of OPG having one or more amino acids deleted from the carboxy terminus.
Nucleic acids encoding OPG may have from 1 to 216 amino acids deleted from the 14 carboxy terminus. Optionally, an antibody Fc region may extend from the new carboxy terminus to yield a biologically active OPG-Fc fusion polypeptide. (see Example 11). In preferred embodiments, nucleic acids encode OPG having the amino acid sequence from residues 22-185, 22-189, 22-194 or 22-201 (using numbering in Figure 9E-F) and optionally, encoding an Fc region of human IgG.
Also included are nucleic acids encoding truncated forms of OPG having one or more amino acids deleted from the amino terminus. Truncated forms include those lacking part or all the 21 amino acids comprising the leader sequence. Additionally, the invention provides for nucleic acids encoding OPG having from 1 to 10 amino acids deleted from the mature amino terminus (at residue 22) and ,optionally, having from 1 to 216 amino acids deleted from the carboxy terminus (at residue 401). Optionally, the S. nucleic acids may encode a methionine residue at the amino terminus. Examples of such OPG irunicated polypepuides aie duescibed in Exaniple 8.
Examples of the nucleic acids of the invention include cDNA, genomic DNA, 15 synthetic DNA and RNA. cDNA is obtained from libraries prepared from mRNA isolated from various tissues expressing OPG. In humans, tissue sources for OPG *4 include kidney, liver, placenta and heart. Genomic DNA encoding OPG is obtained from genomic libraries which are commercially available from a variety of species.
Synthetic DNA is obtained by chemical synthesis of overlapping oligonucleotide 20 fragments followed by assembly of the fragments to reconstitute part or all of the coding Sregion and flanking sequences (see U.S. Patent No. 4,695,623 describing the chemical :synthesis of interferon genes). RNA is obtained most easily by procaryotic expression eo vectors which direct high-level synthesis of mRNA, such as vectors using T7 promoters and RNA polymerase.
Nucleic acid sequences of the invention are used for the detection of OPG sequences in biological samples in order to determine which cells and tissues are expressing OPG mRNA. The sequences may also be used to screen cDNA and genomic libraries for sequences related to OPG. Such screening is well within the capabilities of one skilled in the art using appropriate hybridization conditions to detect homologus sequences. The nucleic acids are also useful for modulating the expression of OPG levels by anti-sense therapy or gene therapy. The nucleic acids are also used for the development of transgenic animals which may be used for the production of the polypeptide and for the study of biological activity (see Example 3).
Vectors and Host Cells Expression vectors containing nucleic acid sequences encoding OPG, host cells transformed with said vectors and methods for the production of OPG are also provided by the invention. An overview of expression of recombinant proteins is found in Methods of Enzymology v. 185, Goeddel, D.V. ed. Academic Press (1990).
Host cells for the production of OPG include procaryotic host cells, such as E. coli, yeast, plant, insect and mammalian host cells. E. coli strains such as HB 101 or JM101 are suitable for expression. Preferred mammalian host cells include COS, CHOd-, 293, CV-1, 3T3, baby hamster kidney (BHK) cells and others. Mammalian host cells are preferred when post-translational modifications, such as glycosylation and polypeptide processing, are important for OPG activity.Mammaian expression allows for the production of secreted polypeptides which may be recovered from the growth medium.
Vectors for the expression of OPG contain at a minimum sequences required for vector propogation and for expression of the cloned insert. These sequences include a replication origin, selection marker, promoter, ribosome binding site, enhancer sequences, RNA splice sites and transcription termination site. Vectors suitable for expression in the aforementioned host cells are readily available and the nucleic acids of 2 0 the invention are inserted into the vectors using standard recombinant DNA techniques.
Vectors for tissue-specific expression of OPG are also included. Such vectors include promoters which function specifically in liver, kidney or other organs for production in mice, and viral vectors for the expression of OPG in targeted human cells.
Using an appropriate host-vector system, OPG is produced recombinantly by culturing a host cell transformed with an expression vector containing nucleic acid sequences encoding OPG under conditions such that OPG is produced, and isolating the product of expression. OPG is produced in the supematant of transfected mammalian cells or in inclusion bodies of transformed bacterial host cells. OPG so produced may be purified by procedures known to one skilled in the art as described below. The expression of OPG in mammalian and bacterial host systems is described in Examples 7 and 8. Expression vectors for mammalian hosts are exemplified by plasmids such as 16 pDSRa described in PCT Application No. 90/14363. Expression vectors for bacterial host cells are exemplified by plasmids pAMG21 and pAMG22-His described in Example 8. Plasmid pAMG21 was deposited with the American Type Culture Collection, Rockville, MD on July 24, 1996 under accession no. 98113. Plasmid pAMG22-His was deposited with the American Type Culture Collection, Rockville, MD on July 24, 1996 under accession no. 98112. It is anticipated that the specific plasmids and host cells described are for illustrative purposes and that other available plasmids and host cells could also be used to express the polypeptides.
The invention also provides for expression of OPG from endogenous nucleic acids by in vivo or ex vivo recombination events to allow modulation of OPG from the host chromosome. Expression of OPG by the introduction of exogenous regulatory sequences promoters or enhancers) capable of directing the production of OPG from endogenous OPG coding regions is also encompassed. Stimulation of endogenous regulatory sequences capable of directing OPG production by exposure to transcriptional enhancing factors) is also provided by the invention.
Polypeptides The invention provides for OPG, a novel member of the TNF receptor superfamily, having an activity associated with bone metabolism and in particular having I the activity of inhibiting bone resorption thereby increasing bone density. OPG refers to a polypeptide having an amino acid sequence of mouse, rat or human OPG or a S. derivative thereof having at least one of the biological activities of OPG. The amino acid sequences of rat, mouse and human OPG are shown in Figures 2B-2C (SEQ ID NO:121), 9A-9B (SEQ ID NO:123), and 9C-9D (SEQ ID NO:125) respectively. A derivative of OPG refers to a polypeptide having an addition, deletion, insertion or substitution of one or more amino acids such that the resulting polypeptide has at least one of the biological activities of OPG. The biological activities of OPG include, but are not limited to, activities involving bone metabolism. Preferably, the polypeptides will have the amino terminal leader sequence of 21 amino acids removed.
OPG polypeptides encompassed by the invention include rat [1-401], rat [22-180], rat [22-401], rat [22-401]-Fc fusion, rat [1-180]-Fc fusion, mouse [1-401], mouse [1- 180], mouse [22-401], human [1-401], mouse [22-180], human [22-401], human [22- 17 180], human [1-180], human [22-180]-Fc fusion and human met-32-401. Amino acid numbering is as shown in SEQ ID NO:121 (rat), SEQ ID NO:123 (mouse) and SEQ ID NO:125 (human). Also encompassed are polypeptide derivatives having deletions or carboxy-terminal truncations of part or all of amino acids residues 180-401 of OPG; one or more amino acid changes in residues 180-401; deletion of part or all of a cysteine-rich domain of OPG, in particular deletion of the distal (carboxy-terminal) cysteine-rich domain; and one or more amino acid changes in a cysteine-rich domain, in particular in the distal (carboxy-terminal) cysteine-rich domain. In one embodiment, OPG has from 1 to about 216 amino acids deleted from the carboxy terminus. In another embodiment, OPG has from 1 to about 10 amino acids deleted from the mature amino terminus (wherein the mature amino terminus is at residue 22) and, optionally, has from 1 to about 216 amino acids deleted from the carboxy terminus.
Additional OPG polypeptides encompassed by the invention include the following: human [22-180]-Fc fusion, human [22-201]-Fc fusion, human [22-401]-Fc fusion, mouse [22-185]-Fc fusion, mouse [22-194]-Fc fusion. These polypeptides are produced in mammalian host cells, such as CHO or 293 cells, Additional OPG polypeptides encompassed by the invention which are expressed in procaryotic host cells include the following: human met[22-401], Fc-human met[22-401] fusion (Fc region is fused at the amino terminus of the full-length OPG coding sequence as described in Example 8), human met[22-401]-Fc fusion (Fc region fused to the full-lengh OPG sequence), Fcmouse met[22-401] fusion, mouse met[22-401]-Fc fusion, human met[27-401], human met[22-185], human met[22-189], human met[22-194], human met[22-194] human met [22-194] (P26A), human met[27-185], human met[27-189], human met[27- 194], human met-arg-gly-ser-(his)6 [22-401], human met-lys [22-401], human met- (lys)3-[22-401], human met[22-401]-Fc (P25A), human met[22-401](P25A), human met[22-401](P26A), human met[22-401] (P26D), mouse met[22-401], mouse met[27- 401], mouse met[32-401], mouse met[27-180], mouse met[22-189], mouse met[22-194], mouse met[27-189], mouse met[27-194], mouse met-lys[22-401], mouse HEK[22- 401](A45T), mouse met-lys-(his)7[22-401], mouse met-lys[22-401]-(his)7 and mouse met[27-401] (P33E, G36S, A45P). It is understood that the above OPG polypeptides produced in procaryotic host cells have an amino-terminal methionine residue, if such a 18 residue is not indicated. In specific examples, OPG-Fc fusion were produced using a 227 amino acid region of human IgGl-gl was used having the sequence as shown in Ellison et al. (Nuc. Acids Res. 10, 4071-4079 (1982)). However, variants of the Fc region of human IgG may also be used.
Analysis of the biological activity of carboxy-terminal OPG truncations fused to the human IgG 1 Fc region indicates a portion of OPG of about 164 amino acids which is required for activity. This region encompasses amino acids 22-185, preferably those in Figure 9C-9D (SEQ ID NO:125), and comprises four cysteine-rich domains characteristic of the cysteine-rich domains of TNFR extraceullular domains.
Using the homology between OPG and the extracellular ligand binding domains of TNF receptor family members, a three-dimensional model of OPG was generated based S"upon the known crystal structure of the extracellular domain of TNFR-I (see Example 6).
This model was ursed to identify those residues within OPG which may be impo:rtant fo biological activity. Cysteine residues that are involved in maintaining the structure of 15 the four cysteine-rich domains were identified. The following disulfide bonds were identified in the model: Domain 1: cys41 to cys54, cys44 to cys62, tyr23 and-his 66 may act to stabilize the structure of this domain; Domain 2: cys65 to cys80, cys83 to cys98, cys87 to cysl05; Domain 3: cysl07 to cysl 18, cysl24 to cysl42; Domain 4: cysl45 to cysl60, cys166 to cys185. Residues were also identified which were in close proximity 20 to TNFb as shown in Figures 11 and 12A-12B. In this model, it is assumed that OPG binds to a corresponding ligand; TNFb was used as a model ligand to simulate the Sinteraction of OPG with its ligand. Based upon this modeling, the following residues in OPG may be important for ligand binding: glu34, lys43, pro66 to gln91 (in particular, pro66, his68, tyr69, tyr70, thr71, asp72, ser73, his76, ser77, asp78, glu79, leu81, tyr82, pro85, va186, lys88, glu90 and gln91), glu153 and Alterations in these amino acid residues, either singly or in combination, may alter the biological activity of OPG. For example, changes in specific cysteine residues may alter the structure of individual cysteine-rich domains, whereas changes in residues important for ligand binding may affect physical interactions of OPG with ligand.
Structural models can aid in identifying analogs which have more desirable properties, such as enhanced biological activity, greater stability, or greater ease of formulation.
19 The invention also provides for an OPG multimer comprising OPG monomers.
OPG appears to be active as a multimer dimer, trimer of a higher number of monomers). Preferably, OPG multimers are dimers or trimers. OPG multimers may comprise monomers having the amino acid sequence of OPG sufficient to promote multimer formation or may comprise monomers having heterologous sequences such as an antibody Fc region. Analysis of carboxy-terminal deletions of OPG suggest that at least a portion of the region 186-401 is involved in association of OPG polypeptides.
Substitution of part or all of the region of OPG amino acids 186-401 with an amino acid sequence capable of self-association is also encompassed by the invention.
Alternatively, OPG polypeptides or derivatives thereof may be modified to form dimers or multimers by site directed mutagenesis to create unpaired cysteine residues for interchain disulfide bond romation, by photochemical crosslinking, such as exposure to -r.-...ultraviolet light, or by chemical crosslink-ing with bifuinctional linker moleclessuch as bifunctional polyethylene glycol and the like.
15 Modifications of OPG polypeptides are encompassed by the invention and include post-translational modifications N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition of an N-terminal methionine residue as a result of procaryotic host 20 cell expression. The polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
Further modifications of OPG include chimeric proteins wherein OPG is fused to a heterologous amino acid sequence. The heterologous sequence may be any sequence which allows the resulting fusion protein to retain the activity of OPG. The heterologous sequences include for example, immunoglobulin fusions, such as Fc fusions, which may aid in purification of the protein. A heterologous sequence which promotes association of OPG monomers to form dimers, trimers and other higher multimeric forms is preferred.
3 0 The polypeptides of the invention are isolated and purified from other polypeptides present in tissues, cell lines and transformed host cells expressing OPG, or purified from components in cell cultures containing the secreted protein. In one embodiment, the polypeptide is free from association with other human proteins, such as the expression product of a bacterial host cell.
Also provided by the invention are chemically modified derivatives of OPG which may provide additional advantages such as increasing stability and circulating time of the polypeptide, or decreasing immunogenicity (see U.S. Patent No. 4,179,337). The chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like. The polypeptides may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
The polymer may be of any molecular weight, and may be branched or unbranched-Forpolyethyleneglyel-theyreferredmolecular-weightisbetween-about kDa and about 100kDa (the term "about" indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and i other known effects of the polyethylene glycol to a therapeutic protein or analog).
S2 0 The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on functional or antigenic domains of the protein. There are a number of attachment methods available to those skilled in the art, e.g. EP 0 401 384 herein incorporated by reference (coupling PEG to G-CSF), see also Malik et al., Exp. Hematol. 20: 1028-1035 (1992) (reporting pegylation of GM-CSF using tresyl chloride). For example, polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue. Sulfhydrl groups may also be used as a reactive group for attaching the 21 polyethylene glycol molecule(s). Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
One may specifically desire N-terminally chemically modified protein. Using polyethylene glycol as an illustration of the present compositions, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (or peptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein. The method of obtaining the Nterminally pegylated preparation separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective N-terminal chemically modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction 15 conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
Synthetic OPG dimers may be prepared by various chemical crosslinking procedures. OPG monomers may be chemically linked in any fashion that retains or S. enhances the biological activity of OPG. A variety of chemical crosslinkers may be used depending upon which properties of the protein dimer are desired. For example, crosslinkers may be short and relatively rigid or longer and more flexible, may be biologically reversible, and may provide reduced immunogenicity or longer pharmacokinetic half-life.
In one example, OPG molecules are linked through the amino terminus by a two step synthesis (see Example 12). In the first step, OPG is chemically modified at the amino terminus to introduce a protected thiol, which after purification is deprotected and used as a point of attachment for site-specific conjugation through a variety of crosslinkers with a second OPG molecule. Amino-terminal crosslinks include, but are not limited to, a disulfide bond, thioether linkages using short-chain, bis-functional aliphatic crosslinkers, and thioether linkages to variable length, bifunctional polyethylene glycol crosslinkers (PEG "dumbbells"). Also encompassed by PEG dumbbell synthesis of OPG dimers is a byproduct of such synthesis, termed a "monobell". An OPG monobell consists of a monomer coupled to a linear bifunctional PEG with a free polymer terminus. Alternatively, OPG may be crosslinked directly through a variety of amine specific homobifunctional crosslinking techniques which include reagents such as: diethylenetriaminepentaacetic dianhydride (DTPA), pbenzoquinone (pBQ) or bis(sulfosuccinimidyl) suberate (BS 3 as well as others known in the art. It is also possible to thiolate OPG directly with reagents such as iminothiolane in the presence of a variety of bifunctional, thiol specific crosslinkers, such as PEG bismaleimide, and achieve dimerization and/or dumbbells in a one step process.
A method for the purification of OPG from natural sources and from transfected host cells is also included. The purification process may employ one or more standard protein purification steps in an appropriate order to obtain purified protein. The hromatgraphysteps cai.luUde ionexchange geI filtrIatIon, hydruphoobicinteraction, reverse phase, chromatofocusing, affinity chromatography employing an anti-OPG 15 antibody or biotin-streptavidin affinity complex and the like.
Antibodies Also encompassed by the invention are antibodies specifically binding to OPG.
Antigens for the generation of antibodies may be full-length polypeptides or peptides spanning a portion of the OPG sequence. Immunological procedures for the generation of polyclonal or monoclonal antibodies reactive with OPG are known to one skilled in the art (see, for example, Harlow and Lane, Antibodies: A Laboratory Manual Cold Spring Harbor Laboratory Press, Cold Spring Harbor N.Y. (1988)). Antibodies so produced are characterized for binding specificity and epitope recognition using standard enzyme-linked immunosorbent assays. Antibodies also include chimeric antibodies having variable and constant domain regions derived from different species. In one embodiment, the chimeric antibodies are humanized antibodies having murine variable domains and human constant domains. Also encompassed are complementary determining regions grafted to a human framework (so-called CDR-grafted antibodies).
Chimeric and CDR-grafted antibodies are made by recombinant methods known to one skilled in the art. Also encompassed are human antibodies made in mice.
Anti-OPG antibodies of the invention may be used as an affinity reagent to purify OPG from biological samples (see Example 10). In one method, the antibody is immobilized on CnBr-activated Sepharose and a column of antibody-Sepharose conjugate is used to remove OPG from liquid samples. Antibodies are also used as diagnostic reagents to detect and quantitate OPG in biological samples by methods described below.
Pharmaceutical compositions The invention also provides for pharmaceutical compositions comprising a therapeutically effective amount of the polypeptide of the invention together with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and/or adjuvant. The term "therapeutically effective amount" means an amount which provides a therapeutic effect for a specified condition and route of administration. The composition may be in a liquid or lyphilized fomancomprisesdiuentTris, acetate or phosphate buffers) having various pH values and ionic strengths, solubilizer such as 15 Tween or Polysorbate, carriers such as human serum albumin or gelatin, preservatives such as thimerosal or benzyl alcohol, and antioxidants such as ascrobic acid or sodium metabisulfite. Also encompassed are compositions comprising OPG modified with water soluble polymers to increase solubility or stability. Compositions may also comprise incorporation of OPG into liposomes, microemulsions, micelles or vesicles for controlled delivery over an extended period of time.
Specifically, OPG compositions may comprise incorporation into polymer matricies such as hydrogels, silicones, polyethylenes, ethylene-vinyl acetate copolymers, or biodegradable polymers. Examples of hydrogels include polyhydroxyalkylmethacrylates (p-HEMA), polyacrylamide, polymethacrylamide, polyvinylpyrrolidone, polyvinyl alcohol and various polyelectrolyte complexes.
Examples of biodegradable polymers include polylactic acid (PLA), polyglycolic acid (PGA), copolymers of PLA and PGA, polyamides and copolymers of polyamides and polyesters. Other controlled release formulations include microcapsules, microspheres, macromolecular complexes and polymeric beads which may be administered by injection.
24 Selection of a particular composition will depend upon a number of factors, including the condition being treated, the route of administration and the pharmacokinetic parameters desired. A more extensive survey of component suitable for pharmaceutical compositions is found in Remington's Pharmaceutical Sciences, 18th ed.
A.R. Gennaro, ed. Mack, Easton, PA (1980).
Compositions of the invention may be administered by injection, either subcutaneous, intravenous or intramuscular, or by oral, nasal, pulmonary or rectal administration. The route of administration eventually chosen will depend upon a number of factors and may be ascertained by one skilled in the art.
The invention also provides for pharmaceutical compositions comprising a therapeutically effective amount of the nucleic acids of the invention together with a pharmaceutically acceptable adjuvant. Nucleic acid compositions will be suitable for the delivery of Dart or all of the OPGT coding reginn tn cells and t;issues as part of an anti; sense or gene therapy regimen.
15 Methods of Treatment Bone tissue provides support for the body and consists of mineral (largely calcium and phosphorous), a matrix of collagenous and noncollagenous proteins, and cells.
Three types of cells found in bone, osteocytes, osteoblasts and osteoclasts, are involved in the dynamic process by which bone is continually formed and resorbed. Osteoblasts 20 promote formation of bone tissue whereas osteoclasts are associated with resorption.
Resorption, or the dissolution of bone matrix and mineral, is a fast and efficient process compared to bone formation and can release large amounts of mineral from bone.
Osteoclasts are involved in the regulation of the normal remodeling of skeletal tissue and in resorption induced by hormones. For instance, resorption is stimulated by the secretion of parathyroid hormone in response to decreasing concentrations of calcium ion in extracellular fluids. In contrast, inhibition of resorption is the principal function of calcitonin. In addition, metabolites of vitamin D alter the responsiveness of bone to parathyroid hormone and calcitonin.
After skeletal maturity, the amount of bone in the skeleton reflects the balance (or imbalance) of bone formation and bone resorption. Peak bone mass occurs after skeletal maturity prior to the fourth decade. Between the fourth and fifth decades, the equilibrium shifts and bone resorption dominates. The inevitable decrease in bone mass with advancing years starts earlier in females than males and is distinctly accelerated after menopause in some females (principally those of Caucasian and Asian descent).
Osteopenia is a condition relating generally to any decrease in bone mass to below normal levels. Such a condition may arise from a decrease in the rate of bone synthesis or an increase in the rate of bone destruction or both. The most common form of osteopenia is primary osteoporosis, also referred to as postmenopausal and senile osteoporosis. This form of osteoporosis is a consequence of the universal loss of bone with age and is usually a result of increase in bone resorption with a normal rate of bone formation. About 25 to 30 percent of all white females in the United States develop symptomatic osteoporosis. A direct relationship exists between osteoporosis and the 0oo. .incidence of hip, femoral, neck and inter-trochanteric fracture in women 45 years and older. Elderly males develop symptomaticosteoporosis between the ages of 50 and but the disease primarily affects females.
15 The cause of postmenopausal and senile osteoporosis is unknown. Several factors have been identified which may contribute to the condition. They include alteration in hormone levels accompanying aging and inadequate calcium consumption attributed to decreased intestinal absorption of calcium and other minerals. Treatments have usually included hormone therapy or dietary supplements in an attempt to retard the process. To 20 date, however, an effective treatment for bone loss does not exist.
The invention provides for a method of treating a bone disorder using a therapeutically effective amount of OPG. The bone disorder may be any disorder characterized by a net bone loss (osteopenia or osteolysis). In general, treatment with OPG is anticipated when it is necessary to suppress the rate of bone resorption. Thus treatment may be done to reduce the rate of bone resorption where the resorption rate is above normal or to reduce bone resorption to below normal levels in order to compensate for below normal levels of bone formation.
Conditions which are treatable with OPG include the following: Osteoporosis, such as primary osteoporosis, endocrine osteoporosis (hyperthyroidism, hyperparathryoidism, Cushing's syndrome, and acromegaly), hereditary and congenital forms of osteoporosis (osteogenesis imperfecta, 26 homocystinuria, Menkes' syndrome, and Riley-Day syndrome) and osteoporosis due to immobilization of extremities.
Paget's disease of bone (osteitis deformans) in adults and juveniles Osteomyelitis, or an infectious lesion in bone, leading to bone loss.
Hypercalcemia resulting from solid tumors (breast, lung and kidney) and hematologic malignacies (multiple myeloma, lymphoma and leukemia), idiopathic hypercalcemia, and hypercalcemia associated with hyperthryoidism and renal function disorders.
Osteopenia following surgery, induced by steroid administration, and associated with disorders of the small and large intestine and with chronic hepatic and renal diseases.
Osteonecrosis, or bone cell death, associated with traumatic injury or nontraumatic necrosis associated with Gaucher's disease, sickle cell anemia, systemic lupus erythematosus and other conditions.
S* 15 Bone loss due to rheumatoid arthritis.
Periodontal bone loss.
Osteolytic metastasis It is understood that OPG may be used alone or in conjunction with other factors S. for the treatment of bone disorders. In one embodiment, osteoprotegerein is used in 20 conjunction with a therapeutically effective amount of a factor which stimulates bone formation. Such factors include but are not limited to the bone morphogenic factors designated BMP-1 through BMP-12, transforming growth factor-b (TGF-b) and TGF-b family members, interleukin-1 inhibitors, TNFa inhibitors, parathyroid hormone and analogs thereof, parathyroid related protein and analogs thereof, E series prostaglandins, bisphosphonates (such as alendronate and others), and bone-enhancing minerals such as fluoride and calcium.
The following examples are offered to more fully illustrate the invention, but are not construed as limiting the scope thereof.
EXAMPLE 1 Identification and isolation of the rat OPG cDNA Materials and methods for cDNA cloning and analysis are described in Maniatis et al, ibid. Polymerase chain reactions (PCR) were performed using a Perkin-Elmer 9600 thermocycler using PCR reaction mixture (Boehringer-Mannheim) and primer concentrations specified by the manufacturer. In general, 25-50 ml reactions were denatured at 94 0 C, followed by 20-40 cycles of 94°C for 5 seconds, 50-60 0 C for seconds, and 72 0 C for 3-5 minutes. Reactions were the treated for 72 °C for minutes. Reactions were then analyzed by gel electrophoresis as described in Maniatis et al., ibid.
A cDNA library was constructed using mRNA isolated from embryonic intestine for EST analysis (Adams et al. Science 252, 1651-1656 (1991)). Rat embryos were dissected, and the entire developing small and large intestine removed and washed oin PBS. Total cell RNA was purified by acid guanidinium thiocyanate-phenol- 15 chloroform extraction (Chomczynski and Sacchi Anal. Biochem. 162. 156-159, (1987)).
The poly mRNA fraction was obtained from the total RNA preparation by adsorption to, and elution from, Dynabeads Oligo (dT)25 (Dynal Corp) using the manufacturer's recommended procedures. A random primed cDNA library was prepared using the Superscript Plasmid System (Gibco BRL, Gaithersburg, Md). The 20 random cDNA primer containing an internal Not I restriction site was used to initiate first strand synthesis and had the following sequence: 5' -AAAGGAAGGAAAAAAGCGGCCGCTACANNNNNNNNT-3' (SEQ ID NO:1) S" Not I For the first strand synthesis three separate reactions were assembled that contained 2.5 mg of poly(A) RNA and 120 ng, 360 ng or 1,080 ng of random primer.
After second strand synthesis, the reaction products were separately extracted with a mixture ofphenol:choroform:isoamyl alcohol (25:24:1 ratio), and then ethanol precipitated. The double strand (ds) cDNA products of the three reactions were combined and ligated to the following ds oligonucleotide adapter: 5'-TCGACCCACGCGTCCG-3' (SEQ ID NO:2) (SEQ ID NO:3) 28 After ligation the cDNA was digested to completion with Not I, extracted with phenol:chloroform:isoamyl (25:24:1) alcohol and ethanol precipitated. The resuspended cDNA was then size fractionated by gel filtration using premade columns provided with the Superscript Plasmid System (Gibco BRL, Gaithersburg, Md) as recommended by the manufacturer. The two fractions containing the largest cDNA products were pooled, ethanol precipitated and then directionally ligated into Not I and Sal I digested pMOB vector DNA (Strathmann et al, 1991). The ligated cDNA was introduced into competent ElectroMAX DH10B E. coli (Gibco BRL, Gaithersburg, MD) by electroporation. For automated sequence analysis approximately 10,000 transformants were plated on 20cm x 20cm agar plates containing ampicillin supplemented LB nutrient media. The colonies that arose were picked and arrayed onto 96 well microtiter plates containing 200 ml of L-broth, 7.5% glycerol, and 50 mg/ml ampicillin. The cultures were grown overnight at *37°C, a duplicate set of microtiter plates were made lusing a sterile 96 pin replicating tool, then both sets were stored at -80 0 C for further analysis. For full-length cDNA 15 cloning approximately one million transformants were plated on 96 bacterial ampicillin plates containing about 10,000 clones each. The plasmid DNA from each pool was separately isolated using the Qiagen Plasmid Maxi Kit (Qiagen Corp.,Germany) and arrayed into 96 microtiter plates for PCR analyses.
To sequence random fetal rat intestine cDNA clones, glycerol stocks were thawed, 20 and small aliquots diluted 1:25 in distilled. Approximately 3.0 ul of diluted bacterial cultures were added to PCR reaction mixture (Boehringer-Mannheim) containing the following oligonucleotides: 5'-TGTAAAACGACGGCCAGT-3' (SEQ ID NO:4) 5'-CAGGAAACAGCTATGACC-3' (SEQ ID The reactions were incubated in a thermocycler (Perkin-Elmer 9600) with the following cycle conditions: 94 C for 2 minutes; 30 cycles of 94°C for 5 seconds, for 5 seconds, and 72 0 C for 3 minutes.; 72 0 C for 4 minutes. After incubation in the thermocycler, the reactions were diluted with 2.0 mL of water. The amplified DNA fragments were further purified using Centricon columns (Princeton Separations) using the manufacturer's recommended procedures. The PCR reaction products were sequenced on an Applied Biosystems 3 73A automated DNA sequencer using T3 primer (oligonucleotide 353-23; 5'-CAATTAACCCTCACTAAAGG-3') (SEQ ID NO:6) Taq dye-terminator reactions (Applied Biosystems) following the manufacturer's recommended procedures.
The resulting 5' nucleotide sequence obtained from randomly picked cDNA clones translated and then compared to the existing database of known protein sequences using a modified version of the FASTA program (Pearson et al. Meth.
Enzymol. 183, (1990)). Translated sequences were also analysed for the presence of a specific cysteine-rich protein motif found in all known members of the tumor necrosis factor receptor (TNFR) superfamily (Smith et al. Cell 76, 959-962 (1994)), using the sequence profile method ofGribskov et al. (Proc. Natl. Acad. Sci. USA 83, 4355-4359 (1987)), as modified by Luethy et al. (Protein Science 3, 139-146 (1994)).
Using the FASTA and Profile search data, an EST, FRI-1 (Fetal Rat Intestine-1), was identified as a possible new member of the TNFR superfamily. FRI-1 contained an approximately 600 bp insert with a LORF of about 150 amino acids. The closest match in the database was the human type II TNFR (TNFR-2). The region compared showed an -43% homology between TNFR-2 and FRI-1 over this 150 aa LORF. Profile analysis using the first and second cysteine-rich repeats of the TNFR superfamily yielded a Z score of indicating that the FRI-1 gene possibly encodes a new family member.
To deduce the structure of the FRI-1 product, the fetal rat intestine cDNA library 2 0 was screened for full length clones. The following oligonucleotides were derived from the original FRI-1 sequence: 5'-GCATTATGACCCAGAAACCGGAC-3' (SEQ ID NO:7) 5'-AGGTAGCGCCCTTCCTCACATTC-3' (SEQ.ID NO:8) These primers were used in PCR reactions to screen 96 pools of plasmid DNA, each pool containing plasmid DNA from 10,000 independent cDNA clones.
Approximately 1 ug ofplasmid pool DNA was amplified in a PCR reaction mixture (Boehringer-Mannheim) using a Perkin-Elmer 96 well thermal cycler with the following cycle conditions: 2 min at 94 0 C,1 cycle; 15 sec at 94 0 C, then 45 sec at 65 0 C, 30 cycles; 7 min at 65 0 C, 1 cycle. PCR reaction products were analysed by gel electrophoresis. 13 out of 96 plasmid DNA pools gave rise to amplified DNA products with the expected relative molecular mass.
DNA from one positive pool was used to transform competent ElectroMAX E. coli (Gibco BRL, Gaithersburg, MD) as described above. Approximately 40,000 transformants were plated onto sterile nitrocellulose filters (BA-85, Schleicher and Schuell), and then screened by colony hybridization using a 3 2 p-dCTP labelled version of the PCR product obtained above. Filters were prehybridized in 5X SSC, 50% deionized formamide, 5X Denhardt's solution, 0.5% SDS, and 100 ug/ml denatured salmon sperm DNA for 2-4 hours at 42 0 C. Filters were then hybridized in 5X SSC, deionized formamide, 2X Denhardt's solution, 0.1% SDS, 100 mg/ml denatured salmon sperm DNA, and -5 ng/ml of labelled probe for -18 hours at 42 0 C. The filters were then washed in 2X SSC for 10 min at RT, 1X SSC for 10 min at 55 0 C, and finally in SSC for 10-15 min at 55 0 C. Hybridizing clones were detected following autoradiography, and then replated onto nitrocellulose filters for secondary screening.
Upon secondary screeinga-piasmticicone(tjBr.A)was-isolated-henampiifediin Lbroth media containing 100 ug/ml ampicillin and the plasmid DNA obtained. Both strands of the 2.4 kb pB1.1 insert were sequenced.
The pB1.1 insert sequence was used for a FASTA search of the public database to 'detect any existing sequence matches and/or similarities. No matches to any known genes or EST's were found, although there was an approximate 45% similarity to the human and mouse TNFR-2 genes. A methionine start codon is found at bp 124 of the 20 nucleotide sequence, followed by a LORF encoding 401 aa residues that terminates at bp 1327. The 401 aa residue product is predicted to have a hydrophobic signal peptide of approximately 31 residues at its N-terminus, and 4 potential sites of N-linked glycosylation. No hydrophobic transmembrane spanning sequence was identified using the PepPlot program (Wisconsin GCG package, version The deduced 401 aa sequence was then used to search the protein database. Again, there were no existing matches, although there appeared to be a strong similarity to many members of the TNFR superfamily, most notably the human and mouse TNFR-2. A sequence alignment of this novel protein with known members of the TNFR-superfamily was prepared using the Pileup program, and then modified by PrettyPlot (Wisconsin GCG package, version This alignment shows a clear homology between the full length FRI-1 gene product and all other TNFR family members. The homologus region maps to the 31 extracellular domain of TNFR family members, and corresponds to the three or four cysteine-rich repeats found in the ligand binding domain of these proteins. This suggested that the FRI-1 gene encoded a novel TNFR family member. Since no transmembrane spanning region was detected we predicted that this may be a secreted receptor, similar to TNFR-1 derived soluble receptors (Kohno et al. Proc. Natl. Acad.
Sci. USA 87, 8331-8335 (1990)). Due to the apparent biological activity of the FRI-1 gene (vide infra), the product was named Osteoprotegerin (OPG).
EXAMPLE 2 OPG mRNA Expression Patterns in Tissues Multiple human tissue northern blots (Clonetech) were probed with a 3 2 P-dCTP labelled FRI-1 PCR product to detect the size of the human transcript and to determine patterns of expression. Northern blots were prehybridized in 5X SSPE, 50% formamide, Denhardt's solution, 0.5% SDS, and 100 mg/ml denatured salmon sperm DNA for 2 4 hr at 42 0 C. The blots were then hybridized in 5X SSPE, 50% formamide, 2X Denhardt's solution, 0.1% SDS, 100 mg/ml denatured salmon sperm DNA, and 5 ng/ml labelled probe for 18-24 hr at 42 0 C. The blots were then washed in 2X SSC for 10 min at RT, IX SSC for 10 min at 50 0 C, then in 0.5X SSC for 10-15 min.
Using a probe derived from the rat gene, a predominant mRNA species with a relative molecular mass of about 2.4 kb is detected in several tissues, including kidney, 20 liver, placenta, and heart. Highest levels are detected in the kidney. A large mRNA species of Mr 4.5 and 7.5 kb was detected in skeletal muscle and pancreas. In human fetal tissue, kidney was found to express relatively high levels of the 2.4 kb mRNA.
Using a human probe (vide infra), only the 2.4 kb transcript is detected in these same tissues. In addition, relatively high levels of the 2.4 kb transcript was detected in the lymph node, thymus, spleen and appendix. The size of the transcript detected by both the rat and human Osteosprotegerin gene is almost identical to the length of the rat pB 1.1 FRI-1 insert, suggesting it was a full length cDNA clone.
EXAMPLE 3 Systemic delivery of OPG in transgenic mice The rat OPG clone pB 1.1 was used as template to PCR amplify the coding region for subcloning into an ApoE-liver specific expression vector (Simonet et al. J. Clin.
32 Invest. 94, 1310-1319 (1994), and PCT Application No. US94/11675 and co-owned U.S.
Serial No. 08/221,767. The following 5' and 3' oligonucleotide primers were used for PCR amplification, respectively: -GACTAGTCCCACAATGAACAAGTGGCTGTG-3' (SEQ ID NO:9) 5' -ATAAGAATGCGGCCGCTAAACTATGAAACAGCCCAGTGACCATTC-3 (SEQ ID The PCR reaction mixture (Boehringer-Mannheim) was treated as follows: 94°C for 1 minute, 1 cycle; 94 0 C for 20 sec, 62 0 C for 30 sec, and 74 C for 1 minute, 25 cycles.
Following amplification, the samples were purified over Qiagen PCR columns and 10 digested overnight with Spel and NotI restriction enzymes. The digested products were extracted and precipitated and subcloned into the ApoE promoter expression vector.
Prior to microinjecting the resulting clone, HE-OPG, it was sequenced to ensure it was mutation-free.
The HE-OPG plasmid was purified through two rounds of CsCI density gradient 15 centrifugation. The purified plasmid DNA was digested with XhoI and Ase I, and the 3.6 kb transgene insert was purified by gel electrophoresis. The purified fragment was diluted to a stock injection solution of 1 mg/ml in 5 mM Tris, pH 7.4, 0.2 mM EDTA.
Single-cell embryos from BDF1 x BDFl-bred mice were injected essentially as described (Brinster et al., Proc. Natl. Acad. Sci. USA 82, 4338 (1985)), except that S 20 injection needles were beveled and siliconized before use. Embryos were cultured overnight in a C02 incubator and 15 to 20 2-cell embryos were transferred to the oviducts of pseudopregnant CD1 female mice.
Following term pregnancy, 49 offspring were obtained from implantation of microinjected embryos. The offspring were screened by PCR amplification of the integrated transgene in genomic DNA samples. The target region for amplification was a 369 bp region of the human Apo E intron which was included in the expression vector.
The oligos used for PCR amplification were: GCC TCT AGA AAG AGC TGG GAC-3' (SEQ ID NO:11) CGC CGT GTT CCA TTT ATG AGC-3' (SEQ ID NO:12) The conditions for PCR were: 94°C for 2 minute, 1 cycle; 94 0 C for 1 min, 63°C for 20 sec, and 72 0 C for 30 sec, 30 cycles. Of the 49 original offspring, 9 were identified as PCR positive transgenic founders.
33 At 8-10 weeks of age, five transgenic founders 11, 16, 17, and 28) and five controls 1, 12, 15, 18, and 30) were sacrificed for necropsy and pathological analysis.
Liver was isolated from the remaining 4 founders by partial hepatectomy. For partial hepatectomy, the mice were anesthetized and a lobe of liver was surgically removed.
Total cellular RNA was isolated from livers of all transgenic founders, and 5 negative control littermates as described (McDonald et al. Meth. Enzymol. 152, 219 (1987)).
Northern blot analysis was performed on these samples to assess the level oftransgene expression. Approximately 10ug of total RNA from each animal liver was resolved by electrophoresis denaturing gels (Ogden et al. Meth. Enzymol 152, 61 (1987)), then 10 transferred to HYBOND-N nylon membrane (Amersham), and probed with 3 2
P
dCTP-labelled pB 1.1 insert DNA. Hybridization was performed overnight at 42 0 C in .50% Formamide, 5 x SSPE, 0.5% SDS, 5 x Denhardt's solution, 100 mg/ml denatured salmon sperm DNA and 2-4 x 10- cpm of labeled probe/ml of hybridization butter.
Following hybridization, blots were washed twice in 2 x SSC, 0.1% SDS at room 15 temperature for 5 min each, and then twice in 0.1 x SSC, 0.1% SDS at 55 0 C for 5-10 min each. Expression of the transgene in founder and control littermates was determined following autoradiography.
The northern blot data indicate that 7 of the transgenic founders express detectable levels of the transgene mRNA (animal 2,1 1,16,17,22,33,and 45). The negative S 20 control mice and one of the founders expressed no transgene-related mRNA.
Since OPG is predicted to be a secreted protein, overexpression of transgene mRNA should be a proxy for the level of systemically delivered gene product. Of the PCR and northern blot positive mice, animal 2, 17 and 22 expressed the highest levels of transgene mRNA, and may show more extensive biological effects on host cells and tissues.
EXAMPLE 4 Biological activity of OPG Five of the transgenic mice (animals 2,11,16,17 and 28) and 5 control littermates (animals 1,12,15,18, and 30) were sacrificed for necropsy and pathological analysis using the following procedures: Prior to euthanasia, all animals had their identification numbers verified, then were weighed, anesthetized and blood drawn. The blood was saved as both serum and whole blood for a complete serum chemistry and hematology panel. Radiography was performed just after terminal anesthesia by lethal C02 inhalation, and prior to the gross dissection. Following this, tissues were removed and fixed in 10% buffered Zn- Formalin for histological examination. The tissues collected included the liver, spleen, pancreas, stomach, duodenum, ileum, colon, kidney, reproductive organs, skin and mammary glands, bone, brain, heart, lung, thymus, trachea, eosphagus, thyroid, jejunem, cecum, rectum, adrenals, urinary bladder, and skeletal muscle. Prior to fixation the 10 whole organ weights were determined for the liver, stomach, kidney, adrenals, spleen, and thymus. After fixation the tissues were processed into paraffin blocks, and 3 um sections were obtained. Bone tissue was decalcified using a formic acid solution, and all _______ections werestanined withhemntnoxyin nnl ein In addition taining with Gomori'., reticulin and Masson's trichrome were performed on certain tissues. Enzyme 15 histochemistry was performed to determine the expression of tartrate resistant acid S"phosphatase (TRAP), an enyzme highly expressed by osteoclasts, multinucleated boneresorbing cells of monocyte-macrophage lineage. Immunohistochemistry for BrdU and F480 monocyte-macrophage surface antigen was also performed to detect replicating cells and cells of the monocyte-macrophage lineage, respectively. To detect F480 20 surface antigen expression, formalin fixed, paraffin embedded 4pm sections were deparaffinized and hydrated to deionized water. The sections were quenched with 3% hydrogen peroxide, blocked with Protein Block (Lipshaw, Pittsburgh, PA), and incubated in rat monoclonal anti-mouse F480 (Harlan, Indianapolis, IN). This antibody was detected by biotinylated rabbit anti-rat immunoglobulins, peroxidase conjugated strepavidin (BioGenex San Ramon, CA) with DAB as chromagen (BioTek, Santa Barbara, CA). Sections were counterstained with hematoxylin.
Upon gross dissection and observation of visceral tissues, no abnormalities were found in the transgene expressors or control littermates. Analysis of organ weight indicate that spleen size increased by approximately 38% in the transgenic mice relative to controls. There was a slight enlargement of platelet size and increased circulating unstained cells in the transgene expressors. There was a marginal decrease in platelet levels in the transgene expressors. In addition, the serum uric acid, urea nitrogen, and alkaline phosphatase levels all trended lower in the transgene expressors. The expressors were found to have increased radiodensity of the skeleton, including long bones (femurs), vertebrae, and flat bones (pelvis). The relative size of femurs in the expressors were not different from the the control mice.
Histological analysis of stained sections of bone from the OPG expressors show severe osteopetrosis with the presence of cartilage remnants from the primary spongiosa seen within bone trabeculae in the diaphysis of the femur. A clearly defined cortex was not identifiable in the sections of femur. In normal animals, the central diaphysis is filled with bone marrow. Sections of vertebra also show osteopetrotic changes implying that the OPG-induced skeletal changes were systemic. The residual bone marrow showed predominantly myeloid elements. Megakaryocytes were present. Reticulin stains showed no evidence for reticulin deposition Imminnhistochemistry for F480, cell surface antigen expressed by cells of monocyte-macrophage derivation in the mouse, showed the presence of F480 positive cells in the marrow spaces. Focally, flattened F480 positive cells could be seen directly adjacent to trabecular bone surfaces.
S. The mesenchymal cells lining the bony trabeculae were flattened and appeared _inactive. Based on H&E and TRAP stains, osteoclasts were rarely found on the trabecular bone surfaces in the OPG expressors. In contrast, osteoclasts and/or 20 chondroclasts were seen in the region of the growth plate resorbing cartilage, but their numbers may be reduced compared to controls. Also, osteoclasts were present on the cortical surface of the metaphysis where modelling activity is usually robust. The predominant difference between the expressors and controls was the profound decrease in trabecular osteoclasts, both in the vertebrae and femurs. The extent of bone accumulation was directly correlated with the level of OPG transgene mRNA detected by northern blotting of total liver RNA.
The spleens from the OPG expressors had an increased amount of red pulp with the expansion due to increased hematopoiesis. All hematopoietic lineages are represented. F480 positive cells were present in both control and OPG expressors in the red pulp. Two of the expressors (2 and 17)had foci of extramedullary hematopoiesis within the liver and this is likely due to the osteopetrotic marrow.
36 There were no observable abnormalities in the thymus, lymph nodes, gastrointestinal tract, pancreato-hepatobiliary tract, respiratory tract, reproductive system, genito-urinary system, skin, nervous system, heart and aorta, breast, skeletal muscle and fat.
EXAMPLE Isolation of mouse and human OPG cDNA A cDNA clone corresponding to the 5' end of the mouse OPG mRNA was isolated from a mouse kidney cDNA library (Clontech) by PCR amplification. The oligonucleotides were derived from the rat OPG cDNA sequence and are shown below: 5'-ATCAAAGGCAGGGCATACTTCCTG-3' (SEQ ID NO:13) -GTTGCACTCCTGTTTCACGGTCTG-3' (SEQ ID NO:14) 5 -CAAGACACCTTGAAGGGCCTGATG-3' (SEQ ID 5' -TAACTTTTACAGAAGAGCATCAGC-3' (SEQ ID NO:16) 5' -AGCGCGGCCGCATGAACAAGTGGCTGTGCTGCG-3 (SEQ ID NO:17) 5' -AGCTCTAGAGAAACAGCCCAGTGACCATTCC-3' (SEQ ID NO:18) The partial and full-length cDNA products obtained in this process were sequenced. The full-length product was digested with Not I and Xba I, then directionally cloned into the plasmid vector pRcCMV (Invitrogen). The resulting plasmid was named pRcCMV-Mu-OPG. The nucleotide sequence of the cloned product was compared to the rat OPG cDNA sequence. Over the 1300 bp region spanning the OPG LORF, the rat and mouse DNA sequences are approximately 88% identical. The mouse cDNA sequence contained a 401 aa LORF, which was compared to the rat OPG protein sequence and found to be -94% identical without gaps. This indicates that the mouse cDNA sequence isolated encodes the murine OPG protein, and that the sequence and structure has been highly conserved throughout evolution. The mouse OPG protein sequence contains an identical putative signal peptide at its N-terminus, and all 4 potential sites of N-linked glycosylation are conserved.
A partial human OPG cDNA was cloned from a human kidney cDNA library using the following rat-specific oligonucleotides: AAG CTG TGC AAG AAC CTG ATG-3' (SEQ ID NO:19) 37 AAA GGC AGG GCA TAC TTC CTG-3' (SEQ ID This PCR product was sequenced and used to design primers for amplifying the 3' end of the human cDNA using a human OPG genomic clone in lambda as template: 5'-TCCGTAAGAAACAGCCCAGTGACC-3' (SEQ ID NO:29) 5'-CAGATCCTGAAGCTGCTCAGTTTG-3' (SEQ ID NO:21) The amplified PCR product was sequenced, and together with the 5' end sequence, was used to design 5' and 3' human-specific primers useful for amplifying the entire human OPG cDNA coding sequences: 5' -AGCGCGGCCGCGGGGACCACAATGAACAAGTTG-3' (SEQ ID NO:22) -AGCTCTAGAATTGTGAGGAAACAGCTCAATGGC-3' (SEQ ID NO: 23) The full-length human PCR product was sequenced, then directionally cloned into :..the plasmid vector pRcCMV (Invitrogen) using Not I and Xba I. The resulting plasmid was named pRcCMV-hnman OPG. The nucleotidP sequenceof hecloed- produ was compared to the rat and mouse OPG cDNA sequences. Over the 1300 bp region spanning the OPG LORF, the rat and mouse DNA sequences are approximately 78-88% identical to the human OPG cDNA. The human OPG cDNA sequence also contained a 401 aa LORF, and it was compared to the rat and mouse protein sequences. The predicted human OPG protein is approximatlely 85% identical, and -90% identical to the rat and mouse proteins, respectively. Sequence alignment of rat, mouse and human proteins show that they have been highly conserved during evolution. The human S" protein is predicted to have a N-terminal signal peptide, and 5 potential sites of N-linked glycosylation, 4 of which are conserved between the rat and mouse OPG proteins.
The DNA and predicted amino acid sequence of mouse OPG is shown in Figure 9A and 9B (SEQ ID NO: 122). The DNA and predicted amino acid sequence of human OPG is shown in Figure 9C an 9D (SEQ ID NO: 124). A comparison of the rat, mouse and human OPG amino acid sequences is shown in Figure 9E and 9F.
Isolation of additional human OPG cDNA clones revealed the presence of a G to C base change at position 103 of the DNA sequence shown in Figure 9C. This nucleotide change results in substitution of an asparagine for a lysine at position 3 of the amino acid sequence shown in Figure 9C. The remainder of the sequence in clones having this change was identical to that in Figure 9C and 9D.
38 EXAMPLE 6 OPG three-dimensional structure modelling The amino-terminal portion of OPG has homology to the extracellular portion of all known members of the TNFR superfamily (Figure 1C). The most notable motif in this region of TNFR-related genes is an -40 amino acid, cysteine-rich repeat sequence which folds into distinct structures (Banner et al. Cell 73, 431-445 (1993)). This motif is usually displayed in four (range 3-6) tandem repeats (see Figure 1C), and is known to be involved in ligand binding (Beutler and van Huffel Science 264, 667-663 (1994)). Each repeat usually contains six interspaced cysteine residues, which are involved in forming three intradomain disulfide bonds, termed SSI, SS2, and SS3 (Banner et al., ibid). In some receptors, such as TNFR2, CD30 and CD40, some of the repeat domains contain only two intrachain disulfide bonds (SS1 and SS3).
The human OPG protein sequence was aligned to a TNFRI1 extracellular domain--- profile using methods described by Luethy, et al., ibid, and the results were graphically 15 displayed using the PrettyPlot program from the Wisconsin Package, version 8.1 (Genetics Computer Group, Madison, WI) (Figure 10). The alignment indicates a clear conservation of cysteine residues involved in formation of domains 1-4. This alignment was then used to construct a three-dimensional model of the human OPG Nterminal domain using the known 3-D structure of the extracellular domain of S 20 TNFR1 (Banner et al., ibid) as the template. To do this the atomic coordinates of the peptide backbone and side chains of identical residues were copied from the crystal structure coordinates of TNFR1. Following this, the remaining coordinates for the insertions and different side chains were generated using the LOOK program (Molecular Applications Group, Palo Alto, CA). The 3-D model was then refined by minimizing its conformational energy using LOOK.
By analogy with other TNFR family members, it is assumed that OPG binds to a ligand. For the purpose of modelling the interaction of OPG with its ligand, the crystal structure of TNF-b was used to simulate a 3-D representation of an "OPG ligand". This data was graphically displayed (see Figure 11) using Molscript (Kraulis, J. Appl. Cryst.
24, 946-950, 1991). A model for the OPG/ligand complex with 3 TNFb and 3 OPG molecules was constructed where the relative positions of OPG are identical to TNFR1 in the crystal structure. This model was then used to find the residues of OPG that could interact with its ligand using the following approach: The solvent accessible area of all residues in the complex and one single OPG model were calculated. The residues that have different accessibility in the complex than in the monomer are likely to interact with the ligand.
The human and mouse OPG amino acid sequences were realigned using this information to highlight sequences comprising each of the cysteine rich domains 1-4 (Figure 12A and 12B). Each domain has individual structural characteristics which can be predicted: Domain 1 Contains 4 cysteines involved in SS2 (C41 to C54) and SS3 (C44 to C62) disulfide S. bonds. Although no SS1 bond is evident based on disulfide bridges, the conserved tyrosine at position 28 is homologous to Y20 in ThNFR1, which is known to be involvedin interacting with H66 to aid in domain formation. OPG has a homologous histidine at S 15 position 75, suggesting OPG Y28 and H75 stack together in the native protein, as do the homologous residues in TNFR1. Therefore, both of these residues may indeed be important for biological activity, and N-terminal OPG truncations up to and beyond Y28 may have altered activity. In addition, residues E34 and K43 are predicted to interact with a bound ligand based on our 3-dimensional model.
20 Domain 2 Contains six cysteines and is predicted to contain SS1 (C65 to C80), SS2 (C83 to C98) and SS3 (C87 to C105) disulfide bonds. This region of OPG also contains an region stretching from P66-Q91 which aligns to the portion of TNFR1 domain 2 which forms close contacts with TNFb (see above), and may interact with an OPG ligand. In particular residues P66, H68, Y69, Y70, T71, D72, S73, H75, T76, S77, D78, E79, L81, Y82, P85, V86, K88, E89, L90, and Q91 are predicted to interact with a bound ligand based on our structural data.
Domain 3 Contains 4 cysteines involved in SS1 (C107 to C 118) and SS3 (C124 to C142) disulfide bonds, but not an SS2 bond. Based on our structural data, residues El 15, LI 18 and K119 are predicted in to interact with an OPG ligand.
Domain 4 Contains 4 cysteines involved in SS1 (C145 to C160) and SS3 (C166 to C185) disulfide bonds, but not an SS2 bond, similar to domain 3. Our structural data predict that E153 and S155 interact with an OPG ligand.
Thus, the predicted structural model for OPG identifies a number of highly conserved residues which are likely to be important for its biological activity.
EXAMPLE 7 Production of recombinant secreted OPG protein in mammalian cells To determine if OPG is actually a secreted protein, mouse OPG cDNA was fused 10 to the human IgG1 Fc domain as a tag (Capon et al. Nature 337, 525-531 (1989)), and expressed in human 293 fibroblasts. Fc fusions were carried out using the vector pFc- A3. pFc-A3 contains the region encoding the Fc portion of human immunoglobulin gG-glheavy chUain r(E1son eVaraobido)arom-rnersramino acid ofthe hinge domain (Glu-99) to the carboxyl terminus and is flanked by a 5'-NotI fusion site and 3'-SalI and 15 XbaI sites. The plasmid was constructed by PCR amplification of the human spleen *o cDNA library (Clontech). PCR reactions were in a final volume of 100 ml and employed 2 units of Vent DNA polymerase (New England Biolabs) in 20 mM Tris-HCl (pH 8.8), 10 mM KCI, 10 mM (NH4)2SO4, 2 mM MgSO4, 0.1% Triton X-100 with 400 mM each dNTP and 1 ng of the cDNA library to be amplified together with 1 mM of each primer.
20 Reactions were initiated by denaturation at 95 0 C for 2 min, followed by 30 cycles of 0 C for 30 s, 55 0 C for 30 s, and 73 0 C for 2 min. The 5' primer ATAGCGGCCGCTGAGCCCAAATCTTGTGACAAAACTCAC 3' (SEQ ID NO:24) incorporated a NotI site immediately 5' to the first residue (Glu-99) of the hinge domain of IgG-gl. The 3'primer -TCTAGAGTCGACTTATCATTTACCCGGAGACAGGGAGAGGCTCTT-3 (SEQ ID incorporated SalI and XbaI sites. The 717-bp PCR product was digested with NotI and Sall, isolated by electrophoresis through 1% agarose (FMC Corp.),purified by the Geneclean procedure (BIO 101, Inc.) and cloned into NotI, SalI-digested pBluescript II KS vector (Stratagene). The insert in the resulting plasmid, pFc-A3, was sequenced to confirm the fidelity of the PCR reaction.
41 The cloned mouse cDNA in plasmid pRcCMV-MuOPG was amplified using the following two sets of primer pairs: Pair 1 -CCTCTGAGCTCAAGCTTCCGAGGACCACAATGAACAAG-3' (SEQ ID NO:26) 5'-CCTCTGCGGCCGCTAAGCAGCTTATTTTCACGGATTGAACCTG-3' (SEQ ID NO:27) Pair 2 -CCTCTGAGCTCAAGCTTCCGAGGACCACAATGAACAAG-3' (SEQ ID NO:28) CCTCTGCGGCCGCTGTTGCATTTCCTTTCTG-3' (SEQ ID S 10 The first pair amplifies the entire OPG LORF, and creates a NotI restriction site which is compatible with the in-frame Not I site in Fc fusion vector pFcA3. pFcA3 was :i prepared by engineering a NotI restriction site 5' to aspartic acid reside 216 of the human IgG1 Fc cDNA. This construct introduces a linker which encodes two irrelevant amino acids which span the junction between the OPG protein and the IgG Fc region. This 15 product, when linked to the Fc portion, would encode all 401 OPG residues directly followed by all 227 amino acid residues of the human IgGI Fc region (Fl.Fc). The second primer pair amplifies the DNA sequences encoding the first 180 amino acid residues of OPG, which encompasses its putative ligand binding domain. As above, the 3' primer creates an artificial Not I restriction site which fuses the C-terminal truncated S. 20 OPG LORF at position threonine 180 directly to the IgG1 Fc domain (CT.fc).
The amino acid sequence junction linking OPG residue 401 and aseptic acid residue 221 of the human Fc region can be modified as follows: The DNA encoding residues 216-220 of the human Fc region can be deleted as described below, or the cysteine residue corresponding to C220 of the human Fc region can be mutated to either serine or alanine. OPF-Fc fusion protein encoded by these modifed vectors can be transfected into human 293 cells, or CHO cells, and recombinant OPG-Fc fusion protein purified as described below.
Both products were directionally cloned into the plasmid vector pCEP4 (Invitrogen). pCEP4 contains the Epstein-Barr virus origin of replication, and is capable of episomal replication in 293-EBNA-1 cells. The parent pCEP4, and pCEP4-Fl.Fc and pCEP4-CT.Fc vectors were lipofected into 293-EBNA-1 cells using the manufacturer's recommended methods. The transfected cells were then selected in 100 mg/ml hygromycin to select for vector expression, and the resulting drug-resistant mass cultures were grown to confluence. The cells were then cultured in serum-free media for 72 hr, and the conditioned media removed and analysed by SDS-PAGE. A silver staining of the polyacrylamide gel detects the major conditioned media proteins produced by the drug resistant 293 cultures. In the pCEP4-Fl.Fc and the pCEP4-CT.Fc conditioned media, unique bands of the predicted sizes were abundantly secreted (see Figures 13B and 13C). The full-length Fc fusion protein accumulated to a high concentration, indicating that it may be stable. Both Fc fusion proteins were detected by anti-human IgG Fc antibodies (Pierce) on western blots, indicating that they are recombinant OPG 10 products.
The full length OPG-Fc fusion protein was purified by Protein-A column chromatography (Pierce) using the manufacturers recommended procedures. The protein was then sujected-toN-terminal sequence analysisoy automatedxEdmandegradation-as essentially described by Matsudaira et al. Biol. Chem. 262, 10-35 (1987)). The 15 following amino acid sequence was read after 19 cycles:
NH
2 -E T L P P KY L H Y D P E T G H Q L L-CO 2
H
(SEQ ID NO:31) This sequence was identical to the predicted mouse OPG amino acid sequence beginning at amino acid residue 22, suggesting that the natural mammalian leader 20 cleavage site is between amino acid residues Q21-E22, not between Y31-D32 as 0 originally predicted. The expression experiments performed in 293-EBNA cells with pCEP4-Fl.Fc and pCEP4-CT.Fc demonstrate that OPG is a secreted protein, and may act systemically to bind its ligand.
Procedures similar to those used to construct and express the muOPG[22-180]-Fc and muOPG[22-401]-Fc fusions were employed for additional mouse and human OPG-Fc fusion proteins.
Murine OPG cDNA encoding amino acids 1-185 fused to the Fc region of human IgG1 [muOPG Ct(185).Fc] was constructed as follows. Murine OPG cDNA from plasmid pRcCMV Mu Osteoprotegerin (described in Example 5) was amplified using the following primer pair in a polymerase chain reaction as described above: 1333-82: CTT GCC CTG ACC ACT CTT-3' (SEQ ID NO:32) 1333-80: CTG CGG CCG CAC ACA CGT TGT CAT GTG TTG C-3' (SEQ ID NO:33) This primer pair amplifies the murine OPG cDNA region encoding amino acid residues 63-185 (corresponding to bp 278-645) of the OPG reading frame as shown in Figure 9A. The 3' primer contains a Not I restriction site which is compatible with the in-frame Not I site of the Fc fusion vector pFcA3. The product also spans a unique EcoRI restriction site located at bp 436. The amplified PCR product was purified, cleaved with NotI and EcoRI, and the resulting EcoRI-NotI restriction fragment was purified. The vector pCEP4 having the murine 1-401 OPG-Fc fusion insert was cleaved with EcoRI and NotI, purified, and ligated to the PCR product generated above. The resulting pCEP4-based expression vector encodes OPG residues 1-185 directly followed by all 227 amino acid residues of the human IgG1 Fc region. The murine OPG 1-185.Fc fusion vector was transfected into 293 cells, drug selected, and conditioned media was produced as described above. The resulting secreted murine OPG 1-185.Fc fusion product was purified by Protein-A column chromatography (Pierce) using the manufacturers recommended procedures.
Murine OPG DNA encoding amino acid residues 1-194 fused to the Fc region of human IgGI (muOPG Ct(194).Fc) was constructed as follows. Mouse OPG cDNA from plasmid pRcCMV Mu-Osteoprotegerin was amplified using the following primer pairs: 1333-82: 5'-TCC CTT GCC CTG ACC ACT CTT-3' (SEQ ID NO:34) 1333-81: CTG CGG CCG CCT TTT GCG TGG CTT CTC TGT T-3' (SEQ ID This primer pair amplifies the murine OPG cDNA region encoding amino acid residues 70-194 (corresponding to bp 298-672) of the OPG reading frame. The 3' primer contains a Not I restriction site which is compatible with the in-frame Not I site of the Fc fusion vector pFcA3. The product also spans a unique EcoRI restriction site located at bp 436. The amplified PCR product was cloned into the murine OPG[1-401] Fc fusion vector as described above. The resulting pCEP4-based expression vector encodes OPG residues 1-194 directly followed by all 227 amino acid residues of the human IgG1 Fc region. The murine OPG 1-194.Fc fusion vector was transfected into 293 cells, drug 44 selected, and conditioned media was produced. The resulting secreted fusion product was purified by Protein-A column chromatography (Pierce) using the manufacturers recommended procedures.
Human OPG DNA encoding amino acids 1-401 fused to the Fc region of human IgGI was constructed as follows. Human OPG DNA in plasmid pRcCMV-hu osteoprotegerin (described in Example 5) was amplified using the following oligonucleotide primers: 1254-90: CTG AGC TCA AGC TTG GTT TCC GGG GAC CAC AAT G-3' (SEQ ID NO:36) 1254-95: CTG CGG CCG CTA AGC AGC TTA TTT TTA CTG AAT GG-3' (SEQ ID NO:37) The resulting PCR product encodes the full-length human OPG protein and creates a Not I restriction site which is compatible with the in-frame Not I site Fc fusion vector FcA3. The PCR product was directionally cloned into the plasmid vector pCEP4 as described above. The resulting expression vector encodes human OPG residues 1-401 directly followed by 227 amino acid residues of the human IgG1 Fc region. Conditioned media from transfected and drug selected cells was produced and the huOPG Fl.Fc fusion product was purified by Protein-A column chromatography (Pierce) using the manufacturers recommended procedures.
S* Human OPG DNA encoding amino acid residues 1-201 fused to the Fc region of human IgG1 [huOPG Ct(201).Fc] was constructed as follows. The cloned human OPG cDNA from plasmid pRrCMV-hu osteoprotegerin was amplified by PCR using the following oligonucleotide primer pair: 1254-90: CTG AGC TCA AGC TTG GTT TCC GGG GAC CAC AAT G-3' (SEQ ID NO:38) 1254-92: 5'-CCT CTG CGG CCG CCA GGG TAA CAT CTA TTC CAC-3' (SEQ ID NO:39) This primer pair amplifies the human OPG cDNA region encoding amino acid residues 1-201 of the OPG reading frame, and creates a Not I restriction site at the 3' end which is compatable with the in-frame Not I site Fc fusion vector FcA3. This product, when linked to the Fc portion, encodes OPG residues 1-201 directly followed by all 221 amino acid residues of the human IgG1 Fc region. The PCR product was directionally cloned into the plasmid vector pCEP4 as described above. Conditioned media from transfected and drug selected cells was produced, and the hu OPG Ct(201).Fc fusion products purified by Protein-A column chromatography (Pierce) using the manufacturer's recommended procedures.
The following procedures were used to construct and express unfused mouse and human OPG.
10 A plasmid for mammalian expression of full-length murine OPG (residues 1-401) *was generated by PCR amplification of the murine OPG cDNA insert from pRcCMV Mu-Osteoprotegerin and subcloned into the expression vector pDSRa (DeClerck et. atl.
J. Biol. Chem. 266, 3893 The ullu.......ollgucl.. primers wr sd 1295-26: 5'-CCG AAG CTT CCA CCA TGA ACA AGT GGC TGT GCT GC-3' (SEQ ID 1295-27: CTG TCG ACT ATT ATA AGC AGC TTA TTT TCA CGG ATT G-3' (SEQ ID NO:41) The murine OPG full length reading frame was amplified by PCR as described above. The PCR product was purified and digested with restriction endonucleases Hind III and Xba I (Boehringer Mannheim, Indianapolis, IN) under the manufacturers recommended conditions, then ligated to Hind III and Xba I digested pDSRa.
Recombinant clones were detected by restriction endonuclease digestion, then sequenced to ensure no mutations were produced during the PCR amplification steps.
The resulting plasmid, pDSRa-muOPG was introduced into Chinese hamster ovary (CHO) cells by calcium mediated transfection (Wigler et al. Cell 11, 233 (1977)).
Individual colonies were selected based upon expression of the dihydrofolate reductase (DHFR) gene in the plasmid vector and several clones were isolated. Expression of the murine OPG recombinant protein was monitored by western blot analysis of CHO cell conditioned media. High expressing cells were selected, and OPG expression was further amplified by treatment with methotrexate as described (DeClerck et al., idid).
Conditioned media from CHO cell lines was produced for further purification of recombinant secreted murine OPG protein.
A plasmid for mammalian expression of full-length human OPG (amino acids 1- 401) was generated by subcloning the cDNA insert in pRcCMV-hu Osteoprotegerin directly into vector pDSRa (DeClerck et al., ibid). The pRcCMV-OPG plasmid was digested to completion with Not I, blunt ended with Klenow, then digested to completion with Xba I. Vector DNA was digested with Hind III, blunt ended with Klenow, then digested with Xba I, then ligated to the OPG insert. Recombinant plasmids were then sequenced to confirm proper orientation of the human OPG cDNA.
10 The resulting plasmid pDSRa-huOPG was introduced into Chinese hamster ovary (CHO) cells as described above. Individual colonies were selected based upon expression of the dihydrofolate reductase (DHFR) gene in the plasmid vector and several clones were isolated. Expression of the numan OPG recombinant protein was monitoredby western blot analysis of CHO cell conditioned media. High expressing clones were selected, and OPG expression was further amplified by treatment with methotrexate.
Conditioned media from CHO cell lines expressing human OPG was produced for protein purification.
S. Expression vectors for murine OPG encoding residues 1-185 were constructed as follows. Murine OPG cDNA from pRcCMV-Mu OPG was amplified using the 20 following oligonucleotide primers: 1333-82: CTT GCC CTG ACC ACT CTT-3' (SEQ ID NO:42) 1356-12: CTG TCG ACT TAA CAC ACG TTG TCA TGT GTT GC-3' (SEQ ID NO:43) This primer pair amplifies the murine OPG cDNA region encoding amino acids 63-185 of the OPG reading frame (bp 278-645) and contains an artificial stop codon directly after the cysteine codon (C185), which is followed by an artificial Sal I restriction endonuclease site. The predicted product contains an internal Eco RI restriction site useful for subcloning into a pre-existing vector. After PCR amplification, the resulting purified product was cleaved with Eco RI and Sal I restriction endonucleases, and the large fragment was gel purified. The purified product was then subcloned into the large restriction fragment of an Eco RI and Sal I digest of pBluescript-muOPG Fl.Fc described above. The resulting plasmid was digested with Hind III and Xho I and the small fragment was gel purified. This fragment, which contains a open reading frame encoding residues 1-185 was then subcloned into a Hind III and Xho I digest of the expression vector pCEP4. The resulting vector, pmuOPG [1- 185], encodes a truncated OPG polypeptide which terminates at a cysteine residue located at position 185. Conditioned media from transfected and drug selected cells was produced as described above.
1333-82: 5'-TCC CTT GCC CTG ACC ACT CTT-3' (SEQ ID NO:44) 1356-13: CTG TCG ACT TAC TTT TGC GTG GCT TCT CTG TT-3' (SEQ ID This primer pair amplifies the murine OPG cDNA region encoding amino acids 70-194 of the OPG reading frame (bp 298-672) and contains an artificial stop codon directly after the lysine codon (K194), which is followed by an artificial Sal I restriction endonuclease site. The predicted product contains an internal Eco RI restriction site useful for subcloning into a pre-existing vector. After PCR amplification, the resulting purified product was cleaved with Eco RI and Sal I restriction endonucleases, and the large fragment was gel purified. The purified product was then subcloned into the large restriction fragment of an Eco RI and Sal I digest of pBluescript-muOPG Fl.Fc described above. The resulting plasmid was digested with Hind III and Xho I and the small fragment was gel purified. This fragment, which contains a open reading frame encoding residues 1-185 was then subcloned into a Hind III and Xho I digest of the expression vector pCEP4. The resulting vector, pmuOPG [1-185], encodes a truncated OPG polypeptide which terminates at a lysine at position 194. Conditioned media from transfected and drug selected cells was produced as described above.
Several mutations were generated at the 5' end of the huOPG [22-401]-Fc gene that introduce either amino acid substitutions, or deletions, of OPG between residues 22 3 0 through 32. All mutations were generated with the "QuickChange T Site-Directed Mutagenesis Kit" (Stratagene, San Diego, CA) using the manfacturer's recommended conditions. Briefly, reaction mix containing huOPG [22-401]-Fc plasmid DNA template and mutagenic primers were treated with Pfu polymerase in the presence of deoxynucleotides, then amplified in a thermocycler as described above. An aliqout of the reaction is then transfected into competent E. coli XL1-Blue by heatshock, then plated.
Plasmid DNA from transformants was then sequenced to verify mutations.
The following primer pairs were used to delete residues 22-26 of the human OPG gene, resulting in the production of a huOPG [27-401]-Fc fusion protein: 1436-11: ACC ACC CAG AAG TAC CTT CAT TAT GAC-3' (SEQ ID NO:140) 1436-12: 5'-GTC ATA ATG AAG GTA CTT CTG GGT GGT CCA-3' (SEQ ID NO:141) The following primer pairs were used to delete residues 22-28 of the human OPG gene, resulting in the production of a huOPG [29-401]-Fc fusion protein: 1436-17: 5'-GGA CCA CCC AGC TTC ATT ATG ACG AAG AAA C-3' (SEQ ID NO:142) 1436-18: 5'-GTT TCT TCG TCA TAA TGA AGC TGG GTG GTC C-3' (SEQ ID NO:143) The following primer pairs were used to delete residues 22-31 of the human OPG gene, resulting in the production of a huOPG [32-401]-Fc fusion protein: 1436-27: GAC CAC CCA GGA CGA AGA AAC CTC TC-3' (SEQ ID NO:144) 1436-28: AGG TTT CTT CGT CCT GGG TGG TCC AC-3' (SEQ ID NO:145) The following primer pairs were used to change the codon for tyrosine residue 28 to phenylalanine of the human OPG gene, resulting in the production of a huOPG [22- 401]-Fc Y28F fusion protein: 1436-29: TTC CTC CAA AGT TCC TTC ATT ATG AC-3' (SEQ ID NO:146) 1436-30: ATA ATG AAG GAA CTT TGG AGG AAA CG-3' (SEQ ID NO:147) The following primer pairs were used to change the codon for proline residue 26 to alanine of the human OPG gene, resulting in the production of a huOPG [22-401]-Fc P26A fusion protein: 1429-83: AAC GTT TCC TGC AAA GTA CCT TCA TTA TG-3 (SEQ ID NO:148) 1429-84: AAT GAA GGT ACT TTG CAG GAA ACG TTT CC-3' (SEQ ID NO:149) Each resulting muOPG [22-401 ]-Fc plasmid containing the appropriate mutation S 15 was then transfected into human 293 cells, the mutant OPG-Fc fusion protein purified from conditioned media as described above. The biological activity of each protein was S[ assessed the in vitro osteoclast forming assay described in Example 11.
-EXAMPLE 8 Expression of OPG in E. coli o 20 A. Bacterial Expression Vectors pAMG21 The expression plasmid pAMG21 can be derived from the Amgen expression *vector pCFM1656 (ATCC #69576) which in turn be derived from the Amgen expression vector system described in US Patent No. 4,710,473. The pCFM1656 plasmid can be derived from the described pCFM836 plasmid (Patent No. 4,710,473) by: destroying the two endogenous NdeI restriction sites by end filling with T4 polymerase enzyme followed by blunt end ligation; replacing the DNA sequence between the unique AatII and Clal restriction sites containing the synthetic PL promoter with a similar fragment obtained from pCFM636 (patent No. 4,710,473) containing the PL promoter AatII
CTAATTCCGCTCTCACCTACCAAACAATGCCCCCCTGCAAAAAATAAATTCATAT-
3 'TGCAGATTAAGGCGAGAGTGGATGGTTTGTTACGGGGGGACGTTTTTTATTTAAGTATA-
-AAAAAACATACAGATAACCATCTGCGGTGATATTATCTCTGGCGGTGTTGACATJA
TTTTTTGTATGTCTATTGGTAGAGCGACTATTTAATAGAGACGGACAJAGTGTATTT -TAGGAGTGGGGTGATACTGAGCAGAT 3' (SEQ ID NO:53) -ATGGTGACCGCCACTATGACTCGTGTAGC51 (SEQ ID NO:54) GlaI and then substituting the small DNA sequence between the unique ClI! and KpnI restriction sites with the following oligonucleotide: CGATTTGATTCTAGAAGGAGGAATAACATATGGTTAACGCGTTGGAATTCGGTAC31 (SEQ ID NO:48) 3' TAAACTAAGATCTTCCTCCTTATTGTATACAATTGCGCAACCTTAJAGC (SEQ ID NO:49) C laI KpnI The expression plasmid pAMG2l can then be derived from pCFMI165 6 by making a series of site directed base changes by PCR overlapping oligo mutagenesis and DNA a. ~~sequence substitutions trigwt h gl ie(lsiib 8)imdaeyYt the plasmid replication promoter PcopB and proceeding toward the plasmid replication genes, the base pair changes are as follows: *PAMG21 bT) bp in PCFM1656 bip changed to in pAMG21 204 T/A G/G 428 A/T G/G 509 G/C A/T 617 insert two G/G bp 679 G/G T/A 980 T/A G/G 994 G/G A/T 1004 A/T G/G 1007 G/G T/A 1028 A/T T/A 1047 G/G T/A 1178 G/G T/A 1466 G/G T/A 2028 G/G bp deletion 2187 G/G T/A 2480 A/T T/A 2499-2SO2 35 49-202AGTG GTGA 51 TCAC CAGT 2642 TCCGAGC 7 bp deletion
AGGCTCG
3435 G/C A/T 3446 G/C A/T 3643 A/T T/A The DNA sequence between the unique AatII (position #4364 in pCFM 1656) and SaclI (position #45 85 in pCFM 1656) restriction sites is substituted with the following DNA sequence: [AatII sticky end] 51 GCGTAACGTATGCATGGTCTCC- (position #4358 in pAMG2l) 3' TGCACGCATTGCATACGTACCAGAGG- -GGAGCCCACCTGAW(51'(C2TATTTATTTTCTTTCCGAGTCAGCTTTCTGA GGGCCTTTCGTTTTATCTGTTGTTTGTCGGTGAACGCTCTCCTGAGTAGGACAAATCCGC -CCCGAAGCAAATAGiACAACAA.CACCTTCAAACTCATCCTGTTTAGGCG CGGGAGCGGATTTGAACGTTGCGAAGCAACGGCCCGGAGGGTGGCGGGCAGGACGCCCGC
-GCCCTCGCCTAAACTTGCAACGCTTCGTTGCCGGGCCTCCCACCGCCCGTCCTGCGGGCG-
GTATTTGACGGTCCGTAGTTTAATTCGTCTTCCGGTAGGACTGCCTACCGGAAAAACGCA-
Aat II TTCTACAAACTCTTTTGTTTATTTTTCTAAATACATTCAAATATGGACGTCGTACTTAAC -AAGATGTTTGAGAAAACAAATAAAAAGATTTATGTAAGTTTATACCTGCAGCATGAATTG TTTTAA.AGTATGGGCAATCAATTGCTCCTGTTAAAATTGCTTTAGAAATACTTTGGCAGC
-AAAATTTCATACCCGTTAGTTAACGAGGACAATTTTAACGAAATCTTTATGAAACCGTCG-
GGTTTGTTGTATTGAGTTTCATTTGCGCATTGGTTAAATGGAAAGTGACCGTGCGCTTAC CCAAACAACATAACTCAAAGTAAACGCGTAACCAATTTACCTTTCACTGGCACGCGAATG TACAGCCTAATATTTTTGAAATATCCCAAGAGCTTTTTCCTTCGCATGCCCACGCTAAAC 52
-AGCGTAAAATTAAGTCCAAAGAGGAGGGGTT-
-ATTCTTTTTCTCTTTTGGTTAAATCGTTGTTTGATTTATTATTTGCTATATTTATTTTTC TAAGAAAAAGAGAAAACCAATTTAGCAACAAACTAATAATAAACGATATAAATAAAAAG
GAATACATGGAGAATAAGTTTCTCCCTTAAT-
CTATTAATAGTTGATCTCTTCCTTGTTATTACCATACAAJGTATGTGCGTACATTTTTAT AACTATCTATATAGTTGTCTTTCTCTGAATGTGCAAAACTAAGCATTCCGA.ZGCCATTAT TTGATAGATATATCAACAGAAAGAGACTTACACGTTTTGATTCGTAGGCTTCGGTAATA -TACGAGAAGGACAACATGTAACGTATTGTCT -ATCGTCATACTTATCCCTTTGATTTGGGTCACTATTCTGGACTACTAGCGAAGAATT 1 5 -TT~CTTGGTTTATAA~TCTTAiTTTCATATGT AATGTAAACCTCTAAAAAATAAATGTCGTAACAAAGTTTATATAAGGTTAATTAGCCAC AATGATTGGAGTTAGAATAATCTACTATAGGATCATATTTTATTAAATTAGCGTCATCAT TTACTAACCTCAATCTTATTAGATGATATCCTAGTATAAAATAATTTAATCGCAGTAGTA *e -AATATTGCCTCCATTTTTTAGGGTAATTATCCAGAATTGAAATATCAGATTTp.ACCATAG TTATAACGGAGGTAAAAAATCCCATTAATAGGTCTTAACTTTATAGTCTAAATTGGTATC AATGAGGATAAATGATCGCGAGTAAATAATATTCACAATGTACCATTTTAGTCATATCAG 25 -TTACTCCTATTTACTAGCGCTCATTTATTATAAGTGTTACATGGTpAATCAGTATAGTC- -ATAAGCATTGATTAATATCATTATTGCTTCTACAGGCTTTAATTTTATTAATTATTCTGT TATTCGTAACTAATTATAGTAATAACGAAGATGTCCGA
TATTATTATAGACA-
-AAGTGTCGTCGGCATTTATGTCTTTCATACCCATCTCTTTATCCTTACCTATTGTTTGTC-
TTCACAGCAGCCGTAAATACAGAAAGTATGGGTAGAGAATAGGAATGGATAACAAACAG
GCAAGTTTTGCGTGTTATATATCATTAACGGTAATAGATTGACATTTGATTCTATAA-
CGTTCAA ACGCACAATATATAGTAATTTTGCCATTATCTAJACTGTAACTAAGATTATT ATTGGATTTTTGTCACACTATTATATCGCTTGAATACAATTGTTTAACATAAGTACCTG TAACCTAAAAACAGTGTGATAATATAGCGACTTTATGTTACAATGTATTCATGGAC TAGGATCGTACAGGTTTACGCAAGAAAATGGTTTGTTATAGTCGATTAATCGATTTGATT
ATCCTAGCATGTCCAAATGCGTTCTTTTACCAACAATATCAGCTAATTAGCTAAACTAA-
CTAGATTTGTTTTAACTAATTAAAGGAGGAATAACATATGGTTAACGCGTTGGAATTCGA-
GATCTAAACAAAATTGATTAATTTCCTCCTTATTGTATACCAATTGCGCAACCTTAAGCT SaclI GCTCACTAGTGTCGACCTGCAGGGTACCATGGAAGCTTACTCGAGGATCCGCGGAAGAjA- CGAGTGATCACAGCTGGACGTCCCATGGTACCTTCGAATGAGCTCCTAGGCGCCTTTCTT GAAGAAGAAGAAGAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCpjATA- CTTCTTCTTCTTCTTTCGGGCTTTCCTTCGACTCAACCGACGACGGTGGCGACTCGTTAT ACTAGCATAACCCCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGG 15 TGATCGTATTGGGGAACCCCGGAGATTTGCCCAGAACTCCCCAAAAAACGACTTTCCTCC- -AACCGCTCTTCACGCTCTTCACGC 3' (SacII sticky end] (SEQ ID *-TTGGCGAGAAGTGCGAGAAGTG 5' (position #5904 in pAMG21) (SEQ ID NO:46) During the ligation of the sticky ends of this substitution DNA sequence, the 20 outside AatII and Sacll sites are destroyed. There are unique AatII and Sacll sites in the substituted DNA.
pAMG22-His The expression plasmid pAMG22-His can be derived from the Amgen expression vector pAMG22 by substituting the small DNA sequence between the unique NdeI( #4795) and EcoRi #4818) restriction sites of pAMG22 with the following oligonucleotide duplex: Nde I NheI EcoRI TATGAAACATCATCACCATCACCATCATGCTAGCGTTAACGCGTTGG 3' (SEQ ID NO:51) 3' ACTTTGTAGTAGTGGTAGTGGTAGTACGATCGCAATTGCGCAACCTTAA (SEQ ID NO:52) MetLysHisHisHisHi sHi sllisHisAlaSerValAsnAlaLeuGlu (SEQ ID NO:168) DAMG22 The expression plasmid pAMG22 can be derived from the Amgen expression vector pCFMl656 (ATCC #69576) which in turn be derived from the Amgen expression vector system described in US Patent No. 4,710,473 granted December 1, 1987. The pCFM1656 plasmid can be derived from the described pCFM836 plasmid (Patent No.
4,710,473) by: destroying the two endogenous NdeI restriction sites by end filling with T4 polymerase enzyme followed by blunt end ligation; replacing the DNA sequence between the unique AatII and Clal restriction sites containing the synthetic PL promoter with a similar fragment obtained from pCFM636 (patent No. 4,710,473) containing the PL promoter AatII
CTAATTCCGCTCTCACCTACCAAACAATGCCCCCCTGCAAAAAATAAATTCATAT-
3 'TGCAGATTAAGGCGAGAGTGGATGGTTTGTTACGGGGGGACGTTTTTTATTTAAGTATA-
-AAAAAACATACAGATAACCATCTGCGGTGATAAATTATCTCTGGCGGTGTTGACATAAA-
-TTTTTTGTATGTCTATTGGTAGACGCCACTATTTAATAGAGACCGCCACAACTGTATTT-
-TACCACTGGCGGTGATACTGAGCACAT 3' (SEQ ID NO:53) -ATGGTGACCGCCACTATGACTCGTGTAGC5 (SEQ ID NO:54) 20 Clal and then substituting the small DNA sequence between the unique Clal and KpnI restriction sites with the following oligonucleotide: CGATTTGATTCTAGAAGGAGGAATAACATATGGTTAACGCGTTGGAATTCGGTAC 3' (SEQ ID 3 TAAACTAAGATCTTCCTCCTTATTGTATACCAATTGCGCAACCTTAAGC (SEQ ID NO:56) Clal KpnI The expression plasmid pAMG22 can then be derived from pCFM1656 by making a series of site directed base changes by PCR overlapping oligo mutagenesis and DNA sequence substitutions. Starting with the BglII site (plasmid bp 180) immediately 5' to the plasmid replication promoter PcopB and proceeding toward the plasmid replication genes, the base pair changes are as follows: PAMv'G22 bp bip in PCFM1656 bip changed to in PAMG22 204 T/A C/G 428 A/T G/C 509 G/C A/T 617 insert two G/C bp 679 G/C T/A 980 T/A C/G 994 G/C A/T 1004 A/T C/G 1007 C/G T/A 1028 A/T T/A 1047 C/G T/A 1178 G/C T/A 15 146 u/uT/A 2028 G/C bp deletion 2187 C/G T/A 2480 A/T T/A 2499-2502 AGTG GTCA 20 TCAC CAGT 2642 TCCGAGC 7 bp deletion
AGGCTCG
3435 G/C A/T 3446 G/C A/T 3643 A/T T/A The DNA sequence between the unique AatII (position #43 64 in pCFM 165 6) and SucII (position #4585 in pCFM 1656) restriction sites is substituted with the following DNA sequence: [AatII sticky end] (position #4358 in pAMG22) 5' GCGTAACGTATGCATGGTCTCCCCATGCGAGAGTAGGGAACTGCCAGGCATCAA- 3' TGCACGCATTGCATACGTACCAGAGGGGTACGCTCTCATCCCTTGACGGTCCGTAGTT-
ATAAAACGAAAGGCTCAGTCGAAAGACTGGGCCTTTCGTTTTATCTGTTGTTTGTCGGTG-
TATTTTGCTTTCCGAGTCAGCTTTCTGACCCGGAAAGCAAAATAGACAACAAACAGCCAC -AACGCTCTCCTGAGTAGGACAAATCCGCCGGGAGCGGATTTGAACGTTGCGAJAGCACGG TTGCGAGAGGACTCATCCTGTTTAGGCGGCCCTCGCCTAA-ACTTGCAACGCTTCGTTGCC CCCGGAGGGTGGCGGGCAGGACGCCCGCCATA AACTGCCAGGCATCAAATTAAGCAGAAG GGGCCTCCCACCGCCCGTCCTGCGGGCGGTATTTGACGGTCCGTAGTTTAATTCGTCTTC GCCATCCTGACGGATGGCCTTTTTGCGTTTCTACAAACTCTTTTGTTTATTTTTCT.AT CGGTAGGACTGCCTACCGGAAAAACGCAAGATGTTTGAGAAAACAATA3AGATTTA- AatII -ACATTCAAATATGGACGTCTCATAATTTTTAATTCATTTGACAATGCTAAAATTC TGTAAGTTTATACCTGCAGAGTATTAAAAATTTTTTAAGTAAACTGTTTACGATTTTAAG TTGATTAATATTCTCAATTGTGAGCGCTCACAATTTATCGATTTGATTCTAGATTTGTTT AACTAATTATAAGAGTTAACACTCGCGAGTGTTAAATAGCTAAACTAAGATCTAAACTCA ow:. TAACTAATTAAAGGAGGAATAACATATGGTTAACGCGTTGGAATTCGAGCTCACTAGTGT
-ATTGATTAATTTCCTCCTTATTGTATACCAATTGCGCAACCTTAAGCTCGAGTGATCACA-
SacII CGACCTGCAGGGTACCATGGAAGCTTACTCGAGGATCCGCGGAAGAAGAAGAAGAkGAA- GCTGGACGTCCCATGGTACCTTCGAATGAGCTCCTAGGCGCCTTTCTTCTTCTTCTTCTT GAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACC CTTTCGGGCTTTCCTTCGACTCAACCGACGACGGTGGCGACTCGTTATTGATCGTATTGG
CCTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACCGCTCTTCA-
-GGAACCCCGGAGATTTGCCCAGAACTCCCCAAAAAACGACTTTCCTCCTTGGCGAGAAGT-
-CGCTCTTCACGC 3' (SEQ ID NO:58) -GCGAGAAGTG 5' (SEQ ID NO:57) [SaclI sticky end] (position #5024 in pAMG22) 57 During the ligation of the sticky ends of this substitution DNA sequence, the outside AatII and SacII sites are destroyed. There are unique AatII and SacII sites in the substituted DNA.
B. Human OPG Metr32-401] In the example, the expression vector used was pAMG21, a derivative of pCFM1656 (ATCC accession no. 69576) which contains appropriate restriction sites for insertion of genes downstream from the lux PR promoter. (See U.S. Patent No.
5,169,318 for description of the lux expression system). The host cell used was GM120 (ATCC accession no. 55764). This host has the lacIQ promoter and lacI gene integrated into a second site in the host chromosome of a prototrophic E. coli K12 host. Other commonly used E. coli expression vectors and host cells are also suitable for expression.
A DNA sequence coding for an N-terminal methionine and amino acids 32-401 of the human OPG polypeptide was placed under control of the iuxPR promoter in the plasmid expression vector pAMG21 as follows. To accomplish this, PCR using 15 oligonucleotides #1257-20 and #1257-19 as primers was performed using as a template plasmid pRcCMV-Hu OPG DNA containing the human OPG cDNA and thermocycling for 30 cycles with each cycle being: 94C for 20 seconds, followed by 37 0 C for o seconds, followed by 72 0 C for 30 seconds. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, and restricted with KpnI and S. 20 BamHI restriction endonucleases and purified. Synthetic oligonucleotides #1257-21 and #1257-22 were phophorylated individually using T4 polynucleotide kinase and ATP, and o were then mixed together, heated at 94 0 C and allowed to slow cool to room temperature to form an oligonucleotide linker duplex containing NdeI and KpnI sticky ends. The phosphorylated linker duplex formed between oligonucleotides #1257-21 and #1257-22 containing NdeI and KpnI cohesive ends (see Figure 14A) and the KpnI and BamHI digested and purified PCR product generated using oligo primers #1257-20 and #1257- 19 (see above) was directionally inserted between two sites of the plasmid vector pAMG21, namely the NdeI site and BamHI site, using standard recombinant DNA methodology (see Figure 14A and sequences below). The synthetic linker utilized E.
3 0 coli codons and provided for a N-terminal methionine.
58 Two clones were selected and plasmid DNA isolated, and the human OPG insert was subsequently DNA sequence confirmed. The resulting pAMG21 plasmid containing amino acids 32-401 of the human OPG polypeptide immediately preceded in frame by a methionine is referred to as pAMG21-huOPG met[32-401] or pAMG21huOPG met[32-401].
Oligo#1257-19 -TACGCACTGGATCCTTATAAGCAGCTTATTTTTACTGATTGGAC-3' (SEQ ID NO:59) Oligo#1257-20 -GTCCTCCTGGTACCTACCTAAAACAAC-3' (SEQ ID Oligo#1257-21
TATGGATGAAGAAACTTCTCATCAGCTGCTGTGTGATAAATGTCC
15 GCCGGGTAC (SEQ ID NO:61) Oligo#1257-22 -CCGGCGGACATTTATCACACAGCAGCTGATGAGAAGTTTCTTCATCCA-3 (SEQ ID NO:47) Cultures of pAMG21-huOPG met[32-401] in E. coli GM120 in 2XYT media containing 20 mg/ml kanamycin were incubated at 30 0 C prior to induction. Induction of S" huOPG met[32-401] gene product expression from the luxPR promoter was achieved following the addition of the synthetic autoinducer N-(3-oxohexanoyl)-DL-homoserine 25 lactone to the culture media to a final concentration of 30 ng/ml and cultures were incubated at either 30 0 C or 37 0 C for a further 6 hours. After 6 hours, the bacterial cultures were examined by microscopy for the presence of inclusion bodies and were then pelletted by centrifugation. Refractile inclusion bodies were observed in induced cultures indicating that some of the recombinant huOPG met[32-401] gene product was produced insolubly in E. coli. Some bacterial pellets were resuspended in 10mM Tris- HCl/pH8, ImM EDTA and lysed directly by addition of 2X Laemlli sample buffer to IX final, and b-mercaptoethanol to 5% final concentration, and analyzed by SDS-PAGE. A substantially more intense coomassie stained band of approximately 42kDa was observed on a SDS-PAGE gel containing total cell lysates of 30°C and 37°C induced 59 cultures versus lane 2 which is a total cell lysate of a 30 0 C uninduced culture (Figure 14B). The expected gene product would be 370 amino acids in length and have an expected molecular weight of about 42.2 kDa. Following induction at 37 0 C for 6 hours, an additional culture was pelleted and either processed for isolation of inclusion bodies (see below) or processed by microfluidizing. The pellet processed for microfluidizing was resuspended in 25mM Tris-HCl/pH8, 0.5M NaCl buffer and passed 20 times through a Microfluidizer Model 1108 (Microfluidics Corp.) and collected. An aliquot was removed of the collected sample (microfluidized total lysate), and the remainder was pelleted at 20,000 x g for 20 minutes. The supernatant following centrifugation was removed (microfluidized soluble fraction) and the pellet resuspended in a 25mM Tris- HCl/pH8, 0.5M NaCI, 6M urea solution (microfluidized insoluble fraction). To an aliquot of either the total soluble, or insoluble fraction was added to an equal volume of 2X Laemalli sample bufftter and b-mercaptoethanol to 5% final concentiation. The samples were then analyzed by SDS-PAGE. A significant amount of recombinant 15 huOPG met[32-401] gene product appeared to be found in the insoluble fraction. To purify the recombinant protein inclusion bodies were purified as follows: Bacterial cells were separated from media by density gradient centrifugation in a Beckman J-6B centrifuge equipped with a JS-4.2 rotor at 4,900 x g for 15 minutes at 4 0 C. The bacterial pellet was resuspended in 5 ml of water and then diluted to a final volume of 10 ml with 20 water. This suspension was transferred to a stainless steel cup cooled in ice and subjected to sonic disruption using a Branson Sonifier equipped with a standard tip (power setting=5, duty cycle=95%, 80 bursts). The sonicated cell suspension was centrifuged in a Beckman Optima TLX ultracentrifuge equipped with a TLA 100.3 rotor at 195,000 x g for 5 to 10 minutes at 23 0 C. The supernatant was discarded and the pellet rinsed with a stream of water from a squirt bottle. The pellets were collected by scraping with a micro spatula and transferred to a glass homogenizer (15 ml capacity). Five ml of Percoll solution (75% liquid Percoll, 0.15 M sodium chloride) was added to the homogenizer and the contents are homogenized until uniformly suspended. The volume was increased to 19.5 ml by the addition of Percoll solution, mixed, and distributed into 3 Beckman Quick-Seal tubes (13 x 32 mm). Tubes were sealed according to manufacturers instructions. The tubes were spun in a Beckman TLA 100.3 rotor at 23°C, 20,000 rpm (21,600 x 30 minutes. The tubes were examined for the appropriate banding pattern. To recover the refractile bodies, gradient fractions were recovered and pooled, then diluted with water. The inclusion bodies were pelleted by centrifugation, and the protein concentration estimated following SDS-PAGE.
An aliquot of inclusion bodies isolated as described below was dissolved into 1X Laemlli sample buffer with 5% b-mercaptoethanol and resolved on a SDS-PAGE gel and the isolated inclusion bodies provide a highly purified recombinant huOPG[32-401] gene product. The major -42 kDa band observed after resolving inclusion bodies on a SDSpolyacrylamide gel was excised from a separate gel and the N-terminal amino acid sequence determined essentially as described (Matsudaira et al. J. Biol. Chem. 262, (1987)). The following sequence was determined after 19 cycles: 'NH2 -MDEETSHQLLCDKCPPGTY-COOH (SEQ ID NO:62) 11 This sequenc was found to be idetical to thefirst inoidsencodedbythe pAMG21 Hu-OPG met[32-401] expression vector, produced by a methionine residue 15 provided by the bacterial expression vector.
C. Human OPG met[22-4011 A DNA sequence coding for an N-terminal methionine and amino acids 22 through 401 of human OPG was placed under control of the luxPR promoter in a prokaryotic plasmid expression vector pAMG21 as follows. Isolated plasmid DNA of S 20 pAMG21-huOPG met[32-401] (see Section B) was cleaved with KpnI and BamHI restriction endonucleases and the resulting fragments were resolved on an agarose gel.
The B fragment (-1064 bp fragment) was isolated from the gel using standard methodology. Synthetic oligonucleotides (oligos) #1267-06 and #1267-07 were phosphorylated individually and allowed to form an oligo linker duplex, which contained NdeI and KpnI cohesive ends, using methods described in Section B. The synthetic linker duplex utilized E. coli codons and provided for an N-terminal methionine. The phosphorylated oligo linker containing NdeI and KpnI cohesive ends and the isolated -1064 bp fragment of pAMG21-huOP met[32-401] digested with KpnI and BamHI restriction endonucleases were directionally inserted between the Ndel and BamHI sites of pAMG21 using standard recombinant DNA methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the 61 manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the huOPG-met[22-401] gene.
Oligo #1267-06 -TAT GGA AAC TTT TCC TCC AAA ATA TCT TCA TTA TGA TGA AGA AAC TTC TCA TCA GCT GCT GTG TGA TAA ATG TCC GCC GGG TAC-3' (SEQ ID NO:63) Oligo #1267-07 GCG GAC ATT TAT CAC ACA GCA GCT GAT GAG AAG TTT CTT CAT CAT AAT GAA GAT ATT TTG GAG GAA AAG TTT CCA-3' (SEQ ID NO:64) Cultures of pAMG21-huOPG-met[22-401] in E. coli host 393 were placed in S2XYT media containing 20 pg/ml kanamycin and were incubated at 30 0 C prior to 15 induction. Induction of recombinant gene product expression from the luxPR promoter of vector pAMG21 was achieved following the addition of the synthetic autoinducer N- (3-oxohexanoyl)-DL-homoserine lactone to the culture media to a final concentration of 30 ng/ml and incubation at either 30°C or 37°C for a further 6 hours. After 6 hours, bacterial cultures were pelleted by centrifugation (=30 0 C 1+6 or 37°C Bacterial cultures were also either pelleted just prior to induction (=30 0 C Prel) or alternatively no autoinducer was added to a separate culture which was allowed to incubate at 30 0 C for a further 6 hours to give an uninduced (UI) culture (=30°C UI). Bacterial pellets of either 0 C Prel, 30 0 C UI, 30 0 C 1+6, or 37 0 C 1+6 cultures were resuspended, lysed, and analyzed by SDS-polyacrylamide gel electrophoresis (PAGE) as described in Section B.
Polyacrylamide gels were either stained with coomassie blue and/or Western transferred to nitrocellulose and immunoprobed with rabbit anti-mu OPG-Fc polyclonal antibody as described in Example 10. The level of gene product following induction compared to either an uninduced (30 0 C UI) or pre-induction (30 0 C Prel) sample.
D. Murine OPG met[22-4011 A DNA sequence coding for an N-terminal methionine and amino acids 22 through 401 of the murine (mu) OPG (OPG) polypeptide was placed under control of the luxPR promoter in a prokaryotic plasmid expression vector pAMG21 as follows. PCR was performed using oligonucleotides #1257-16 and #1257-15 as primers, plasmid pRcCMV-Mu OPG DNA as a template and thermocycling conditions as described in Section B. The PCR product was purified and cleaved with KpnI and BamHI restriction endonucleases as described in Section B. Synthetic oligos #1260-61 and #1260-82 were phosphorylated individually and allowed to form an oligo linker duplex with Ndel and KpnI cohesive ends using methods described in Section B. The synthetic linker duplex utilized E. coli codons and provided for an N-terminal methionine. The phosphorylated linker duplex formed between oligos #1260-61 and #1260-82 containing NdeI and KpnI cohesive ends and the KpnI and BamHI digested and purified PCR product generated using oligo primers #1257-16 and #1257-15 were directionally inserted between the NdeI and BamnHI sites of pAMG21 using standard methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verity the NA sequence oe u G me[22-4] ge.
Expression of recombinant muOPG met[22-401] polypeptide from cultures of 393 15 cells harboring plasmid pAMG21-MuOPG met[22-401] following induction was determined using methods described in Section C.
Oligo #1257-15 GCA CTG GAT CCT TAT AAG CAG CTT ATT TTC ACG GAT 20 TGA AC-3' (SEQ ID Oligo #1257-16 5'-GTG CTC CTG GTA CCT ACC TAA AAC AGC ACT GCA CAG TG- 3' (SEQ ID NO:66) Oligo #1260-61 GGA AAC TCT GCC TCC AAA ATA CCT GCA TTA CGA TCC GGA AAC TGG TCA TCA GCT GCT GTG TGA TAA ATG TGC TCC GGG TAC-3' (SEQ ID NO:67) Oligo #1260-82 GAG CAC ATT TAT CAC ACA GCA GCT GAT GAC CAG TTT CCG GAT CGT AAT GCA GGT ATT TTG GAG GCA GAG TTT CCA-3' (SEQ ID NO:68) E. Murine OPG met[32-401] A DNA sequence coding for an N-terminal methionine and amino acids 32 through 401 of murine OPG was placed under control of the luxPR promoter in a prokaryotic plasmid expression vector pAMG21 as follows. To accomplish this, Synthetic oligos #1267-08 and #1267-09 were phosphorylated individually and allowed to form an oligo linker duplex using methods described in Section B. The synthetic linker duplex utilized E. coli codons and provided for an N-terminal methionine. The phosphorylated linker duplex formed between oligos #1267-08 and #1267-09 containing NdeI and KpnI cohesive ends, and the KpnI and BamHI digested and purified PCR product described earlier (see Section was directionally inserted between the NdeI and BamHI sites of pAMG21 using standard methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencig was performed to verify the DNA sequence of the muOPG-met[32-401] gene.
15 Expression of recombinant muOPG-met [32-401] polypeptide from cultures of 393 cells harboring the pAMG21 recombinant plasmid following induction was determined using methods described in Section C.
Oligo #1267-08 5'-TAT GGA CCC AGA AAC TGG TCA TCA GCT GCT GTG TGA TAA 20 ATG TGC TCC GGG TAC-3' (SEQ ID NO:69) Oligo #1267-09 GAG CAC ATT TAT CAC ACA GCA GCT GAT GAC CAG TTT SCTG GGT CCA-3' (SEQ ID F. Murine OPG met-lys[22-4011 A DNA sequence coding for an N-terminal methionine followed by a lysine residue and amino acids 22 through 401 of murine OPG was placed under control of the lux PR promoter in prokaryotic expression vector pAMG21 as follows. Synthetic oligos #1282-95 and #1282-96 were phosphorylated individually and allowed to form an oligo linker duplex using methods described in Section B. The synthetic linker duplex utilized E. coli codons and provided for an N-terminal methionine. The phosphorylated linker duplex formed between oligos #1282-95 and #1282-96 containing NdeI and KpnI 64 cohesive ends and the KpnI and BamHI digested and purified PCR product described in Section D was directionally inserted between the NdeI and BamHI sites in pAMG21 using standard methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the MuOPG--Met-Lys[22-401] gene.
Expression of recombinant MuOPG Met-Lys[22-401] polypeptide from transformed 393 cells harboring the recombinant pAMG21 plasmid following induction was determined using methods described in Section C.
Oligo #1282-95 GAA AGA AAC TCT GCC TCC AAA ATA CCT GCA TTA CGA TCC GGA AAC TGG TCA TCA GCT GCT GTG TGA TAA ATG TGC TCC GGG TAC-3' (SEQ ID NO:71) Oligo #1282-96 5'-CCG GAG CAC ATT TAT CAC ACA GCA GCT GAT GAC CAG TTT CCG GAT CGT AAT GCA GGT ATT TTG GAG GCA GAG TTT CTT TCA-3' (SEQ ID NO:72) 20 G. Murine OPG met-lvs-(his)7r22-4011 A DNA sequence coding for N-terminal residues Met-Lys-His-His-His-His-His- His-His (=MKH) followed by amino acids 22 through 401 of Murine OPG was placed under control of the lux PR promoter in prokaryotic expression vector pAMG21 as follows. PCR was performed using oligonucleotides #1300-50 and #1257-15 as primers and plasmid pAMG21 -muOPG-met[22-401 DNA as template. Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, cleaved with NdeI and BamHI restriction endonucleases and purified. The NdeI and BamHI digested and purified PCR product generated using oligo primers #1300-50 and #1257-15 was directionally inserted between the NdeI and BamHI sites ofpAMG21 using standard DNA methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing performed to verify the DNA sequence of the muOPG-MKH[22-401] gene.
Expression of recombinant MuOPG-MKH[22-401] polypeptide from transformed 393 cultures harboring the recombinant pAMG21 plasmid following induction was determined using methods described in Section C.
Oligo #1300-50 5'-GTT CTC CTC ATA TGA AAC ATC ATC ACC ATC ACC ATC ATG AAA CTC TGC CTC CAA AAT ACC TGC ATT ACG AT-3' (SEQ ID NO:73) Oligo #1257-15 (see Section D) H. Murine OPG met-lvsr22-4011(his)7 A DNA sequence coding for a N-terminal met-lys, amino acids 22 through 401 murine OPG, and seven histidine residues following amino acid 401 (=muOPG MK[22- 401]-H7), was placed under control of the lux PR promoter in prokaryotic expression vector pAMG21 as follows. PCR was performed using oligonucleotides #1300-49 and 15 #1300-51 as primers and pAMG21-muOPG met[22-401] DNA as template.
Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, restricted with Ndel and BamHI restriction endonucleases, and purified. The NdeI and BamHI digested and purified PCR product was directionally inserted between the NdeI and BamHI sites 20 in pAMG21 using standard methodology. The ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the muOPG MK[22-401]-H7 gene.
Expression of the recombinant muOPG MK-[22-401]-H7 polypeptide from a transformed 393 cells harboring the recombinant pAMG21 plasmid following induction was determined using methods described in Section C.
Oligo #1300-49 5'-GTT CTC CTC ATA TGA AAG AAA CTC TGC CTC CAA AAT ACC TGC A-3' (SEQ ID NO:74) Oligo #1300-51 66 GCA CTG GAT CCT TAA TGA TGG TGA TGG TGA TGA TGT AAG CAG CTT ATT TTC ACG GAT TGA ACC TGA TTC CCT A-3' (SEQ ID I. Murine OPG met[27-4011 A DNA sequence coding for a N-terminal methionine and amino acids 27 through 401 of murine OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. PCR was performed with oligonucleotides #1309-74 and #1257-15 as primers and plasmid pAMG21-muOPG-met[22-401] DNA as template. Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, cleaved with NdeI and BamHI restriction endonucleases, and purified. The NdeI and BamHI digested and purified PCR product was directionally inserted between the NdeI and BamHI sites of pAMG21 using standard methodology. The ligation mixture was 15 transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the muOPG-met[27-401] gene.
Expression of recombinant muOPG-met[27-401 polypeptide from a transfected 393 culture harboring the recombinant pAMG21 plasmid following induction was 20 determined using methods described in Section C.
Oligo#1309-74 5'-GTT CTC CTC ATA TGA AAT ACC TGC ATT ACG ATC CGG AAA S. CTG GTC AT-3' (SEQ ID NO:76) Oligo#1257-15 (See Section D) J. Human OPG metr27-4011 A DNA sequence coding for a N-terminal methionine and amino acids 27 through 401 of human OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. PCR was performed using oligonucleotides #1309-75 and #1309-76 as primers and plasmid pAMG21-huOPG-met[22-401] DNA as template. Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, restricted with Asel and BamHI restriction endonucleases, and purified. The Asel and BamHI digested and purified PCR product above was directionally inserted between the NdeI and BamHI sites ofpAMG21 using standard methodology. The ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the huOPG-met[27-401] gene.
Expression of the recombinant huOPG-met[27-401] polypeptide following induction of from transfected 393 cells harboring the recombinant pAMG21 plasmid was determined using methods described in Section C.
'Oligo #1309-/75 5'-GTT CTC CTA TTA ATG AAA TAT CTT CAT TAT GAT GAA GAA 15 ACT T-3' (SEQ ID NO:77) Oligo #1309-76 5'-TAC GCA CTG GAT CCT TAT AAG CAG CTT ATT TTT ACT GAT T-3' (SEQ ID NO:78) K. Murine OPG met[22-1801 A DNA sequence coding for a N-terminal methionine and amino acids 22 through 180 of murine OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. PCR was performed with oligonucleotides #1309-72 and #1309-73 as primers and plasmid pAMG21-muOPG-met[22-401] DNA as template. Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the PCR product was excised, purified, restricted with NdeI and BamHI restriction endonucleases, and purified. The NdeI and BamHI digested and purified PCR product above was directionally inserted between the NdeI and BamHI sites of pAMG21 using standard methodology. The ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the muOPG-met[22-180] gene.
Expression of recombinant muOPG-met[22-180] polypeptide from transformed 393 cultures harboring the recombinant pAMG21 plasmid following induction was determined using methods described in Section C.
Oligo #1309-72 5'-GTT CTC CTC ATA TGG AAA CTC TGC CTC CAA AAT ACC TGC A-3' (SEQ ID NO:79) Oligo #1309-73 GCA CTG GAT CCT TAT GTT GCA TTT CCT TTC TGA ATT AGC A-3' (SEQ ID L. Murine OPG metf27-1801 A DNA sequence coding for a N-terminal methionine and amino acids 27 through 180 of muirinP OPG was placed under the rcntrol of the lux PR promoter of prokaryotic 15 expression vector pAMG21 as follows. PCR was performed using oligonucleotides #1309-74 (see Section I) and #1309-73 (see Section K) as primers and plasmid pAMG21-muOPG met[22-401] DNA as template. Thermocycling conditions were as described in Section B. The resulting PCR sample was resolved on an agarose gel, the €o S PCR product excised, purified, restricted with NdeI and BamHI restriction endonucleases, and purified. The NdeI and BamHI digested and purified PCR product above was directionally inserted between the NdeI and BamHI sites in pAMG21 using S* standard methodology. The ligation mixture was transformed into E. coli host 393 by SS.. electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of the muOPG met[27-180] gene.
Expression of recombinant muOPG met[27-180] polypeptide from cultures of transformed 393 cells harboring the recombinant pAMG21 plasmid following induction was determined using methods described in Section C.
M. Murine OPG metr22-1891 and metr22-1 94 1 A DNA sequence coding for a N-terminal methionine and either amino acids 22 through 189, or 22 through 194 ofmurine OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. The pair of synthetic oligonucleotides #1337-92 and #1337-93 (=muOPG-189 linker) or #1333-57 and #1333- 58 (=muOPG-194 linker) were phosphorylated individually and allowed to form an oligo linker duplex pair using methods described in Section B. Purified plasmid DNA of pAMG21-muOPG-met[22-401] was cleaved with KpnI and BspEl restriction endonucleases and the resulting DNA fragments were resolved on an agarose gel. The -413 bp B fragment was isolated using standard recombinant DNA methodology. The phosphorylated oligo linker duplexes formed between either oligos #1337-92 and #1337- 93 (muOPG-189 linker) or oligos #1333-57 and #1333-58 (muOPG-194 linker) containing BspEI and BamHI cohesive ends, and the isolated -413 bp B fragment of plasmid pAMG21 -muOPG-met[22-401 digested with KpnI and BspEI restriction endonucleases above, was directionally inserted between the KpnI and BamHI sites of pAMG21-muOPG met[22-401] using standard methodology. Each ligation mixture was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of either the muOPG-met[22-189] or muOPGmet[22-194] genes.
Expression of recombinant muOPG-met[22-189] and muOPG-met[22-194] polypeptides from recombinant pAMG21 plasmids transformed into 393 cells was determined using methods described in Section C.
Oligo #1337-92 5'-CCG GAA ACA GAT AAT GAG-3' (SEQ ID NO:81) Oligo #1337-93 CCT CAT TAT CTG TTT-3' (SEQ ID NO:82) Oligo #1333-57 GAA ACA GAG AAG CCA CGC AAA AGT AAG-3' (SEQ ID NO:83) Oligo #1333-58 CCT TAC TTT TGC GTG GCT TCT CTG TTT-3' (SEQ ID NO:84) N. Murine OPG met[27-18 9 1 and metr27-194] A DNA sequence coding for a N-terminal methionine and either amino acids 27 through 189, or 27 through 194 of murine OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. Phosphorylated oligo linkers either "muOPG-189 linker" or "muOPG-194 linker" (see Section M) containing BspEI and BamHI cohesive ends, and the isolated -413 bp B fragment of plasmid pAMG21-muOPG-met[22-401] digested with KpnI and BspEl restriction endonucleases were directionally inserted between the KpnI and BamHI sites of plasmid pAMG21muOPG-met[27-401] using standard methodology. Each ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of either the muOPG met[27-189] or muOPG met[27-194] genes.
•Expression of recombinant muOPG met[27-189] and muOPG met[27-194] following induction of 393 cells harboring recombinant pAMG21 plasmids was determined using methods described in Section C.
O. Human OPG metr22-1851, met[22-1891, met[22-194] A DNA sequence coding for a N-terminal methionine and either amino acids 22 through 185, 22 through 189, or 22 through 194 of the human OPG polypeptide was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 20 as follows. The pair of synthetic oligonucleotides #1331-87 and #1331-88 (=huOPG- :185 linker), #1331-89 and #1331-90 (=huOPG-189 linker), or #1331-91 #1331-92 (=huOPG-194 linker) were phosphorylated individually and each allowed to form an oligo linker duplex pair using methods described in Section B. Purified plasmid DNA of pAMG21-huOPG-met[27-401] was restricted with KpnI and NdeI restriction endonucleases and the resulting DNA fragments were resolved on an agarose gel. The -407 bp B fragment was isolated using standard recombinant DNA methodology. The phophorylated oligo linker duplexes formed between either oligos #1331-87 and #1331- 88 (huOPG-185 linker), oligos #1331-89 and #1331-90 (huOPG-189 linker), or oligos #1331-91 and #1331-92 (huOPG-194 linker)[each linker contains NdeI and BamHI cohesive ends], and the isolated -407 bp B fragment of plasmid pAMG21-huOPGmet[27-401] digested with KpnI and Ndel restriction endonucleases above, was 71 directionally inserted between the KpnI and BamHI sites of plasmid pAMG21-huOPGmet[22-401] using standard methodology. Each ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA was isolated, and DNA sequencing was performed to verify the DNA sequence of either the huOPG-met[22-185], huOPG-met[22-189], or huOPG-met[22- 194] genes.
Expression of recombinant huOPG-met[22-185], huOPG-met[22-189] or huOPGmet[22-194] in transformed 393 cells harboring recombinant pAMG21 plasmids following induction was determined using methods described in Section C.
Oligo #1331-87 GTT AAT GAG-3' (SEQ ID Oligo #1331-88 5'-GAT CCT CAT TAA CA-3' (SEQ ID N0:86) Oligo #1331-89 GTT CCG GAA ACA GTT AAG-3' (SEQ ID NO:87) 20 Oligo #1331-90 S 5'-GAT CCT TAA CTG TTT CCG GAA CA-3' (SEQ ID NO:88) Oligo #1331-91 5'-TAT GTT CCG GAA ACA GTG AAT CAA CTC AAA AAT AAG-3' (SEQ ID NO:89) Oligo #1331-92 CCT TAT TTT TGA GTT GAT TCA CTG TTT CCG GAA CA- 3' (SEQ ID P. Human OPG met[27-1851. met[27-1891. met [27-1941 A DNA sequence coding for a N-terminal methionine and either amino acids 27 through 185, 27 through 189, or 27 through 194 of the human OPG polypeptide was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 72 as follows. Phosphorylated oligo linkers "huOPG-185 linker", "huOPG-189 linker", or "huOPG-194 linker" (See Section O) each containing Ndel and BamHI cohesive ends, and the isolated -407 bp B fragment of plasmid pAMG21 -huOPG-met[27-401] digested with KpnI and NdeI restriction endonucleases (See Section O) were directionally inserted between the KpnI and BamHI sites of plasmid pAMG21-huOPG-met[27-401] (See Section J) using standard methodology. Each ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA isolated, and DNA sequencing performed to verify the DNA sequence of either the huOPG-met[27-185], huOPG-met[27-189], or huOPG-met[27-194] genes.
Expression of recombinant huOPG-met[27-185], huOPG-met[27-189], and huOPG-met[27-194] from recombinant pAMG21 plasmids transformed into 393 cells was determined using methods described in Section C.
0 Murine OPi metl27-4UI (P33E. G36S. A DNA sequence coding for an N-terminal methionine and amino acids 27 15 through 48 of human OPG followed by amino acid residues 49 through 401 of murine OPG was placed under control of the lux PR promoter of prokaryotic expression vector pAMG21 as follows. Purified plasmid DNA of pAMG21-huOPG-met[27-401] (See Section J) was cleaved with AatII and KpnI restriction endonucleases and a -1075 bp B ifragment isolated from an agarose gel using standard recombinant DNA methodology.
Additionally, plasmid pAMG21 -muOPG-met[22-401] DNA (See Section D) was digested with KpnI and BamHI restriction endonucleases and the -1064 bp B fragment isolated as described above. The isolated -1075 bp pAMG21-huOPG-met[27-401] restriction fragment containing AatII KpnI cohesive ends (see above), the -1064 bp pAMG21 -muOPG-met[22-401] restriction fragment containing KpnI and BamHI sticky ends and a -5043 bp restriction fragment containing AatII and BamHI cohesive ends and corresponding to the nucleic acid sequence of pAMG21 between AatII BamHI were ligated using standard recombinant DNA methodology. The ligation was transformed into E. coli host 393 by electroporation utilizing the manufacturer's protocol. Clones were selected, and the presence of the recombinant insert in the plasmid verified using standard DNA methodology. muOPG-27-401 (P33E, G36S, gene. Amino acid changes in muOPG from proline-33 to glutamic acid-33, 73 glycine-36 to serine-36, and alanine-45 to proline-45, result from replacement of muOPG residues 27 through 48 with huOPG residues 27 through 48.
Expression of recombinant muOPG-met[27-401] (P33E, G36S, A45P) from transformed 393 cells harboring the recombinant pAMG21 plasmid was determined using methods described in Section C.
R. Murine OPG met-lys-(his)7-ala-ser-(asp)4-lvs[22-4011 A DNA sequence coding for an N-terminal His tag and enterokinase recognition sequence which is (NH2 to COOH terminus): Met-Lys-His-His-His-His-His-His-His- Ala-Ser-Asp-Asp-Asp-Asp-Lys followed by amino acids 22 through 401 of the murine OPG polypeptide was placed under control of the lac repressor regulated Ps4 promoter as follows. pAMG22-His (See Section A) was digested with NheI and BamHI restriction endonucleases, and the large fragment (the A fragment) isolated from an agarose gel using standard recombinant DNA methodology. Oligonucleotides #1282-91 and #1282-92 were phosphorylated individually and allowed to form an oligo linker 15 duplex using methods previously described (See Section The phosphorylated linker duplex formed between oligos #1282-91 and #1282-92 containing NheI and KpnI cohesive ends, the KpnI and BamHI digested and purified PCR product described (see Section and the A fragment of vector pAMG22-His digested with NheI and BamHI were ligated using standard recombinant DNA methodology. The ligation was transformed into E. coli host GM120 by electroporation utilizing the manufacturer's protocol. Clones were selected, plasmid DNA isolated and DNA sequencing performed to verify the DNA sequence of the muOPG-HEK[22-401] gene. DNA sequencing revealed a spurious mutation in the natural muOPG sequence that resulted in a single amino acid change of Alanine-45 of muOPG polypeptide to a Threonine.
Expression of recombinant muOPG-HEK[22-401] (A45T) from GM120 cells harboring the recombinant pAMG21 plasmid was determined using methods similar to those described in Section C, except instead of addition of the synthetic autoinducer, IPTG was added to 0.4 mM final to achieve induction.
Oligo #1282-91 5'-CTA GCG ACG ACG ACG ACA AAG AAA CTC TGC CTC CAA AAT ACC TGC ATT ACG ATC CGG AAA CTG GTC ATC AGC TGC TGT GTG ATA AAT GTG CTC CGG GTA C-3' (SEQ ID NO:91) Oligo #1282-92 GAG CAC ATT TAT CAC ACA GCA GCT GAT GAC CAG TTT CCG GAT CGT AAT GCA GGT ATT TTG GAG GCA GAG TTT CTT TGT CGT CGT CGT CG-3' (SEQ ID NO:92) S. Human OPG met-arg-glv-ser-(his)6[22-4011 Eight oligonucleotides (1338-09 to 1338-16 shown below) were designed to produce a 175 base fragment as overlapping, double stranded DNA. The oligos were annealed, ligated, and the 5' and 3' oligos were used as PCR primers to produce large quantities of the 175 base fragment. The final PCR gene products were digested with restriction endonucleases Clal and KpnI to yield a fragment which replaces the Nterminal 28 codons of human OPG. The Clal and KpnI digested PCR product was inserted into pAMG21-huOPG [27-401] which had also been cleaved with Clal and KpnI. Ligated DNA was transformed into competent host cells of E. coli strain 393.
Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. Protein expression levels were determined from 50 ml shaker flask studies. Whole cell lysate and sonic pellet were analyzed for expression of the construct by Coomassie stained PAGE gels 2 0 and Western analysis with murine anti-OPG antibody. Expression of huOPG Met-Arg- Gly-Ser-(His)6 [22-401] resulting in the formation of large inclusion bodies and the protein was localized to the insoluble (pellet) fraction.
1338-09 ACA AAC ACA ATC GAT TTG ATA CTA GA (SEQ ID NO:93) 1338-10 TTT GTT TTA ACT AAT TAA AGG AGG AAT AAA ATA TGA GAG GAT CGC ATC AC (SEQ ID NO:94) 1338-11 CAT CAC CAT CAC GAA ACC TTC CCG CCG AAA TAC CTG CAC TAC GAC GAA GA (SEQ ID 1338-12 AAC CTC CCA CCA GCT GCT GTG CGA CAA ATG CCC GCC GGG TAC CCA AAC A (SEQ ID NO:96) 1338-13 TGT TTG GGT ACC CGG CGG GCA TTT GT (SEQ ID NO:97) r r rr 1338-14 CGC ACA GCA GCT GGT GGG AGG TTT CTT CGT CGT AGT GCA GGT ATT TCG GC (SEQ ID NO:98) 15 1338-15 r GGG AAG GTT TC TAT T (SEQ ID NO:99) G TGA TGG TGA TGG TGA TGC GAT CCT CTC ATA TTT a 1338-16 CCT CCT TTA ATT AGT TAA AAC AAA TCT AGT ATC AAA TCG ATT GTG TTT GT (SEQ ID NO:100) T. Human OPG met-lys[22-4011 and met(lys)3[22-4011 To construct the met-lys and met-(lys)3 versions of human OPG[22-401], overlapping oligonucleotides were designed to add the appropriate number of lysine residues. The two oligos for each construct were designed to overlap, allowing two rounds of PCR to produce the final product. The template for the first PCR reaction was a plasmid DNA preparation containing the human OPG 22-401 gene. The first PCR added the lysine residue(s). The second PCR used the product of the first round and added sequence back to the first restriction site, Clal.
The final PCR gene products were digested with restriction endonucleases Clal and KpnI, which replace the N-terminal 28 codons of hu OPG, and then ligated into plasmid pAMG21-hu OPG [27-401] which had been also digested with the two restriction endonucleases. Ligated DNA was transformed into competent host cells of E coli strain 393. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. Protein expression levels were determined from 50 ml shaker flask studies. Whole cell lysate and sonic pellet were analyzed for expression of the construct by Coomassie stained PAGE gels and Western analysis with murine anti-OPG antibody. Neither construct had a detectable level of protein expression and inclusion bodies were not visible. The DNA sequences were confirmed by DNA sequencing.
Oligonucleotide primers to prepare Met-Lys huOPG[22-401]: 1338-17 SACA AAC ACA ATC GAT TTG ATA CTA GAT TTG TTT TAA CTA ATT AAA GGA GGA ATA AAA TG (SEQ ID NO:101) 1338-18 15 CTA ATT AAA GGA GGA ATA AAA TGA AAG AAA CTT TTC CTC CAA AAT ATC (SEQ ID NO:102) *e 1338-20 TGT TTG GGT ACC CGG CGG ACA TTT ATC ACA C (SEQ ID NO:103) S* Oligonucleotide primers to prepare Met-(Lys)3-huOPG[22- 401] *.1338-17 S" ACA AAC ACA ATC GAT TTG ATA CTA GAT TTG TTT TAA CTA ATT AAA GGA GGA ATA AAA TG (SEQ ID NO:104) 1338-19 CTA ATT AAA GGA GGA ATA AAA TGA AAA AAA AAG AAA CTT TTC CTC CAA AAT ATC (SEQ ID NO:105) 1338-20 TGT TTG GGT ACC CGG CGG ACA TTT ATC ACA C (SEQ ID NO:106) U. Human and Murine OPG [22-4011/Fc Fusions 77 Four OPG-Fc fusions were constructed where the Fc region of human IgGI was fused at the N-terminus of either human or murine Osteoprotegerin amino acids 22 to 401 (referred to as Fc/OPG [22-401]) or at the C-terminus (referred to as OPG[22- 401]/Fc). Fc fusions were constructed using the fusion vector pFc-A3 described in Example 7.
All fusion genes were constructed using standard PCR technology. Template for PCR reactions were plasmid preparations containing the target genes. Overlapping oligos were designed to combine the C-terminal portion of one gene with the N terminal portion of the other gene. This process allows fusing the two genes together in the correct reading frame after the appropriate PCR reactions have been performed. Initially one "fusion" oligo for each gene was put into a PCR reaction with a universal primer for the vector carrying the target gene. The complimentary "fusion" oligo was used with a :universal primer to PCR the other gene. At the end of this first PCR reaction, two separate products were obtained, with each individual gene having the fusion site present, creating enough overlap to drive the second round of PCR and create the desired fusion. In the second round of PCR, the first two PCR products were combined along with universal primers and via the overlapping regions, the full length fusion DNA *.sequence was produced.
The final PCR gene products were digested with restriction endonucleases Xbal and BamHI, and then ligated into the vector pAMG21 having been also digested with the two restriction endonicleases. Ligated DNA was transformed into competent host cells of E. coli strain 393. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence.
Protein expression levels were determined from 50 ml shaker flask studies. Whole cell lysate, sonic pellet, and supernatant were analyzed for expression of the fusion by Coomassie stained PAGE gels and Western analysis with murine anti-OPG antibody.
Fc/huOPG [22-4011 Expression of the Fc/hu OPG [22-401] fusion peptide was detected on a Coomassie stained PAGE gel and on a Western blot. The cells have very large inclusion 78 bodies, and the majority of the product is in the insoluble (pellet) fraction. The following primers were used to construct this OPG-Fc fusion: 1318-48 CAG CCC GGG TAA AAT GGA AAC GTT TCC TCC AAA ATA TCT TCA TT (SEQ ID NO:107) 1318-49 CGT TTC CAT TTT ACC CGG GCT GAG CGA GAG GCT CTT CTG CGT GT (SEQ ID NO:108) Fc/muOPG [22-4011 Expression of the fusion peptide was detected on a Coomassie stained gel and on a Western blot. The cells have very large inclusion bodies, and the majority of the product is in the insoluble (pellet) fraction. The following primers were used to construct this S* 15 OPG-Fc fusion: i 1318-50 CGC TCA GCC CGG GTA AAA TGG AAA CGT TGC CTC CAA AAT ACC TGC (SEQ ID NO:109) 1318-51 CCA TTT TAC CCG GGC TGA GCG AGA GGC TCT TCT GCG TGT (SEQ ID NO:110) muOPG [22-4011/Fc Expression of the fusion peptide was detected on a Coomassie stained gel and on a Western blot. The amount of recombinant product was less than the OPG fusion proteins having the Fc region in the N terminal position. Obvious inclusion bodies were not detected. Most of the product appeared to be in the insoluble (pellet) fraction. The following primers were used to construct this OPG-Fc fusion: 1318-54 GAA AAT AAG CTG CTT AGC TGC AGC TGA ACC AAA ATC (SEQ ID NO:111) 1318-55 CAG CTG CAG CTA AGC AGC TTA TTT TCA CGG ATT G (SEQ ID NO:112) huOPG r22-4011/Fc Expression of the fusion peptide was not detected on a Coomassie stained gel, although a faint Western positive signal was present. Obvious inclusion bodies were not detected. The following primers were used to prepare this OPG-Fc fusion: 1318-52 AAA AAT AAG CTG CTT AGC TGC AGC TGA ACC AAA ATC (SEQ ID NO:113) 15 CAG CTG CAG CTA AGC AGC TTA TTT TTA CTG ATT GG (SEQ ID NO:114) V. Human OPG met[22-4011-Fc fusion This construct combines a proline to alanine amino acid change at position 20 (P25A) with the huOPG met[22-401]-Fc fusion. The plasmid was digested with Srestriction endonucleases Clal and KpnI, which removes the N-terminal 28 codons of the gene, and the resulting small (less than 200 base pair) fragment was gel purified. This fragment containing the proline to alanine change was then ligated into plasmid pAMG21-huOPG [22-401]-Fc fusion which had been digested with the two restriction endonucleases. The ligated DNA was transformed into competent host cells of E. coli strain 393. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. Protein expression levels were determined from 50 ml shaker flask studies. Whole cell lysate and sonic pellet were analyzed for expression of the construct by Coomassie stained PAGE gels and Western analysis with murine anti-OPG antibody. The expression level of the fusion peptide was detected on a Coomassie stained PAGE gel and on a Western blot. The protein was in the insoluble (pellet) fraction. The cells had large inclusion bodies.
I
W. Human OPG metr22-4011 A DNA sequence coding for an N-terminal methionine and amino acids 22 through 401 of human OPG with the proline at position 25 being substituted by alanine under control of the lux PR promoter in prokaryotic expression vector pAMG21 was constructed as follows: Synthetic oligos 1289-84 and 1289-85 were annealed to form an oligo linker duplex with XbaI and KpnI cohesive ends. The synthetic linker duplex utilized optimal E. coli codons and encoded an N-terminal methionine. The linker also included an Spel restriction site which was not present in the original sequence. The linker duplex was directionally inserted between the XbaI and KpnI sites in pAMG21- 10 huOPG-22-401 using standard methods. The ligation mixture was introduced into E.
coli host GM221 by transformation. Clones were initially screened for production of the recombinant protein. Plasmid DNA was isolated from positive clones and DNA sequencing was performed to verify the DNA sequence of the HuOPG-Met[22- 401](P25A) gene. The following oligonucleotides were used to generate the XbaI 15 KpnI linker: Oligo #1289-84 GAA GGA GGA ATA ACA TAT GGA AAC TTT TGC TCC AAA ATA TCT TCA TTA TGA TGA AGA AAC TAG TCA TCA GCT GCT GTG TGA TAA ATG TCC GCC GGG TAC (SEQ ID NO:115) Oligo #1289-85 CCG GCG GAC ATT TAT CAC ACA GCA GCT GAT GAC TAG TTT CTT CAT CAT AAT GAA GAT ATT TTG GAG CAA AAG TTT CCA TAT GTT ATT CCT CCT T-3' (SEQ ID NO:116) X. Human OPG met[22-4011 (P26A) and (P26D) A DNA sequence coding for an N-terminal methionine and amino acids 22 through 401 of human OPG with the proline at position 26 being substituted by alanine under control of the lux PR promoter in prokaryotic expression vector pAMG21 was constructed as follows: Synthetic oligos 1289-86 and 1289-87 were annealed to form an oligo linker duplex with XbaI and Spel cohesive ends. The synthetic linker duplex utilized optimal E. coli codons and encoded an N-terminal methionine. The linker duplex was directionally inserted between the XbaI and Spel sites in pAMG21- 81 huOPG[22-401](P25A) using standard methods. The ligation mixture was introduced into E. coli host GM221 by transformation. Clones were initially screened for production of the recombinant protein. Plasmid DNA was isolated from positive clones and DNA sequencing was performed to verify the DNA sequence of the huOPG-met[22- 401](P26A) gene. One of the clones sequenced was found to have the proline at position.
26 substituted by aspartic acid rather than alanine, and this clone was designated huOPG-met[22-401](P26D). The following oligonucleotides were used to generate the Xbal Spel linker: Oligo #1289-86 5' CTA GAA GGA GGA ATA ACA TAT GGA AAC TTT TCC TGC TAA ATA TCT TCA TTA TGA TGA AGA AA 3' (SEQ ID NO:117) 01-O igo CTA GTT TCT TCA TCA TAA TGA AGA TAT TTA GCA GGA AAA GTT TCC ATA TGT TAT TCC TCC TT 3' (SEQ ID NO:118) Y. Human OPG met[22-1941 S* 20 A DNA sequence coding for an N-terminal methionine and amino acids 22 through 194 of human OPG with the proline at position 25 being substituted by alanine :under control of the lux PR promoter in prokaryotic expression vector pAMG21 was constructed as follows: The plasmids pAMG21-huOPG[27-194] and pAMG21huOPG[22-401] (P25A) were each digested with KpnI and BamHI endonucleases. The 450 bp fragment was isolated from pAMG21-huOPG[27-194] and the 6.1 kbp fragment was isolated from pAMG21-huOPG[22-401] (P25A). These fragments were ligated together and introduced into E. coli host GM221 by transformation. Clones were initially screened for production of the recombinant protein. Plasmid DNA was isolated from positive clones and DNA sequencing was performed to verify the DNA sequence of the huOPG-Met[22-194](P25A) gene.
EXAMPLE 9 Association of OPG Monomers CHO cells engineered to overexpress muOPG [22-401] were used to generate conditioned media for the analysis of secreted recombinant OPG using rabbit polyclonal anti-OPG antibodies. An aliquot of conditioned media was concentrated 20-fold, then analysed by reducing and non-reducing SDS-PAGE (Figure 15). Under reducing conditions, the protein migrated as a Mr 50-55 kd polypeptide, as would be predicted if the mature product was glycosylated at one or more of its consensus N-linked glycosylation sites. Suprisingly, when the same samples were analysed by non-reducing SDS-PAGE, the majority of the protein migrated as an approximately 100 kd polypeptide, twice the size of the reduced protein. In addition, there was a smaller amount of the Mr 50-55 kd polypeptide. This pattern of migration on SDS-PAGE was consistent with the notion that the OPG product was forming dimers through oxidation i1 of a free sulfhydryl group(s).
°The predicted mature OPG polypeptide contains 23 cysteine residues, 18 of which 15 are predicted to be involved in forming intrachain disulfide bridges which comprise the four cysteine-rich domains (Figure 12A). The five remaining C-terminal cysteine residues are not involved in secondary structure which can be predicted based upon homology with other TNFR family members. Overall there is a net uneven number of cysteine residues, and it is formally possible that at least one residue is free to form an 20 intermolecular disulfide bond between two OPG monomers.
To help elucidate patterns of OPG kinesis and monomer association, a pulse-chase labelling study was performed. CHO cells expressing muOPG [22-401] were metabolically labelled as described above in serum-free medium containing 35
S
methionine and cysteine for 30 min. After this period, the media was removed, and replaced with complete medium containing unlabelled methionine and cysteine at levels approximately 2,000-fold excess to the original concentration of radioactive amino acids.
At 30 min, Ihr, 2 hr, 4 hr, 6 hr and 12 hr post addition, cultures were harvested by the removal of the conditioned media, and lysates of the conditioned media and adherent monolayers were prepared. The culture media and cell lysates were clarified as described above, and then immunoprecipitated using anti-OPG antibodies as described above. After the immunoprecipitates were washed, they were released by boiling in nonreducing SDS-PAGE buffer then split into two equal halves. To one half, the reducing agent b-mercaptothanol was added to 5% final concentration, while the other half was maintained in non-reducing conditions. Both sets of immunoprecipitates were analysed by SDS-PAGE as described above, then processed for autoradiography and exposed to film. The results are shown in Figure 16. The samples analysed by reducing SDS-PAGE are depicted in the bottom two panels. After synthesis, the OPG polypeptide is rapidly processed to a slightly larger polypeptide, which probably represents modification by N-linked glycoslyation. After approximately 1-2 hours, the level of OPG in the cell decreases dramatically, and concomitantly appears in the culture supernatant. This appears to be the result of the vectoral transport of OPG from the cell into the media over time, consistent with the notion that OPG is a naturally secreted protein. Analysis of the same immunoprecipitates under nonreducing conditions reveals Sthie relationship between te fnormation ofOPG diiners and secretion into theconditioned media (Figure 16, upper panels). In the first 30-60 minutes, OPG monomers are processed in the cell by apparent glycoslylation, followed by dimer formation. Over time, the bulk of OPG monomers are driven into dimers, which subsequently disappear from the cell. Beginning about 60 minutes after synthesis, OPG dimers appear in the :conditioned media, and accumulate over the duration of the experiment. Following this period, OPG dimers are formed, which are then secreted into the culture media. OPG monomers persist at a low level inside the cell over time, and small amounts also appear Sin the media. This does not appear to be the result of breakdown of covalent OPG dimers, but rather the production of sub-stoichiometric amounts of monomers in the cell and subsequent secretion.
Recombinantly produced OPG from transfected CHO cells appears to be predominantly a dimer. To determine if dimerization is a natural process in OPG synthesis, we analysed the conditioned media of a cell line found to naturally express OPG. The CTLL-2 cell line, a murine cytotoxic T lymphocytic cell line (ATCC accession no. TIB-214), was found to express OPG mRNA in a screen of tissue and cell line RNA. The OPG transcript was found to be the same as the cloned and sequenced 3 0 2.5-3.0 kb RNA identified from kidney and found to encode a secreted molecule.
Western blot analysis of conditioned media obtained from CTLL-2 cells shows that 84 most, if not all, of the OPG protein secreted is a dimer (Figure 17). This suggests that OPG dimerization and secretion is not an artifact of overexpression in a cell line, but is likely to be the main form of the product as it is produced by expressing cells.
Normal and transgenic mouse tissues and serum were analysed to determine the nature of the OPG molecule expressed in OPG transgenic mice. Since the rat OPG cDNA was expressed under the control of a hepatocyte control element, extracts made from the parenchyma of control and transgenic mice under non-reducing conditions were analysed (Figure 18). In extract from transgenic, but not control mice, OPG dimers are readily detected, along with substoichiometric amounts of monomers. The OPG dimers and monomers appear identical to the recombinant murine protein expressed in the oo, genetically engineered CHO cells. This strongly suggests that OPG dimers are indeed a natural form of the gene product, and are likely to be key active components. Serum samples obtained from control and transgnic mic wre similarly analysd ywestern blot analysis. In control mice, the majority of OPG protein migrates as a dimer, while small amounts of monomer are also detected. In addition, significant amounts of a larger S• OPG related protein is detected, which migrates with a relative molecular mass
S
consistent with the predicted size of a covalently-linked trimer. Thus, recombinant OPG is expressed predominantly as a dimeric protein in OPG transgenic mice, and the dimer form may be the basis for the osteopetrotic phenotype in OPG mice. OPG recombinant protein may also exist in higher molecular weight "trimeric" forms.
To determine if the five C-terminal cysteine residues of OPG play a role in o homodimerization, the murine OPG codons for cytsteine residues 195 (C 195), C202, C277, C319, and C400 were changed to serine using the QuickChange T M Site-Directed Mutagenesis Kit (Stratagene, San Diego, CA) as described above. The muOPG gene was subcloned between the Not I and Xba I sites of the pcDNA 3.1 vector (Invitrogen, San Diego, CA). The resulting plasmid, pcDNA3.1-muOPG, and mutagenic primers were treated with Pfu polymerase in the presence of deoxynucleotides, then amplified in a thermocycler as described above. An aliqout of the reaction is then transfected into competent E. coli XL1-Blue by heatshock, then plated. Plasmid DNA from transformants was then sequenced to verify mutations.
The following primer pairs were used to change the codon for cysteine residue 195 to serine of the murine OPG gene, resulting in the production of a muOPG [22-401] C195S protein: 1389-19: 5' -CAC GCA AAA GTC GGG AAT AGA TGT CAC-3' (SEQ ID NO:150) 1406-38: -GTG ACA TCT ATT CCC GAC TTT TGC GTG-3' (SEQ ID NO:151) The following primer pairs were used to change the codon for cysteine residue 202 to serine of the murine OPG gene, resulting in the production of a muOPG [22-401] C202S protein: 1389-21: -CAC CCT GTC GGA AGA GGC CTT CTT C-3' (SEQ ID NO:152) 1389-22: 5' -GAA GAA GGC CTC TTC CGA CAG GGT G-3' (1389-22) (SEQ ID NO:153) The following primer pairs were used to change the codon for cysteine residue 277 25 to serine of the murine OPG gene, resulting in the production of a muOPG [22-401] C277S protein: 1389-23: -TGA CCT CTC GGA AAG CAG CGT GCA-3' (SEQ ID NO:154) 1389-24: -TGC ACG CTG CTT TCC GAG AGG TCA-3' (SEQ ID NO:155) 86 The following primer pairs were used to change the codon for cysteine residue 319 to serine of the murine OPG gene, resulting in the production of a muOPG [22-401] C319S protein: 1389-17: 5' -CCT CGA AAT CGA GCG AGC AGC TCC-3' (SEQ ID NO:156) 1389-18: -CGA TTT CGA GGT CTT TCT CGT TCT C-3' (SEQ ID NO:157) The following primer pairs were used to change the codon for cysteine residue 400 to serine of the murine OPG gene, resulting in the production of a muOPG [22-401] C400S protein: 15 1406-72: -CCG TGA AAA TAA GCT CGT TAT AAC TAG GAA TGG-3' (SEQ ID NO:158) 1406-75: 5' -CCA TTC CTA GTT ATA ACG AGC TTA TTT TCA CGG-3' (SEQ ID NO:159) Each resulting muOPG [22-401] plasmid containing the appropriate mutation was then transfected into human 293 cells, the mutant OPG-Fc fusion protein purified from conditioned media as described above. The biological activity of each protein was assessed the in vitro osteoclast forming assay described in example 11. Conditioned media from each transfectant was analysed by non-reducing SDS-PAGE and western blotting with anti-OPG antibodies.
Mutation of any of the five C-terminal cysteine residues results in the production of predominantly monomeric 55 kd OPG molecules. This strongly suggests that the C-terminal cysteine residues together play a role in OPG homodimerization.
C-terminal OPG deletion mutants were constructed to map the region(s) of the OPG C-terminal domain which are important for OPG homodimerization. These OPG mutants were constructed by PCR amplification using primers which introduce 87 premature stop translation signals in the C-terminal region ofmurine OPG. The 5' oligo was designed to the MuOPG start codon (containing a HindIII restriction site) and the 3' oligonucleotides (containing a stop codon and XhoI site) were designed to truncate the C-terminal region of muOPG ending at either threonine residue 200 (CT 200), proline 212 (CT212), glutamic acid 293 (CT-293), or serine 355 (CT-355).
The following primers were used to construct muOPG [22-200]: 1091-39: -CCT CTG AGC TCA AGC TTC CGA GGA CCA CAA TGA ACA AG- 3' (SEQ ID NO:160) 1391-91: 5' -CCT CTC TCG AGT CAG GTG ACA TCT ATT CCA GTG GC-3' (1391-91) (SEQ ID NO:161) CAC TTT TGC 15 The following primers were used to construct muOPG [22-212]: 1091-39: 5' -CCT CTG AGC TCA AGC TTC CGA GGA CCA CAA 3' (SEQ ID NO:162) 1391-90: 20 5' -CCT CTC TCG AGT CAA GGA ACA GCA AAC CTG (SEQ ID NO:163) The following primers were used to construct muOPG [22-293]: 1091-39: 5' -CCT CTG AGC TCA AGC TTC CGA GGA CCA CAA 3' (SEQ ID NO:164) 1391-89: TGA ACA AG- AAG AAG GC TGA ACA AG- CCT CTC TCG AGT CAC TCT GTG GTG AGG TTC GAG TGG CC- 3' (SEQ ID NO:165) The following primers were used to construct muOPG [22-355]: 1091-39: 88 -CCT CTG AGC TCA AGC TTC CGA GGA CCA CAA TGA ACA AG- 3' (SEQ ID NO:166) 1391-88: 5' CCT CTC TCG AGT CAG GAT GTT TTC AAG TGC TTG AGG GC-3' (SEQ ID NO:167) Each resulting muOPG-CT plasmid containing the appropriate truncation was then transfected into human 293 cells, the mutant OPG-Fc fusion protein purified from conditioned media as described above. The biological activity of each protein was assessed the in vitro osteoclast forming assay described in example 11. The conditioned medias were also analysed by non-reducing SDS-PAGE and western blotting using anti- OPG antibodies.
S.Truncation of the C-terminal reginn of OPG effects thP ability f nOP tn fnrm 15 homodimers. CT 355 is predominantly monomeric, although some dimer is formed. CT 293 forms what appears to be equal molar amounts of monomer and dimer, and also high molecular weight aggregates. However, CT 212 and CT 200 are monomeric.
EXAMPLE i 20 Purification of OPG A. Purification of mammalian OPG-Fc Fusion Proteins 5 L of conditioned media from 293 cells expressing an OPG-Fc fusion protein were prepared as follows. A frozen sample of cells was thawed into 10 ml of 293S media (DMEM-high glucose, Ix L-glutamine, 10% heat inactivated fetal bovine serum (FBS) and 100 ug/ml hygromycin) and fed with fresh media after one day. After three days, cells were split into two T175 flasks at 1:10 and 1:20 dilutions. Two additional 1:10 splits were done to scale up to 200 T175 flasks. Cells were at 5 days post-thawing at this point. Cells were grown to near confluency (about three days) at which time serum-containing media was aspirated, cells were washed one time with 25 ml PBS per flask and 25 ml of SF media (DMEM-high glucose, Ix L-glutamine) was added to each flask. Cells were maintained at 5% C02 for three days at which point the media was harvested, centrifuged, and filtered through 0.45m cellulose nitrate filters (Coming).
89 OPG-Fc fusion proteins were purified using a Protein G Sepharose column (Pharmacia) equilibrated in PBS. The column size varied depending on volume of starting media. Conditioned media prepared as described above was loaded onto the column, the column washed with PBS, and pure protein eluted using 100mM glycine pH 2.7. Fractions were collected into tubes containing 1M Tris pH 9.2 in order to neutralize as quickly as possible. Protein containing fractions were pooled, concentrated in either an Amicon Centricon 10 or Centriprep 10 and diafiltered into PBS. The pure protein is stored at Murine [22-401]-Fc, Murine [22-180]-Fc, Murine [22-194]-Fc, human [22-401]-Fc and human [22-201]Fc were purified by this procedure. Murine [22-185]-Fc is purified by this procedure.
B. Preparation of anti-OPG antibodies S. three New Zealand White rabbits (5-8 lbs initial wt) were injected subcutaneously with muOPG[22-401]-Fc fusion protein. Each rabbit was immunized on day 1 with mg of antigen emulsified in an equal volume of Freunds complete adjuvant. Further boosts (Days 14 and 28) were performed by the same procedure with the substitution of Freunds incomplete adjuvant. Antibody titers were monitored by EIA. After the second boost, the antisera revealed high antibody titers and 25ml production bleeds were obtained from each animal. The sera was first passed over an affinity column to which murine OPG-Fc had be immobilized. The anti-OPG antibodies were eluted with Pierce Gentle Elution Buffer containing 1% glacial acetic acid. The eluted protein was then dialyzed into PBS and passed over a Fc column to remove any antibodies specific for the Fc portion of the OPG fusion protein. The run through fractions containing anti-OPG specific antibodies were dialyzed into PBS.
C. Purification of murine OPGr22-4011 Antibody Affinity Chromatographv Affinity purified anti-OPG antibodies were diafiltered into coupling buffer (0.1M sodium carbonate pH 8.3, 0.5M NaCl), and mixed with CNBr-activated sepharose beads (Pharmacia) for two hours at room temperature. The resin was then washed with coupling buffer extensively before blocking unoccupied sited with 1M ethanolamine (pH 8.0) for two hours at room temperature. The resin was then washed with low pH (0.1M sodium acetate pH 4.0, 0.5M NaCI) followed by a high pH wash (0.1M Tris-HCl pH 8.0, 0.5M NaCI). The last washes were repeated three times. The resin was finally equilibrated with PBS before packing into a column. Once packed, the resin was washed with PBS. A blank elution was performed with 0.1M glycine-HCl, pH followed by re-equilibration with PBS.
Concentrated conditioned media from CHO cells expressing muOPG[22-410] was applied to the column at a low flow rate. The column was washed with PBS until UV absorbance measured at 280nm returned to baseline. The protein was eluted from the column first with 0.1M glycine-HCl (pH re-equilibrated with PBS, and eluted with a second buffer (0.1M CAPS, pH 10.5), 1M NaC1). The two elution pools were diafiltered separately into PBS and sterile filtered before freezing at -20 0
C.
Conventional Chromatography CHO cell conditioned media was concentrated 2 3x i1 an Amcon spiral wound cartridge (S10Y10) and diafiltered into 20mM tris pH 8.0. The diafiltered media was then applied to a Q-sepharose HP (Pharmacia) column which had been equilibrated with 20mM tris pH 8.0. The column was then washed until absorbence at 280nm reached baseline. Protein was eluted with a 20 column volume gradient of 0-300mM NaCI in tris S: pH 8.0. OPG protein was detected using a western blot of column fractions.
Fractions containing OPG were pooled and brought to a final concentration of 300mM NaC1, 0.2mM DTT. A NiNTA superose (Qiagen) column was equilibrated with 20mM tris pH 8.0, 300mM NaCI, 0.2mM DTT after which the pooled fractions were applied. The column was washed with equilibration buffer until baseline absorbence was reached. Proteins were eluted from the column with a 0-30mM Imidazole gradient in equilibration buffer. Remaining proteins were washed off the column with 1M Imidazole. Again a western blot was used to detect OPG containing fractions.
Pooled fractions from the NiNTA column were dialyzed into 10mm potassium phosphate pH 7.0, 0.2mM DTT. The dialyzed pool was then applied to a ceramic hydroxyapatite column (Bio-Rad) which had been equilibrated in 10mM phosphate buffer. After column washing, the protein was eluted with a 10-100mM potassium phosphate gradient over 20 column volumes. This was then followed by a 20 column volume gradient of 100-400 mM phosphate.
OPG was detected by coomassie blue staining of SDS-polyacrylamide gels and by western blotting. Fractions were pooled and diafiltered onto PBS and frozen at -80 0
C.
The purified protein runs as a monomer and will remain so after diafiltration into PBS.
The monomer is stable when stored frozen or at pH 5 at 4°C. However if stored at 4 0 C in PBS, dimers and what appears to be trimers and tetramers will form after one week.
D. Purification of human OPG met[22-4011 from E. coli The bacterial cell paste was suspended into 10 mM EDTA to a concentration of using a low shear homogenizer at 5 0 C. The cells were then disrupted by two homogenizations at 15,000 psi each at 5 0 C. The resulting homogenate was centrifuged at 5,000x g for one hour at 5 0 C. The centrifugal pellet was washed by low shear homogenization into water at the original homogenization volume followed by centrifugation as before. The washed pellet was then solubilized to 15% by a solution of (final concentration) 6M guan, 10 mM TrisHCl, pH 8.5 at ambient temperature for 30 minutes. This solution was diluted fold into 2M urea containing 50 mM CAPS, pH 10.5, 1 mM reduced glutathione and then stirred for 72 hours at 5 0 C. The OPG was purified from this solution at 25 0 C by first adjustment to pH 4.5 with acetic acid and then chromatography over a column of SP-HP Sepharose resin.equilibrated with 25 mM sodium acetate, pH 4.5. The column elution was carried out with a linear sodium chloride gradient from 50 mM to 550 mM in the same buffer using 20 column volumes at a flow rate of 0.1 column volumes/minute. The peak fractions containing only the desired OPG form were pooled S"and stored at 5 0 C or buffer exchanged into phosphate buffered saline, concentrated by ultrafiltration, and then stored at 5 0 C. This material was analyzed by reverse phase HPLC, SDS-PAGE, limulus amebocyte lysate assay for the presence of endotoxin, and N-terminal sequencing. In addition, techniques such as mass spectrometry, pH/temperature stability, fluoresence, circular dichroism, differential scanning calorimetry, and protease profiling assays may also be used to examine the folded nature of the protein.
92 EXAMPLE 11 Biological Activity of Recombinant OPG Based on histology and histomorphometry, it appeared that hepatic overexpression of OPG in transgenic mice markedly decreased the numbers of osteoclasts leading to a marked increase in bone tissue (see Example To gain further insight into potential mechanism(s) underlying this in vivo effect, various forms of recombinant OPG have been tested in an in vitro culture model of osteoclast formation (osteoclast forming assay). This culture system was originally devised by Udagawa (Udagawa et al.
10 Endocrinology 125, 1805-1813 (1989), Proc. Natl. Acad. Sci. USA 87, 7260-7264 (1990)) and employs a combination of bone marrow cells and cells from bone marrow stromal cell lines. A description of the modification of this culture system used for these studies has been previously published (Lacey et al. Endocrinology 136, 2367-2376 (1995)). In this method, bone marrow cells, flushed from the femurs and tibiae of mice, are cultured overnight in culture media (alpha MEM with 10% heat inactivated fetal bovine serum) supplemented with 500 U/ml CSF-1 (colony stimulating factor 1, also called M-CSF), a hematopoietic growth factor specific for cells of the monocyte/macrophage family lineage. Following this incubation, the non-adherent cells .o are collected, subjected to gradient purification, and then cocultured with cells from the bone marrow cell line ST2 (1 x 106 non-adherent cells 1 x 105 ST2 cells/ ml media).
The media is supplemented with dexamethasone (100 nM) and the biologically-active metabolite of vitamin D3 known as 1,25 dihydroxyvitamin D3 (1,25 (OH)2 D3, 10 nM).
To enhance osteoclast appearance, prostaglandin E2 (250 nM) is added to some cultures.
The coculture period usually ranges from 8 10 days and the media, with all of the supplements freshly added, is renewed every 3-4 days. At various intervals, the cultures are assessed for the presence of tartrate acid phosphatase (TRAP) using either a histochemical stain (Sigma Kit 387A, Sigma, St. Louis, MO) or TRAP solution assay.
The TRAP histochemical method allows for the identification of osteoclasts phenotypically which are multinucleated (3 3 nuclei) cells that are also TRAP+. The solution assay involves lysing the osteoclast-containing cultures in a citrate buffer (100 mM, pH 5.0) containing 0.1% Triton X-100. Tartrate resistant acid phosphatase activity 93 is then measured based on the conversion of p-nitrophenylphosphate (20 nM) to p-nitrophenol in the presence of 80 mM sodium tartrate which occurs during a minute incubation at RT. The reaction is terminated by the addition ofNaOH to a final concentration of 0.5 M. The optical density at 405 nm is measured and the results are plotted.
Previous studies (Udagawa et al. ibid) using the osteoclast forming assay have demonstrated that these cells express receptors for 12 5 I-calcitonin (autoradiography) and can make pits on bone surfaces, which when combined with TRAP positivity confirm that the multinucleated cells have an osteoclast phenotype. Additional evidence in support of the osteoclast phenotype of the multinucleated cells that arise in vitro in the osteoclast forming assay are that the cells express av and b3 integrins by immunocytochemistry and calcitonin receptor and TRAP mRNA by in situ hybridization
(ISH).
i.
The huOPG [22-401 ]-Fc fusion was purified from CHO cell conditioned media and subsequently utilized in the osteoclast forming assay. At 100 ng/ml ofhuOPG [22- 401]-Fc, osteoclast formation was virtually 100% inhibited (Figure 19A). The levels of TRAP measured in lysed cultures in microtitre plate wells were also inhibited in the presence of OPG with an ID50 of approximately 3 ng/ml (Figure 20). The level of TRAP activity in lysates appeared to correlate with the relative number of osteoclasts seen by TRAP cytochemistry (compare Figures 19A-19G and 20). Purified human IgG and TNFbp were also tested in this model and were found to have no inhibitory or stimulatory effects suggesting that the inhibitory effects of the huOPG [22-401 ]-Fc were due to the OPG portion of the fusion protein. Additional forms of the human and murine molecules have been tested and the cumulative data are summarized in Table 1.
94 Table I Effects of various OPG forms on in vitro osteoclast formation OPG Construct Relative Bioactivity in vitro muOPG [22-401] -Fe muOPG 122-194] -Fe muOPG [22-185]-Fc muOPG [22-180]-Fc muOPG [22-401] muOPG [22-401] C195..
muOPG (22-401] C202 muOPG [22-401] C277 muOPG [22-4011 C319 *muOPG [22-401] C400 muOPG [22-185] *muOPG [22-194] *muOPG [22-200] 20 muOPG [22-212] muOPG [22-293] *muOPG [22-355] huOPG [22-401]-Fc huOPG [22-201] -Fe *huOPG [22-401]-Fc P26A..
huOPG [22-4011-Fe Y28F..
huOPG [22-401] huOPG [27-401] -Fe huOPG [29-4011-Fe huOPG [32-4011-Fe
ED
5 0 =0.4-2 ng/tnl
ED
5 0 =2-10 ng/ml
ED
5 0 =10.-lao ng/ml
ED
5 0 100 ng/ml The cumulative data suggest that murine and human OPG amino acid sequences 22-401 are fully active in vitro, when either fused to the Fc domain, or unfused. They inhibit in a dose-dependent manner and possess half-maximal activities in the 2-10 ng/ml range. Truncation of the murine C-terminus at threonine residue 180 inactivates the molecule, whereas truncations at cysteine 185 and beyond have full activity. The cysteine residue located at position 185 is predicted to form an SS3 bond in the domain 4 region of OPG. Removal of this residue in other TNFR-related proteins has previously been shown to abrogate biological activity (Yan et al. J. Biol. Chem. 266, 12099-12104 (1994)). Our finding that muOPG[22-180]-Fc is inactive while muOPG[22-185]-Fc is active is consistent with these findings. This suggests that amino acid residues 22-185 define a region for OPG activity.
These findings indicate that like transgenically-expressed OPG, recombinant OPG protein also suppressed osteoclast formation as tested in the osteoclast forming assay.
Time course experiments examining the appearance of TRAP+ cells, b3+ cells, F480+ cells in cultures continuously exposed to OPG demonstrate that OPG blocks the appearance TRAP+ and b3+ cells, but not F480+ cells. In contrast, TRAP+ and b3+ cells begin to appear as early as day 4 following culture establishment in control cultures. Only F480+ cells can be found in OPG-treated cultures and they appear to be Spresent at qualitatively the same numbers as the control cultures. Thus, the mechanism of OPG effects in vitro appears to involve a blockade in osteoclast differentiation at a step beyond the appearance of monocyte-macrophages but before the appearance of cells expressing either TRAP or b3 integrins. Collectively these findings indicate that OPG does not interfere with the general growth and differentiation of monocyte-macrophage precursors from bone marrow, but rather suggests that OPG specifically blocks the selective differentiation of osteoclasts from monocyte-macrophage precursors.
To determine more specifically when in the osteoclast differentiation pathway that OPG was inhibitory, a variation of the in vitro culture method was employed. This variation, described in (Lacey et al. supra), employs bone marrow macrophages as osteoclast precursors. The osteoclast precursors are derived by taking the nonadherent bone marrow cells after an overnight incubation in CSF-1/M-CSF, and culturing the cells for an additional 4 days with 1,000 2,000 U/ml CSF-1. Following 4 days of 96 culture, termed the growth phase, the non-adherent cells are removed. The adherent cells, which are bone marrow macrophages, can then be exposed for up to 2 days to various treatments in the presence of 1,000 2,000 U/ml CSF-1. This 2 day period is called the intermediate differentiation period. Thereafter, the cell layers are again rinsed and then ST-2 cells (1 X 10 5 cell/ml), dexamethasone (100 nM) and 1,25 (OH)2 D3 nM) are added for the last 8 days for what is termed the terminal differentiation period.
Test agents can be added during this terminal period as well. Acquisition of phenotypic markers of osteoclast differentiation are acquired during this terminal period (Lacey et al. ibid).
huOPG [22-401]-Fc (100 ng/ml) was tested for its effects on osteoclast formation in this model by adding it during either the intermediate, terminal or, alternatively, both differentiation periods. Both TRAP cytochemistry and solution assays were performed.
The results of the solution assay are shown in Figure 21. HuOPG [22-401]-Fc inhibited the appearance of TRAP activity when added to both the intermediate and terminal or only the terminal differentiation phases. When added to the intermediate phase and then removed from the cultures by rinsing, huOPG [22-401]-Fc did not block the appearance of TRAP activity in culture lysates. The cytochemistry results parallel the solution assay data. Collectively, these observations indicate that huOPG [22-401]-Fc only needs to be present during the terminal differentiation period for it to exert its all of its suppressive effects on osteoclast formation.
B. In vivo IL1-a and IL -b challenge experiments ILl increases bone resorption both systemically and locally when injected subcutaneously over the calvaria of mice (Boyce et al., Endocrinology 125, 1142-1150 (1989)). The systemic effects can be assessed by the degree of hypercalcemia and the local effects histologically by assessing the relative magnitude of the osteoclast-mediated response. The aim of these experiments was to determine if recombinant muOPG [22- 401]-Fc could modify the local and/or systemic actions of ILl when injected subcutaneously over the same region of the calvaria as IL1.
IL-1 b experiment Male mice (ICR Swiss white) aged 4 weeks were divided into the following treatment groups (5 mice per group): Control group: IL1 treated animals (mice received 1 injection/day of 2.5 ug of ILl-b); Low dose muOPG [22-401]-Fc treated animals (mice received 3 injections/day of 1 mg ofmuOPG [22-401]-Fc); Low dose muopg [22-401]- Fc and IL High dose muOPG [22-401]-Fc treated animals (mice receive 3 injections/day of 10 mg muOPG [22-401]-Fc); High dose muOPG [22-401]-Fc and ILlb. All mice received the same total number of injections of either active factor or vehicle bovine serum albumin in phosphate buffered saline). All groups are sacrificed on the day after the last injection. The weights and blood ionized calcium levels are measured before the first injections, four hours after the second injection and 24 hours after the third IL1 injection, just before the animals were sacrificed. After sacrifice the calvaria were removed and processed for paraffin sectioning.
IL1-a experiment Male mice (ICR Swiss white) aged 4 weeks were divided into the following treatment groups (5 mice per group): Control group; IL1 alpha treated animals (mice received 1 injection/day of 5 ug of ILl-alpha); Low dose muOPG [22-401]-Fc treated animals (mice received 1 injection/day of 10 mg ofmuOPG [22-401]-Fc; Low dose muopg [22-401]-Fc and IL1-alpha, (dosing as above); High dose muopg [22-401]-Fc treated animals (mice received 3 injections/day of 10 mg muOPG [22-401]-Fc; High *dose muOPG [22-401]-Fc and ILl-a. All mice received the same number of injections/day of either active factor or vehicle. All groups were sacrificed on the day after the last injection. The blood ionized calcium levels were measured before the first injection, four hours after the second injection and 24 hours after the third IL1 injection, just before the animals were sacrificed. The animal weights were measured before the first injection, four hours after the second injection and 24 hours after the third IL1 injection, just before the animals were sacrificed. After sacrifice the calvaria were removed and processed for paraffin sectioning.
Histological methods Calvarial bone samples were fixed in zinc formalin, decalcified in formic acid, dehydrated through ethanol and mounted in paraffin. Sections (5mm thick) were cut 98 through the calvaria adjacent to the lambdoid suture and stained with either hematoxylin and eosin or reacted for tartrate resistant acid phosphatase activity (Sigma Kit# 387A) and counterstained with hematoxylin. Bone resorption was assessed in the IL 1-a treated mice by histomorphometric methods using the Osteomeasure (Osteometrics, Atlanta, GA) by tracing histologic features onto a digitizor platen using a microscope-mounted camera lucida attachment. Osteoclast numbers, osteoclast lined surfaces, and eroded surfaces were determined in the marrow spaces of the calvarial bone. The injected and non-injected sides of the calvaria were measured separately.
Results IL 1-a and IL 1-b produced hypercalcemia at the doses used, particularly on the second day, presumably by the induction of increased bone resorption systemically. The hypercalcemic response was blocked by muOPG [22-401]-Fc in the IL1-beta treated mice and significantly diminished in mice treated with IL1-alpha, an effect most apparent on day 2 (Figure 22A-22B).
Histologic analysis of the calvariae of mice treated with IL 1-alpha and beta shows that IL 1 treatments alone produce a marked increase in the indices of bone resorption including: osteoclast number, osteoclast lined surface, and eroded surface (surfaces showing deep scalloping due to osteoclastic action (Figure 23B, Table In response to o ILl-a or ILl-b, the increases in bone resorption were similar on the injected and noninjected sides of the calvaria. Muopg [22-401]-Fc injections reduced bone resorption in both IL 1-alpha and beta treated mice and in mice receiving vehicle alone but this reduction was seen only on the muopg [22-401]-Fc injected sides of the calvariae.
The most likely explanation for these observations is that muOPG [22-401]-Fc inhibited bone resorption, a conclusion supported by the reduction of both the total osteoclast number and the percentage of available bone surface undergoing bone resorption, in the region of the calvaria adjacent to the muOPG [22-401]-Fc injection sites. The actions ofmuOPG [22-401]-Fc appeared to be most marked locally by histology, but the fact that muOPG [22-401]-Fc also blunted IL1-induced hypercalcemia suggests that muOPG [22-401]-Fc has more subtle effects on bone resorption systemically.
*4 4 4 4* Se* 4 ~4* 4** Table 2. Effects of OPG on variables of bone resorption in IL-1 i jected mice.
Osteoclast Surface Bone Surface (mean Eroded Surface %Bone Surface Osteoclast Number/mm 2 Tissue S.D) (mean S.D) Area (mean S.D) Experiment 1 Non-injected side Injected side Non-injected Inject d side Non-injected Injected side side side Control 12.36 3.44 9.54 j 2.46 8.07 3.90 9.75 t 3.16 32.51 11.09 23.50 10.83 ILI-P (2.5pg/d) 17.18 1.30 16.40 2.16 40.66 t 4.28 37.53 10.28 71.80 18.76 60.89 5.16 OPO (40pg/d) 10.12 3.71 5.04 1.66 9.73 4.33 4.19 t 3.61 32.73 11.09 15.24 7.54 OPG+ILI-p 18.61 2.46 13.26 2.50 44.87 t 8.63 25. 6.82 69.42 36.29 47.13 t 24.26 Experiment 2 Control 11.56 4.22 11.95 2.97 12.67 5.04 10.03 5.13 51.72 23.93 56.03 30.70 ILI-a (Spg/d) 28.81 4.84 23.46 t 5.76 37.51 4 5.16 41.1 12.53 113.60 18.04 102.70 32.09 OPO (40pg/d) 14.40 1.00 8 4.26 2.54 11.55 4.14 4.29 3.16 72.28 14.11 I 22.65 16.68 326 2 OPO+ILI-a 129.58 t 8.80 N 1783 3.34 33.66 9.21 24 38 8.88 140.10U 42.31 6 o.3 1 .U I 0 3.3 Different to non-injected side p 0.05 (by paired t test) 100 C. Systemic Effects of muOPG [22-4011-Fc in Growing Mice Male BDF1 mice aged 3-4 weeks, weight range 9.2- 15.7g were divided into groups of ten mice per group. These mice were injected subcutaneously with saline or muOPG [22-401]-Fc 2.5mg/kg bid for 14 days (5mg/kg/day). The mice were radiographed before treatment, at day 7 and on day 14. The mice were sacrificed 24 hours after the final injection. The right femur was removed, fixed in zinc formalin, decalcified in formic acid and embedded in paraffin. Sections were cut through the mid region of the distal femoral metaphysis and the femoral shaft. Bone density, by histomorphometry, was determined in six adjacent regions extending from the metaphyseal limit of the growth plate, through the primary and secondary spongiosa and into the femoral diaphysis (shaft). Each region was 0.5 X 0.5 mm 2 Radiographic changes After seven days of treatment there was evidence of a zone of increased bone density in the spongiosa associated with the growth plates in the OPG treated mice relative to that seen in the controls. The effects were particularly striking in the distal femoral and the proximal tibial metaphases (Figure 24A-24B). However bands of increased density were also apparent in the vertebral bodies, the iliac crest and the distal tibia. At 14 days, the regions of opacity had extended further into the femoral and tibial shafts though the intensity of the radio-opacity was diminished. Additionally, there were no differences in the length of the femurs at the completion of the experiment or in the change in length over the duration of the experiment implying that OPG does not alter 'bone growth.
Histological Changes The distal femoral metaphysis showed increased bone density in a regions 1.1 to 2.65 mm in distance from the growth plate (Figures 25 and 26A-26B). This is a region where bone is rapidly removed by osteoclast-mediated bone resorption in mice. In these rapidly growing young mice, the increase in bone in this region observed with OPG treatment is consistent with an inhibition of bone resorption.
D. Effects of Osteoprotegerin on Bone Loss Induced by Ovariectomy in the Rat 3 0 Twelve week old female Fisher rats were ovariectomized (OVX) or sham operated and dual xray absorptiometry (DEXA) measurements made of the bone density in the distal femoral metaphysis. After 3 days recovery period, the animals received daily injections for 14 days as follows: Ten sham operated animals received vehicle (phosphate buffered saline); Ten OVX animals received vehicle (phosphate buffered saline); Six OVX animals received OPG-Fc 5mg/kg SC; Six OVX animals received pamidronate (PAM) 5mg/kg SC; Six OVX animals received estrogen (ESTR) SC. After 7 and 14 days treatment the animals had bone density measured by DEXA.
Two days after the last injection the animals were killed and the right tibia and femur removed for histological evaluation.
The DEXA measurements of bone density showed a trend to reduction in the bone density following ovariectomy that was blocked by OPG-Fc. Its effects were similar to the known antiresorptive agents estrogen and pamidronate. (Figure 27). The histomorphometric analysis confirmed these observations with OPG-Fc treatment producing a bone density that was significantly higher in OVX rats than that seen in Suntreated OVX rats (Figure 28). These results confirm the activity of OPG in the bone loss associated with withdrawal of endogenous estrogen following ovariectomy.
.In vivo Summary The in vivo actions of recombinant OPG parallel the changes seen in OPG transgenic mice. The reduction in osteoclast number seen in the OPG transgenic is reproduced by injecting recombinant OPG locally over the calvaria in both normal mice and in mice treated with IL1-a or IL1-b. The OPG transgenic mice develop an osteopetrotic phenotype with progressive filling of the marrow cavity with bone and unremodelled cartilage extending from the growth plates from day 1 onward after birth.
In normal three week old (growing) mice, OPG treatments also led to retention of bone and unremodelled cartilage in regions of endochondral bone formation, an effect observed radiographically and confirmed histologically. Thus, recombinant OPG produces phenotypic changes in normal animals similar to those seen in the transgenic animals and the changes are consistent with OPG-induced inhibition of bone resorption.
Based on in vitro assays of osteoclast formation, a significant portion of this inhibition is due to impaired osteoclast formation. Consistent with this hypothesis, OPG blocks 3 0 ovariectomy-induced osteoporosis in rat. Bone loss in this model is known to be mediated by activated osteoclasts, suggesting a role for OPG in treatment of primary osteoporosis.
EXAMPLE 12 Pegylation Derivatives of OPG Preparation of N-terminal PEG-OPG conjugates by reductive alkvlation HuOPG met [22-194] P25A was buffer exchanged into 25-50 mM NaOAc, pH 4.5-4.8 and concentrated to 2-5 mg/ml. This solution was used to conduct OPG reductive alkylation with monofunctional PEG aldehydes at 5-7 o C. PEG monofunctional aldehydes, linear or branched, MW=I to 57 kDa (available from Shearwater Polymers) were added to the OPG solution as solids in amounts constituting 2-4 moles of PEG aldehyde per mole of OPG. After dissolution of polymer into the protein solution, sodium cyanoborohydride was added to give a final concentration of to 20 mM in the reaction mixture from 1-1.6 M freshly prepared stock solution in cold DI water. The progress of the reaction and the extent of OPG PEGylation was monitored by size exclusion HPLC on a G3000SWXL column (Toso Haas) eluting with 100 mM NaP04, 0.5 M NaC1, 10% ethanol, pH 6.9. Typically the reaction was allowed to proceed for 16-18 hours, after which the reaction mixture was diluted 6-8 times and the pH lowered to 3.5-4. The reaction mixture was fractionated by ion exchange chromatography (HP SP HiLoad 16/10, Pharmacia) eluting with 20 mM NaOAc pH 4 with a linear gradient to 0.75M NaCl over 25 column volumes at a flow rate of 30 cm/h.
Fractions of mono-, di- or poly- PEGylated OPG were pooled and characterized by SEC HPLC and SDS-PAGE. By N-terminal sequencing, it was determined that the monoPEG-OPG conjugate, the major reaction product in most cases, was 98% Nterminally PEG-modified OPG.
This procedure was generally used to prepare the following N-terminal PEG-OPG conjugates (where OPG is HuOPG met [22-194] P25A: 5 kD monoPEG, 10 kD mono branched PEG, 12 kD monoPEG, 20 kD monoPEG, 20 kD mono branched PEG, 25 kD monoPEG, 31 kD monoPEG, 57 kD monoPEG, 12 kD diPEG, 25 kD diPEG, 31 kD diPEG, 57 kD diPEG, 25 kD triPEG.
103 Preparation of PEG-OPG conjugates by acvlation HuOPG met [22-194] P25A was buffer exchanged into 50 mM BICINE buffer, pH 8 and concentrated to 2-3 mg/ml. This solution was used to conduct OPG acylation with monofunctional PEG N-hydroxysuccinimidyl esters at room temperature. PEG Nhydroxysuccinimidyl esters, linear or branched, MW=1 to 57 kDa (available from Shearwater Polymers) were added to the OPG solution as solids in amounts constituting 4-8 moles of PEG N-hydroxysuccinimidyl ester per mole of OPG. The progress of the reaction and the extent of OPG PEGylation was monitored by size exclusion HPLC on a G3000SWXL column (Toso Haas) eluting with 100 mM NaPO4, 0.5 M NaCI, ethanol, pH 6.9. Typically the reaction was allowed to proceed for 1 hour, after which the reaction mixture was diluted 6-8 times and the pH lowered to 3.5-4. The reaction mixture was fractionated by ion exchange chromatography (HP SP HiLoad 16/10, Pharmacia) eluting with 20 mM NaOAc pH 4 with a linear gradient to 0.75M NaCI over 25 column volumes at a flow rate of 30 cm/h. Fractions of mono-, di- or poly- PEGylated OPG were pooled and characterized by SEC HPLC and SDS-PAGE.
"This procedure was generally used to prepare the following PEG-OPG conjugates: 5 kD polyPEG, 20 kD polyPEG, 40 kD poly branched PEG, 50 kD poly PEG.
Preparation of dimeric PEG-OPG HuOPG met [22-194] P25A is prepared for thiolation at 1-3 mg/ml in a phosphate buffer at near neutral pH. S-acetyl mecaptosuccinic anhydride (AMSA) is added in a 3-7 i: fold molar excess while maintaining pH at 7.0 and the rxn stirred at 4_C for 2 hrs. The monothiolated-OPG is separated from unmodified and polythiolated OPG by ion exchange chromatography and the protected thiol deprotected by treatment with hydroxylamine. After deprotection, the hydroxylamine is removed by gel filtration and the resultant monothiolated-OPG is subjected to a variety of thiol specific crosslinking chemistries. To generate a disulfide bonded dimer, the thiolated OPG at >1mg/ml is allowed to undergo air oxidation by dialysis in slightly basic phosphate buffer. The covalent thioether OPG dimer was prepared by reacting the bis-maleimide crosslinker, N,N-bis(3-maleimido propianyl)-2-hydroxy 1,3 propane with the thiolated OPG at >1mg/ml at a 0.6x molar ratio of crosslinker:OPG in phosphate buffer at pH Similarly, the PEG dumbbells are produced by reaction of substoichiometric amounts of 104 bis-maleimide PEG crosslinkers with thiolated OPG at >1mg/ml in phosphate buffer at pH 6.5. Any of the above dimeric conjugates may be further purified using either ion exchange or size exclusion chromatographies.
Dimeric PEG-OPG conjugates (where OPG is HuOPG met [22-194] prepared using the above procedures include disulfide-bonded OPG dimer, covalent thioether OPG dimer with an aliphatic amine type crosslinker, 3.4 kD and 8kD PEG dumbbells and monobells.
PEG-OPG conjugates were tested for activity in vitro using the osteoclast maturation assay described in Example 11A and for activity in vivo by measuring increased bone density after injection into mice as described in Example 11C. The in vivo activity is shown below in Table 3.
erg.
I
6 0*ee 0@ OS S
I
*5 C. C C *6 I @0 *5 C
S
S. C
S.
C
S.
S.
Table 3 In vivo biological activity of Pegylated OPG Increase in Tibial Bone Density OPG Construct muOPG muOPG muOPG 20 huOPG huOPG huOPG huOPG huOPG huOPG huOPG huOPG huOPG met met met met met met met met met met met met [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] [22-194] 5k PEG 20k PEG P25A 5k PEG P25A 20k PEG P25A 31k PEG P25A 57k PEG P25A 12k PEG P25A 20k Branched PEG P25A 8k PEG dimer P25A disulfide crosslink 105 While the invention has been described in what is considered to be its preferred embodiments, it is not to be limited to the disclosed embodiments, but on the contrary, is intended to cover various modifications and equivalents included within the spirit and scope of the appended claims, which scope is to be accorded the broadest interpretation so as to encompass all such modifications and equivalents.
o.o *o o 106 SEQUENCE LISTING GENERAL INFORMATION: APPLICANT:Amgen Inc.
(ii) TITLE OF INVENTION: OSTEOPROTEGERIN (iii) NUMBER OF SEQUENCES: 168 (iv) CORRESPONDENCE
ADDRESS:
ADDRESSEE: Amgen Inc.
STREET: 1840 Dehavilland Drive CITY: Thousand Oaks STATE: California COUNTRY: United States ZI7P: 91320 COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk 15 COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS SOFTWARE: PatentIn Release Version #1.30 (vi) CURRENT APPLICATION DATA: APPLICATION NUMBER: S. 20 FILING DATE: .i CLASSIFICATION: (viii) ATTORNEY/AGENT INFORMATION: NAME: Winter, Robert B.
REFERENCE/DOCKET NUMBER: A-378-CIP2 INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 36 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 107 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1: AAAGGAAGGA AAAAAGCGGC CGCTACANNN NNNNNT 36 INFORMATION FOR SEQ ID NO:2: SEQUENCE CHARACTERISTICS: LENGTH: 16 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: TCGACCCACG CGTCCG 16 INFORMATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: 25 LENGTH: 12 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: GGGTGCGCAG GC 12 108 INFORMATION FOR SEQ ID NO:4: SEQUENCE CHARACTERISTICS: LENGTH: 18 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: 1b TGTAAAACGA CGGCCAGT 18 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 18 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID CAGGAAACAG CTATGACC 18 INFORMATION FOR SEQ ID NO:6: SEQUENCE CHARACTERISTICS: SEQUENCE CHARACTERISTICS: LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: CAATTAACCC TCACTAAAGG IJflJ lIU 'J Ii NJ SEQUENCE CHARACTERISTICS: LENGTH: 23 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: GCATTATGAC CCAGAAACCG GAC INFORMATION FOR SEQ ID NO:8: SEQUENCE CHARACTERISTICS: LENGTH: 23 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 110 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: AGGTAGCGCC CTTCCTCACA TTC 23 INFORMATION FOR SEQ ID NO:9: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs *o TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: GACTAGTCCC ACAATGAACA AGTGGCTGTG INFORMATION FOR SEQ ID
S*
25 SEQUENCE CHARACTERISTICS: LENGTH: 45 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID ATAAGAATGC GGCCGCTAAA CTATGAAACA GCCCAGTGAC CATTC 111 INFORMATION FOR SEQ ID NO:11: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: GCCTCTAGAA AGAGCTGGGA C 21 INFORMATION FOR SEQ ID NO:12: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 25 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: CGCCGTGTTC CATTTATGAG C 21 INFORMATION FOR SEQ ID NO:13: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: ATCAAAGGCA GGGCATACTT CCTG 24 INFORMATION FOR SEQ ID NO:14: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: a.
25 GTTGCACTCC TGTTTCACGG TCTG 24 .a INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH:.24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 113 (xi) SEQUENCE DESCRIPTION: SEQ ID CAAGACACCT TGAAGGGCCT GATG 24 INFORMATION FOR SEQ ID NO:16: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16: TAACTTTTAC AGAAGAGCAT CAGC 24 INFORMATION FOR SEQ ID NO:17: SEQUENCE CHARACTERISTICS: 25 LENGTH: 33 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17: AGCGCGGCCG CATGAACAAG TGGCTGTGCT GCG 33 114 INFORMATION FOR SEQ ID NO:18: SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA INFORMATION FOR SEQ ID NO:19: S*-1 SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: GTGAAGCTGT GCAAGAACCT GATG 24 INFORMATION FOR SEQ ID *o **2O 115 SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID .ATCAAAGGCA GGGCATACTT CCTG 24 INFORMATION FOR SEQ ID NO:21: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: CAGATCCTGA AGCTGCTCAG TTTG 24 INFORMATION FOR SEQ ID NO:22: SEQUENCE CHARACTERISTICS: LENGTH: 33 base pairs TYPE: nucleic acid 116 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: AGCGCGGCCG CGGGGACCAC AATGAACAAG TTG 33 INFORMATION FOR SEQ ID NO:23: I* SEQUENCE CHARACTERISTICS: 15 1NA33Lpar TYPE: nucleic acid STRANYDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 5030 25 SEQUENCE DCRIPTRTI:SQI: N: LENGTH: 39 base pairs TYPE: nucleic acid STRA3NDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 117 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: ATAGCGGCCG CTGAGCCCAA ATCTTGTGAC AAAACTCAC 39 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 45 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 0* (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID TCTAGAGTCG ACTTATCATT TACCCGGAGA CAGGGAGAGG CTCTT INFORMATION FOR SEQ ID NO:26: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs 25 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 118 INFORMATION FOR SEQ ID NO:27: SEQUENCE CHARACTERISTICS: LENGTH: 43 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:27: *1 CCTCTGCGGC CGCTAAGCAG CTTATTTTCA CGGATTGAAC CTG 43 INFORMATION FOR SEQ ID NO:28: e SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:28: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 INFORMATION FOR SEQ ID NO:29: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs 119 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:29: TCCGTAAGAA ACAGCCCAGT GACC 24 S* 15 INFORMATION FOR SEQ ID *ee SEQUENCE CHARACTERISTICS: e LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNESS: single e S(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA e (xi) SEQUENCE DESCRIPTION: SEQ ID CCTCTGCGGC CGCTGTTGCA TTTCCTTTCT G 31 INFORMATION FOR SEQ ID NO:31: SEQUENCE CHARACTERISTICS: LENGTH: 19 amino acids TYPE: amino acid STRANDEDNESS: single 120 TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:31: Glu Thr Leu Pro Pro Lys Tyr Leu His Tyr Asp Pro Glu Thr Gly His 1 5 10 Gin Leu Leu 9 15 INFORMATION FOR SEQ ID NO:32: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 9 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:32: TCCCTTGCCC TGACCACTCT T 21 INFORMATION FOR SEQ ID NO:33: SEQUENCE CHARACTERISTICS: LENGTH: 34 base pairs TYPE: nucleic acid STRANDEDNESS: single 121 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:33: CCTCTGCGGC CGCACACACG TTGTCATGTG TTGC 34 INFORMATION FOR SEQ ID NO:34: SEQUENCE CHARACTERISTICS: 15 LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 25 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:34: TCCCTTGCCC TGACCACTCT T 21 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 34 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 122 (xi) SEQUENCE DESCRIPTION: SEQ ID CCTCTGCGGC CGCCTTTTGC GTGGCTTCTC TGTT 34 INFORMATION FOR SEQ ID NO:36: SEQUENCE CHARACTERISTICS: LENGTH: 37 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear S:(ii) MOLECULE TYPE: cDNA S(xi) SEQUENCE DESCRIPTION: SEQ ID NO:36: e 20 CCTCTGAGCT CAAGCTTGGT TTCCGGGGAC CACAATG 37 INFORMATION FOR SEQ ID NO:37: SEQUENCE CHARACTERISTICS: 25 LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:37: CCTCTGCGGC CGCTAAGCAG CTTATTTTTA CTGAATGG 38 123 INFORMATION FOR SEQ ID NO:38: SEQUENCE CHARACTERISTICS: LENGTH: 37 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 15 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:38: CCTCTGAGCT CAAGCTTGGT TTCCGGGGAC CACAATG 37 INFORMATION FOR SEQ ID NO:39: SEQUENCE CHARACTERISTICS: LENGTH: 33 base pairs TYPE: nucleic acid STRANDEDNESS: single 25 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:39: CCTCTGCGGC CGCCAGGGTA ACATCTATTC CAC 33 INFORMATION FOR SEQ ID 124 SEQUENCE CHARACTERISTICS: LENGTH: 35 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID CCGAAGCTTC CACCATGAAC AAGTGGCTGT GCTGC 15 INFORMATION FOR SEQ ID NO:41: SEQUENCE CHARACTERISTICS: ee LENGTH: 40 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 4* (xi) SEQUENCE DESCRIPTION: SEQ ID NO:41: CCTCTGTCGA CTATTATAAG CAGCTTATTT TCACGGATTG INFORMATION FOR SEQ ID NO:42: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single 125 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:42: TCCCTTGCCC TGACCACTCT T 21 INFORMATION FOR SEQ ID NO:43: SEQUENCE CHARACTERISTICS: 15 LENGTH: 35 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 20 (ii) MOLECULE TYPE: cDNA g e (xi) SEQUENCE DESCRIPTION: SEQ ID NO:43: CCTCTGTCGA CTTAACACAC GTTGTCATGT GTTGC INFORMATION FOR SEQ ID NO:44: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 126 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:44: TCCCTTGCCC TGACCACTCT T 21 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 35 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID 20 CCTCTGTCGA CTTACTTTTG CGTGGCTTCT CTGTT e• INFORMATION FOR SEQ ID NO:46: SEQUENCE CHARACTERISTICS: 25 LENGTH: 1537 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 127 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:46: GTGAAGAGCG TGAAGAGCGG TTCCTCCTTT CAGCAAAAAA CCCCTCAAGA CCCGTTTAGA GGCCCCAAGG GGTTATGCTA GTTATTGCTC AGCGGTGGCA GCAGCCAACT CAGCTTCCTT 120 TCGGGCTTTC TTCTTCTTCT TCTTCTTTCC GCGGATCCTC GAGTAAGCTT CCATGGTACC 180 CTGCAGGTCG ACACTAGTGA GCTCGAATTC CAACGCGTTA ACCATATGTT ATTCCTCCTT 240 TAATTAGTTA AAACAAATCT AGAATCAAAT CGATTAATCG ACTATAACAA ACCATTTTCT 300 *TGCGTAAACC TGTACGATCC TACAGGTACT TATGTTAAAC AATTGTATTT CAAGCGATAT 360 15 AATAGTGTGA CAAAAATCCA AT-TTAAACA'~±~ J-CATCCTTAT 2 ATATATAACA CGCAAAACTT GCGACAAACA ATAGGTAAGG ATAAAGAGAT GGGTATGAAA 480 *GACATAAATG CCGACGACAC TTACAGAATA ATTAATAAAA TTAAAGCCTG TAGAAGCAAT 540 **AATGATATTA ATCAATGCTT ATCTGATATG ACTAAAATGG TACATTGTGA ATATTATTTA 600 CTCGCGATCA TTTATCCTCA TTCTATGGTT AAATCTGATA TTTCAATTCT GGATAATTAC 660 CCTAAAAAAT GGAGGCAATA TTATGATGAC GCTAATTTAA TAAAATATGA TCCTATAGTA 720 GATTATTCTA ACTCCAATCA TTCACCGATT AATTGGAATA TATTTGAAAA CAATGCTGTA 780 AATAAAAAAT CTCCAAATGT AATTAAAGAA GCGAAATCAT CAGGTCTTAT CACTGGGTTT 840 *AGTTTCCCTA TTCATACTGC TAATAATGGC TTCGGAATGC TTAGTTTTGC ACATTCAGAG 900 AAAGACAACT ATATAGATAG TTTATTTTTA CATGCGTGTA TGAACATACC ATTAATTGTT 960 CCTTCTCTAG TTGATAATTA TCGAAAAATA AATATAGCAA ATAATAAATC AAACAACGAT 1020 TTAACCAAAA GAGAAAAAGA ATGTTTAGCG TGGGCATGCG AAGGAAAAAG CTCTTGGGAT 1080 128 ATTTCAAAAA TATTAGGCTG TAGTAAGCGC ACGGTCACTT TCCATTTAAC CAATGCGCAA 1140 ATGAAACTCA ATACAACAAA CCGCTGCCA.A AGTATTTCTA AAGCAATTTT AACAGGAGCA 1200 ATTGATTGCC CATACTTTAA AAGTTAAGTA CGACGTCCAT ATTTGAATGT ATTTAGAAAA 1260 ATAAACAAAA GAGTTTGTAG AAACGCAAAA AGGCCATCCG TCAGGATGGC CTTCTGCTTA 1320 ATTTGATGCC TGGCAGTTTA TGGCGGGCGT CCTGCCCGCC ACCCTCCGGG CCGTTGCTTC 1380 GCAACGTTCA AATCCGCTCC CGGCGGATTT GTCCTACTCA GGAGAGCGTT CACCGACAAA 1440 *CAACAGATAA AACGAAAGGC CCAGTCTTTC GACTGAGCCT TTCGTTTTAT TTGATGCCTG 1500 GCAGTTCCCT ACTCTCGCAT GGGGAGACCA TGCATAC 1537 INFORMATION FOR SEQ ID NO:47: SEQUENCE CHARACTERISTICS: LENGTH: 48 base pairs TYPE: nucleic acid STRANDEDNESS: single V. TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:47: CCGGCGGACA TTTATCACAC AGCAGCTGAT GAGAAGTTTC TTCATCCA 48 INFORMATION FOR SEQ ID NO:48: 129 SEQUENCE CHARACTERISTICS: LENGTH: 55 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA Soo. (xi) SEQUENCE DESCRIPTION: SEQ ID NO:48: *oo* *q CGATTTGATT CTAGAAGGAG GAATAACATA TGGTTAACGC GTTGGAATTC GGTAC S INFORMATION FOR SEQ ID NO:49: *5 a o0u, SEQUENCE CHARACTERISTICS: 0 6 LENGTH: 49 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:49: CGAATTCCAA CGCGTTAACC ATATGTTATT CCTCCTTCTA GAATCAAAT 49 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 1546 base pairs TYPE: nucleic acid 130 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA (xi) SEQUENCE DESCRIPTION: SEQ ID GCGTAACGTA TGCATGGTCT CCCCATGCGA GAGTAGGGAA CTGCCAGGCA TCAAATAAAA *CGAAAGGCTC AGTCGAAAGA CTGGGCCTTT CGTTTTATCT GTTGTTTGTC GGTGAACGCT 120 15 CTCCTGAGTA GGACAAATCC GCCGGGAGCG GATTTGAACG TTGCAGAAGcc~ d GGGTGGCGGG CAGGACGCCC GCCATAAACT GCCAGGCATC AAATTAAGCA GAAGGCCATC 240 CTGACGGATG GCCTTTTTGC GTTTCTACAA ACTCTTTTGT TTATTTTTCT AAATACATTC 300 .:AAATATGGAC GTCGTACTTA ACTTTTAAAG TATGGGCAAT CAATTGCTCC TGTTAAAATT 360 GCTTTAGAAA TACTTTGGCA GCGGTTTGTT GTATTGAGTT TCATTTGCGC ATTGGTTAAA 420 TGGAAAGTGA CCGTGCGCTT ACTACAGCCT AATATTTTTG AAATATCCCA AGAGCTTTTT 480 CCTTCGCATG CCCACGCTAA ACATTCTTTT TCTCTTTTGG TTAAATCGTT GTTTGATTTA 540 TTATTTGCTA TATTTATTTT TCGATAATTA TCAACTAGAG AAGGAACAAT TAATGGTATG 600 *TTCATACACG CATGTAAAAA TAAACTATCT ATATAGTTGT CTTTCTCTGA ATGTGCAAAA 660 CTAAGCATTC CGAAGCCATT ATTAGCAGTA TGAATAGGGA AACTAAACCC AGTGATAAGA 720 CCTGATGATT TCGCTTCTTT AATTACATTT GGAGATTTTT TATTTACAGC ATTGTTTTCA 780 AATATATTCC AATTAATCGG TGAATGATTG GAGTTAGAAT AATCTACTAT AGGATCATAT 840 131 TTTATTAAAT TAGCGTCATC ATAATATTGC CTCCATTTTT TAGGGTAATT ATCCAGAATT 900 GAAATATCAG ATTTAACCAT AGAATGAGGA TAAATGATCG CGAGTAAATA ATATTCACAA 960 TGTACCATTT TAGTCATATC AGATAAGCAT TGATTAATAT CATTATTGCT TCTACAGGCT 1020 TTAATTTTAT TAATTATTCT GTAAGTGTCG TCGGCATTT.A TGTCTTTCAT ACCCATCTCT 1080 TTATCCTTAC CTATTGTTTG, TCGCAAGTTT TGCGTGTTAT ATATCATTAA AACGGTAATA 1140 GATTGACATT TGATTCTAAT AAATTGGATT TTTGTC!ACAC TATTATATCG CTTGAAATAC 1200 AATTGTTTAA CATAAGTACC TGTAGGATCG TACAGGTTTA CGCAAGAAAA TGGTTTGTTA 1260 TAGTCGATTA ATCGATTTGA TTCTAGATTT GTTTTAACTA ATTAAAGGAG GAATAACATA 1320 *TGGTTAACGC GTTGGAATTC GAGCTCACTA GTGTCGACCT GCAGGGTACC ATGGAAGCTT 1380 ACTCGAGGAT CCGCGGAAAG AAGAAGAAGA AGAAGAA.AGC CCGAAAGGAA GCTGAGTTGG 1440 **:CTGCTGCCAC CGCTGAGCAA TAACTAGCAT AACCCCTTGG GGCCTCTAAA CGGGTCTTGA 1500 :GGGGTTTTTT GCTGAAAGGA GGAACCGCTC TTCACGCTCT TCACGC 1546 INFORMATION FOR SEQ ID NO:51: SEQUENCE CHARACTERISTICS: LENGTH: 47 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 132 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:51: TATGAAACAT CATCACCATC ACCATCATGC TAGCGTTAAC GCGTTGG INFORMATION FOR SEQ ID NO:52: SEQUENCE CHARACTERISTICS: LENGTH: 49 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA S" x) 15 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:52: *e AATTCCAACG CGTTAACGCT AGCATGATGG TGATGGTGAT GATGTTTCA INFORMATION FOR SEQ ID NO:53: SEQUENCE CHARACTERISTICS: LENGTH: 141 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 133 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:53: CTAATTCCGC TCTCACCTAC CAAACAATGC CCCCCTGCAA AAAATAAATT CATATAAAAA ACATACAGAT AACCATCTGC GGTGATAAAT TATCTCTGGC GGTGTTGACA TAAATACCAC 120 TGGCGGTGAT ACTGAGCACA T 141 INFORMATION FOR SEQ ID NO:54: SEQUENCE CHARACTERISTICS: *o LENGTH: 147 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA o (xi) SEQUENCE DESCRIPTION: SEQ ID NO:54: CGATGTGCTC AGTATCACCG CCAGTGGTAT TTATGTCAAC ACCGCCAGAG ATAATTTATC *A ACCGCAGATG GTTATCTGTA TGTTTTTTAT ATGAATTTAT TTTTTGCAGG GGGGCATTGT 120 TTGGTAGGTG AGAGCGGAAT TAGACGT INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 55 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 147 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID CGATTTGATT CTAGAAGGAG GAATAACATA TGGTTAACGC GTTGGAATTC GGTAC INFORMATION FOR SEQ ID NO:56: SEQUENCE CHARACTERISTICS: LENGTH: 49 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:56: CGAATTCCAA CGCGTTAACC ATATGTTATT CCTCCTTCTA GAATCAAAT INFORMATION FOR SEQ ID NO:57: SEQUENCE CHARACTERISTICS: LENGTH: 668 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 135 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:57: GTGAAGAGCG TGAAGAGCGG TTCCTCCTTT CAGCAAAAAA CCCCTCAAGA CCCGTTTAGA GGCCCCAAGG GGTTATGCTA GTTATTGCTC AGCGGTGGCA GCAGCCAACT CAGCTTCCTT 120 ***TCGGGCTTTC TTCTTCTTCT TCTTCTTTCC GCGGATCCTC GAGTAAGCTT CCATGGTACC 180 :15 CTGCAGGTCG ACACTAGTGA GCTCGAATTC CAACGCGTTA ACCATATGT TAATTAGTTA ACTCAAATCT AGAATCAAAT CGATAAATTG TGAGCGCTCA CAATTGAGAA 300 *TATTAATCAA GAATTTTAGC ATTTGTCAAA TGAATTTTTT AAAAATTATG AGACGTCCAT 360 :ATTTGAATGT ATTTAGAAAA ATAAACAAAA GAGTTTGTAG AAACGCAAAA AGGCCATCCG 420 TCAGGATGGC CTTCTGCTTA ATTTGATGCC TGGCAGTTTA TGGCGGGCGT CCTGCCCGCC 480 .:25 ACCCTCCGGG CCGTTGCTTC GCAACGTTCA AATCCGCTCC CGGCGGATTT GTCCTACTCA 540 GGAGAGCGTT CACCGACAAA CAACAGATAA AACGAAAGGC CCAGTCTTTC GACTGAGCCT 600 TTCGTTTTAT TTGATGCCTG GCAGTTCCCT ACTCTCGCAT GGGGAGACCA TGCATACGTT 660 *ACGCACGT 668 INFORMATION FOR SEQ ID NO:58: SEQUENCE CHARACTERISTICS: LENGTH: 726 base pairs TYPE: nucleic acid 136 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 58: GCGTAACGTA TGCATGGTCT CCCCATGCGA GAGTAGGGAA CTGCCAGGCA TCAAATAAAA CGAAAGGCTC AGTCGAAAGA CTGGGCCTTT CGTTTTATCT GTTGTTTGTC GGTGAACGCT 120 *15 CTCCTGAGTA GGACAAATCC GCCGGGAGCG GATTTGCGTU(A, RGCGA GGGTGGCGGG CAGGACGCCC GCCATAAACT GCCAGGCATC AAATTAAGCA GAAGGGGCCT 240 CCCACCGCCC GTCCTGCGGG CGGTATTTGA CGGTCCGTAG TTTAATTCGT CTTCGCCATC 300 *CTGACGGATG GCCTTTTTGC GTTTCTACAA ACTCTTTTGT TTATTTTTCT AAATACATTC 360 *AAATATGGAC GTCTCATAAT TTTTAAAAAA TTCATTTGAC AAATGCTAAA ATTCTTGATT 420 AATATTCTCA ATTGTGAGCG CTCACAATTT ATCGATTTGA TTCTAGATTT GTTTTAACTA 480 ATTAAAGGAG GAATAACATA TGGTTAACGC GTTGGAATTC GAGCTCACTA GTGTCGACCT 540 GCAGGGTACC ATGGAAGCTT ACTCGAGGAT CCGCGGAAAG AAGAAGAAGA AGAAGAAAGC 600 CCGAAAGGAA GCTGAGTTGG CTGCTGCCAC CGCTGAGCAA TAACTAGCAT AACCCCTTGG 660 GGCCTCTAAA CGGGTCTTGA GGGGTTTTTT GCTGAAAGGA GGAACCGCTC TTCACGCTCT 720 TCACGC 726 137 INFORMATION FOR SEQ ID NO:59: SEQUENCE CHARACTERISTICS: LENGTH: 44 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear S' (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:59: C C TACGCACTGG ATCCTTATAA GCAGCTTATT TTTACTGATT GGAC 44 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs S(B) TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA (xi) SEQUENCE DESCRIPTION: SEQ ID GTCCTCCTGG TACCTACCTA AAACAAC 27 138 INFORMATION FOR SEQ ID NO:61: SEQUENCE CHARACTERISTICS: LENGTH: 102 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 0 *@o 000*, (xi) SEQUENCE DESCRIPTION: SEQ ID NO:61: TATGGATGAA GAAACTTCTC ATCAGCTGCT GTGTGATAAA TGTCCGCCGG GTACCCGC GACATTTATC ACACAGCAGC TGATGAGAAG TTTCTTCATC CA 102
S
20 INFORMATION FOR SEQ ID NO:62: 00 SEQUENCE CHARACTERISTICS: LENGTH: 19 amino acids TYPE: amino acid 25 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:62: Met Asp Glu Glu Thr Ser His Gln Leu Leu Cys Asp Lys Cys Pro Pro 1 5 10 Gly Thr Tyr 139 INFORMATION FOR SEQ ID NO:63: SEQUENCE CHARACTERISTICS: LENGTH: 84 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:63: TATGGAAACT TTTCCTCCAA AATATCTTCA TTATGATGAA GAAACTTCTC ATCAGCTGCT GTGTGATAAA TGTCCGCCGG GTAC 84 INFORMATION FOR SEQ ID NO:64: SEQUENCE CHARACTERISTICS: LENGTH: 78 base pairs e 25 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:64: CCGGCGGACA TTTATCACAC AGCAGCTGAT GAGAAGTTTC TTCATCATAA TGAAGATATT 140 TTGGAGGAAA AGTTTCCA 78 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 44 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID •TACGCACTGG ATCCTTATAA GCAGCTTATT TTCACGGATT GAAC 44 INFORMATION FOR SEQ ID NO:66: "25 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:66: GTGCTCCTGG TACCTACCTA AAACAGCACT GCACAGTG 38 INFORMATION FOR SEQ ID NO:67: SEQUENCE CHARACTERISTICS: LENGTH: 84 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA *c (xi) SEQUENCE DESCRIPTION: SEQ ID NO:67: *15 TATGGAAACT CTGCCTCCAA AATACCTGCA TTACGATCCG GAAACTGGTC ATCAGCTGCT GTGTGATAAA TGTGCTCCGG GTAC 84 INFORMATION FOR SEQ ID NO:68: SEQUENCE CHARACTERISTICS: LENGTH: 78 base pairs TYPE: nucleic acid STRANDEDNESS: single 25 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:68: CCGGAGCACA TTTATCACAC AGCAGCTGAT GACCAGTTTC CGGATCGTAA TGCAGGTATT TTGGAGGCAG AGTTTCCA 78 142 INFORMATION FOR SEQ ID NO:69: SEQUENCE CHARACTERISTICS: LENGTH: 54 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:69: 15 TATGGACCCA GAAACTGGTC ATCAGCTGCT GTGTGATAAA TGTGCTCCGG GTAC 54 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 48 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 25 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID CCGGAGCACA TTTATCACAC AGCAGCTGAT GACCAGTTTC TGGGTCCA 48 INFORMATION FOR SEQ ID NO:71: SEQUENCE CHARACTERISTICS: LENGTH: 87 base pairs 143 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:71: TATGAAAGAA ACTCTGCCTC CAAAATACCT GCATTACGAT CCGGAAACTG GTCATCAGCT GCTGTGTGAT AAATGTGCTC CGGGTAC 87 S* INFORMATION FOR SEQ ID NO:72: SEQUENCE CHARACTERISTICS: LENGTH: 81 base pairs 20 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:72: CCGGAGCACA TTTATCACAC AGCAGCTGAT GACCAGTTTC CGGATCGTAA TGCAGGTATT TTGGAGGCAG AGTTTCTTTC A 81 144 INFORMATION FOR SEQ ID NO:73: SEQUENCE CHARACTERISTICS: LENGTH: 71 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:73: S/ GTTCTCCTCA TATGAAACAT CATCACCATC ACCATCATGA AACTCTGCCT CCAAAATACC TGCATTACGA T 71 INFORMATION FOR SEQ ID NO:74: SEQUENCE CHARACTERISTICS: LENGTH: 43 base pairs TYPE: nucleic acid 25 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:74: GTTCTCCTCA TATGAAAGAA ACTCTGCCTC CAAAATACCT GCA 43 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 76 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 15 (xi) SEQUENCE DESCRIPTION: SEQ ID TACGCACTGG ATCCTTAATG ATGGTGATGG TGATGATGTA AGCAGCTTAT TTTCACGGAT TGAACCTGAT TCCCTA INFORMATION FOR SEQ ID NO:76: SEQUENCE CHARACTERISTICS: LENGTH: 47 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:76: GTTCTCCTCA TATGAAATAC CTGCATTACG ATCCGGAAAC TGGTCAT 146 INFORMATION FOR SEQ ID NO:77: SEQUENCE CHARACTERISTICS: LENGTH: 43 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:77: GTTCTCCTAT TAATGAAATA TCTTCATTAT GATGAAGAAA CTT 43 e INFORMATION FOR SEQ ID NO:78: S" 20 SEQUENCE CHARACTERISTICS: LENGTH: 40 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:78: TACGCACTGG ATCCTTATAA GCAGCTTATT TTTACTGATT INFORMATION FOR SEQ ID NO:79: SEQUENCE CHARACTERISTICS: 147 LENGTH: 40 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:79: GTTCTCCTCA TATGGAAACT CTGCCTCCAA AATACCTGCA 15 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 43 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear S(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID TACGCACTGG ATCCTTATGT TGCATTTCCT TTCTGAATTA GCA 43 INFORMATION FOR SEQ ID NO:81: SEQUENCE CHARACTERISTICS: LENGTH: 18 base pairs TYPE: nucleic acid STRANDEDNESS: single 148 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:81: CCGGAAACAG ATAATGAG 18 INFORMATION FOR SEQ ID NO:82: S(i) SEQUENCE CHARACTERISTICS: 15 LENGTH: 18 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 20 (ii) MOLECULE TYPE: cDNA 25 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:82: GATCCTCATT ATCTGTTT 18 INFORMATION FOR SEQ ID NO:83: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 149 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:83: CCGGAAACAG AGAAGCCACG CAAAAGTAAG INFORMATION FOR SEQ ID NO:84: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:84: GATCCTTACT TTTGCGTGGC TTCTCTGTTT INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 12 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 150 (xi) SEQUENCE DESCRIPTION: SEQ ID TATGTTAATG AG 12 INFORMATION FOR SEQ ID NO:86: SEQUENCE CHARACTERISTICS: LENGTH: 14 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:86: GATCCTCATT AACA 14 INFORMATION FOR SEQ ID NO:87: 25 SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:87: 151 TATGTTCCGG AAACAGTTAA G 21 INFORMATION FOR SEQ ID NO:88: SEQUENCE CHARACTERISTICS: LENGTH: 23 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA ee* .9 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:88: 9* 9 GATCCTTAAC TGTTTCCGGA ACA 23 INFORMATION FOR SEQ ID NO:89: %oto% SEQUENCE CHARACTERISTICS: 25 LENGTH: 36 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:89: TATGTTCCGG AAACAGTGAA TCAACTCAAA AATAAG 36 152 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID GATCCTTATT TTTGAGTTGA TTCACTGTTT CCGGAACA 38 S(2) INFORMATION FOR SEQ ID NO:91: go SEQUENCE CHARACTERISTICS: *a LENGTH: 100 base pairs TYPE: nucleic acid STRANDEDNESS: single 25 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:91: CTAGCGACGA CGACGACAAA GAAACTCTGC CTCCAAAATA CCTGCATTAC GATCCGGAAA 153 CTGGTCATCA GCTGCTGTGT GATAAATGTG CTCCGGGTAC 100 INFORMATION FOR SEQ ID NO:92: SEQUENCE CHARACTERISTICS: LENGTH: 92 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:92: CCGGAGCACA TTTATCACAC AGCAGCTGAT GACCAGTTTC CGGATCGTAA TGCAGGTATT 20 TTGGAGGCAG AGTTTCTTTG TCGTCGTCGT CG 92 *6 INFORMATION FOR SEQ ID NO:93: SEQUENCE CHARACTERISTICS: LENGTH: 26 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 154 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:93: ACAAACACAA TCGATTTGAT ACTAGA 26 INFORMATION FOR SEQ ID NO:94: SEQUENCE CHARACTERISTICS: LENGTH: 50 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear e (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:94: TTTGTTTTAA CTAATTAAAG GAGGAATAAA ATATGAGAGG ATCGCATCAC e INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: 25 LENGTH: 50 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID CATCACCATC ACGAAACCTT CCCGCCGAAA TACCTGCACT ACGACGAAGA 155 INFORMATION FOR SEQ ID NO:96: SEQUENCE CHARACTERISTICS: LENGTH: 49 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:96: AACCTCCCAC CAGCTGCTGT GCGACAAATG CCCGCCGGGT ACCCAAACA 49 INFORMATION FOR SEQ ID NO:97: SEQUENCE CHARACTERISTICS: ee LENGTH: 26 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:97: TGTTTGGGTA CCCGGCGGGC ATTTGT 26 INFORMATION FOR SEQ ID NO:98: INFORMATION FOR SEQ ID NO:98: 156 SEQUENCE CHARACTERISTICS: LENGTH: 50 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:98: CGCACAGCAG CTGGTGGGAG GTTTCTTCGT CGTAGTGCAG GTATTTCGGC ^1 15 INFORMATION FOR SEQ ID NO:99: SEQUENCE CHARACTERISTICS: LENGTH: 49 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:99: GGGAAGGTTT CGTGATGGTG ATGGTGATGC GATCCTCTCA TATTTTATT 49 INFORMATION FOR SEQ ID NO:100: SEQUENCE CHARACTERISTICS: LENGTH: 50 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:100: CCTCCTTTAA TTAGTTAAAA CAAATCTAGT ATCAAATCGA TTGTGTTTGT INFORMATION FOR SEQ ID NO:101: SEQUENCE CHARACTERISTICS: LENGTH: 59 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear e (ii) MOLECULE TYPE: cDNA e e (xi) SEQUENCE DESCRIPTION: SEQ ID NO:101: ACAAACACAA TCGATTTGAT ACTAGATTTG TTTTAACTAA TTAAAGGAGG AATAAAATG 59 INFORMATION FOR SEQ ID NO:102: SEQUENCE CHARACTERISTICS: LENGTH: 48 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 158 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:102: CTAATTAAAG GAGGAATAAA ATGAAAGAAA CTTTTCCTCC AAAATATC 48 INFORMATION FOR SEQ ID NO:103: SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid 15 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:103: 25 TGTTTGGGTA CCCGGCGGAC ATTTATCACA C 31 INFORMATION FOR SEQ ID NO:104: SEQUENCE CHARACTERISTICS: LENGTH: 59 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 159 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:104: ACAAACACAA TCGATTTGAT ACTAGATTTG TTTTAACTAA TTAAAGGAGG AATAAAATG 59 INFORMATION FOR SEQ ID NO:105: SEQUENCE CHARACTERISTICS: LENGTH: 54 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:106: 160 TGTTTGGGTA CCCGGCGGAC ATTTATCACA C 31 INFORMATION FOR SEQ ID NO:107: SEQUENCE CHARACTERISTICS: LENGTH: 44 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA CAGCCCGGGT AAAATGGAAA CGTTTCCTCC AAAATATCTT CATT 44 INFORMATION FOR SEQ ID NO:108: S* LENGTH: 44 base pairs 25 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:108: CGTTTCCATT TTACCCGGGC TGAGCGAGAG GCTCTTCTGC GTGT 44 161 INFORMATION FOR SEQ ID NO:109: SEQUENCE CHARACTERISTICS: LENGTH: 45 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA *99 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:109: CGCTCAGCCC GGGTAAAATG GAAACGTTGC CTCCAAAATA CCTGC INFORMATION FOR SEQ ID NO:110: **9 SEQUENCE CHARACTERISTICS: S. LENGTH: 39 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear .o (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:110: CCATTTTACC CGGGCTGAGC GAGAGGCTCT TCTGCGTGT 39 INFORMATION FOR SEQ ID NO:111: SEQUENCE CHARACTERISTICS: 162 LENGTH: 36 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:111: GAAAATAAGC TGCTTAGCTG CAGCTGAACC AAAATC 36 15 INFORMATION FOR SEQ ID NO:112: SEQUENCE CHARACTERISTICS: LENGTH: 34 base pairs 9* TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear o (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:112: CAGCTGCAGC TAAGCAGCTT ATTTTCACGG ATTG 34 INFORMATION FOR SEQ ID NO:113: SEQUENCE CHARACTERISTICS: LENGTH: 36 base pairs TYPE: nucleic acid STRANDEDNESS: single 163 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:113: AAAAATAAGC TGCTTAGCTG CAGCTGAACC AAAATC 36 INFORMATION FOR SEQ ID NO:114: SEQUENCE CHARACTERISTICS: LENGTH: 35 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 20 (ii) MOLECULE TYPE: cDNA e* (xi) SEQUENCE DESCRIPTION: SEQ ID NO:114: CAGCTGCAGC TAAGCAGCTT ATTTTTACTG ATTGG INFORMATION FOR SEQ ID NO:115: SEQUENCE CHARACTERISTICS: LENGTH: 102 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 164 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:115: CTAGAAGGAG GAATAACATA TGGAAACTTT TGCTCCAAAA TATCTTCATT ATGATGAAGA AACTAGTCAT CAGCTGCTGT GTGATAAATG TCCGCCGGGT AC 102 INFORMATION FOR SEQ ID NO:116: SEQUENCE CHARACTERISTICS: o(A) LENGTH: 94 base pairs 15 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:116: CCGGCGGACA TTTATCACAC AGCAGCTGAT GACTAGTTTC TTCATCATAA TGAAGATATT TTGGAGCAAA AGTTTCCATA TGTTATTCCT CCTT 94 INFORMATION FOR SEQ ID NO:117: SEQUENCE CHARACTERISTICS: LENGTH: 62 base pairs TYPE: nucleic acid STRANDEDNESS: single 165 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:117: CTAGAAGGAG GAATAACATA TGGAAACTTT TCCTGCTAAA TATCTTCATT ATGATGAAGA AA 62 INFORMATION FOR SEQ ID NO:118: SEQUENCE CHARACTERISTICS: LENGTH: 62 base pairs TYPE: nucleic acid STRANDEDNESS: single 20 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:118: CTAGTTTCTT CATCATAATG AAGATATTTA GCAGGAAAAG TTTCCATATG TTATTCCTCC TT 62 INFORMATION FOR SEQ ID NO:119: SEQUENCE CHARACTERISTICS: LENGTH: 51 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:119: Tyr His Tyr Tyr Asp Gln Asn Gly Arg Met Cys Glu Glu Cys His Met Cys Gln Pro Gly His Phe Leu Val Lys His Cys Lys Gln Pro Lys Arg 25 Asp Thr Val Cys His Lys Pro Cys 40 Glu Pro Gly Val Thr Tyr Thr Asp Asp Trp His INFORMATION FOR SEQ ID NO:120: SEQUENCE CHARACTERISTICS: LENGTH: 2432 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: CDS LOCATION: 124..1326 167 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:120: ATCAAAGGCA GGGCATACTT CCTGTTGCCC AGACCTTATA TAAAACGTCA TGTTCGCCTG GGCAGCAGAG AAGCACCTAG CACTGGCCCA GCGGCTGCCG CCTGAGGTTT CCAGAGGACC ACA ATG AAC Met Asn 1.
AAG TGG Lys Trp CTG TGC TGT GCA CTC CTG Leu Cys Cys Ala Leu Leu 5 GTG TTC TTG GAC ATC Val Phe Leu Asp Ile ATT GAA TGG ACA ACC CAG GAA ACC TTT Ilie Giu Trp, Thr Thr Gin Giu Thr Phe CCA AAA TAC TTG CAT TAT Pro Lys Tyr Leu His Tyr GAC CCA GAA ACC GGA CGT CAG CTC TTG TGT GAC AAA TGT GCT CCT GGC Asp Pro Giu Thr Giy Arg Gin Leu Leu Cys Asp Lys Cys Aia Pro Gly CTG TGT GTC Leu Cys Val ACC TAC Thr Tyr AAA CAG CAC TGC ACA GTC AGG AGG AAG ACA Lys Gin His Cys Thr Vai Arg Arg Lys Thr 322 CCT TGC Pro Cys CCT GAC TAC TCT TAT ACA GAC AGC TGG Pro Asp Tyr Ser Tyr Thr Asp Ser Trp ACG AGT GAT GAA 360 Thr Ser Asp Giu
TGC
Cys GTG TAC TGC AGC CCC Val Tyr Cys Ser Pro GTG TGC AAG GAA Vai Cys Lys Giu CAG ACC GTG AAA Gin Thr Val Lys GAG TGC AAC CGC Giu Cys Asn Arg CAC AAC CGA His Asn Arg GTG TGC Vai Cys 105 GAA TGT GAG GAA Glu Cys Giu Giu GGG CGC Gly Arg 110 TAC CTG GAG CTC GAA TTC TGC TTG AAG CAC CGG AGC TGT CCC CCA GGC50 S04 Tyr Leu Glu Leu Giu Phe Cys Leu 115 His Arg Ser Cys Pro Pro Gly 125 TTG GGT Leu Gly GTG CTG Val Leu 130 CAG GCT GGG Gin Ala Gly CCA GAG CGA AAC ACG GTT TGC AAA Pro Glu Arg Asn Thr Val Cys Lys 140 AGA TGT CCG GAT Arg Cys Pro Asp 145 GGG TTC TTC Gly Phe Phe 150 TCA GGT GAG ACG TCA Ser Gly Glu Thr Ser 155 TCG AAA GCA CCC Ser Lys Ala Pro 600 AGG AAA CAC Arg Lys His ACC AAC Thr Asn 165 TGC AGC TCA CTT GGC CTC CTG CTA ATT CAG Cys Ser Ser Leu Gly Leu Leu Leu Ile Gin 170 175 AAA GGA AAT GCA Lys Gly Asn Ala ACA CAT GAC AAT GTA TGT Thr His Asp Asn Val Cys 180 185 TCC GGA AAC AGA Ser Gly Asn Arg GAA GCA Glu Ala 190 20 ACT CAA AAT TGT GGA ATA GAT Thr Gin Asn Cys Gly Ile Asp 195 AGG TTT GCT GTG CCT ACC AAG 25 Arg Phe Ala Val Pro Thr Lys 210 GTC ACC CTG TGC GAA GAG GCA TTC TTC Val Thr Leu Cys Giu Glu Ala Phe Phe 200 205 744 ATA CCG AAT TGG Ile Pro Asn Trp, CTG AGT GTT CTG Leu Ser Val Leu 220 AGT GTA GAG AGG Ser Val Glu Arg GTG GAC Val Asp 225 AGT TTG CCT GGG ACC AAA GTG AAT GCA Ser Leu Pro Gly Thr Lys Val Asn Ala 230
GAG
Glu 235 ATA AAA CGG AGA CAC AGC Ile Lys Arg Arg His Ser 240 245 CTG TGG AAG CAT CAA AAC Leu Trp Lys His Gin Asn TCG CAA GAG CAA Ser Gin Giu Gin TTC CAG CTA Phe Gin Leu CTT AAG Leu Lys 255 AGA GAC CAG Arg Asp Gin ATG GTG AAG AAG ATC ATC Met Val Lys Lys Ile Ile 270 CAA GAC ATT GAC Gin Asp Ile Asp 275 CTC TGT GAA AGC AGT Leu Cys Giu Ser Ser 280 GTG CAA CGG CAT Val Gin Arg His ATC GGC CAC Ile Giy His 285 GCG AAC CTC Aia Asn Leu 290 ACC ACA GAG CAG CTC CGC Thr Thr Giu Gin Leu Arg 295 ATC TTG ATG GAG AGC TTG CCT Ile Leu Met Giu Ser Leu Pro 300 1032 GGG AAG Giy Lys 305 AAG ATC AGC CCA GAC Lys Ile Ser Pro Asp 310 GAG ATT GAG AGA ACG AGA Giu Ile Giu Arg Thr Arg AAG, ACC TGC Lys Thr Cys 1080 *0 a a a.
AAA
Lys 320 CCC AGC GAG CAG CTC Pro Ser Giu Gin Leu 325 CTG AAG CTA CTG AGC TTG TGG AGG ATC Leu Lys Leu Leu Ser Leu Trp Arg Ile 330 1128 AAT GGA GAC CAA 20 Asn Gly Asp Gin ACC TTG AAG GGC Thr Leu Lys Giy CTG ATG Leu Met 345 TAC GCA CTC AAG CAC His 1176 Tyr Ala Leu TTG AAA GCA TAC CAC TTT CCC AAA Leu Lys Ala Tyr His Phe Pro Lys
ACC
Thr 360 GTC ACC CAC AGT Val Thr His Ser CTG AGG AAG Leu Arg Lys 365 1224 1272 ACC ATC AGG Thr Ilie Arg 370 TTC TTG CAC AGC Phe Leu His Ser ACC ATG TAC CGA TTG TAT CAG AAA Thr Met Tyr Arg Leu Tyr Gin Lys 380 CTC TTT CTA GAA ATG ATA GGG AAT Leu Phe Leu Giu Met Ile Gly Asn CAG GTT CAA TCA GTG Gin Val Gin Ser Val 395 AAG ATA AGC Lys Ile Ser 1320 TGC TTA Cys Leu 400 TAGTTAGGAA TGGTCACTGG GCTGTTTCTT CAGGATGGGC CAACACTGAT 1376 170
GGAGCAGATG
GTGGGAATGA
GGCAGGCTAT
TGAGAAAGAG
TGAGTACTCA
TTATTTTTTT
GCAAGTGCTC
GCTGCTTCTC
AGATCCTCCA
TTGATAATTG
GATATTTTTA
GAAGGCTTCT
ATTCTTTTTT
TACCACTGAG
CGGCTCTTGA
GCCCAACACA
TGCAAAGCTG
TAACCTCAAA
ACTATCTTCT
TCGGAGCTGG
CTAAATCTCC
AATGGCAGTT
CACACTGGGG
CCAGGTGTAC
CATAGGCCCT
GTGTCATCCC
GGACCGAACC
AACCCCTGAA
GATTCCTTTC
AGTCTGAGTC
ACCTAGAAAG
TTCCTTCCTC
TAGATGAAGG
CAGGGCCTTG
GGCCTCTTTC
TCATCAGTTG
AGGAGAGTGA
TCAAGCACCC
TCCTTATGGA
CCTCTTTTAT
CGCTTGCGAG
TTTCTGCCTC
1436 1496 1556 1616 1676 1736 1796 TGATAGTCTA TGACATTCTT TTGTAGGTTT CTAGGCAAGT 20 TTGCAGACTT GGCTAGACA CAGGAGTCCA GTGTTTCTTG AGTATGAAAA ATAATCAACA GGGCACTAAA AGAAACTACT CCCAATAGTT TATCCAGCTG TTACTGCATG CAGTAATTCA CTCCATTGGA TCTCTCTGAA GTGTTAAAGG CTTTTATTAA AAGACTATTA CAGTATTGCT
TTTTCTACAA
TGACCGTTAG
GCAGGGGTAG
TTCCTCTGTA
AATTTTATTC
ATATGGAGAA
TCATGCCTGG
ACTGGAAATA
TATGGGAATA
AAAGCTGATG
ATTTATATCC
TTCGTATCAG
CTATTTTTCC
GTTATGGTAG
GTTGTACCTA
CTTCTATCAA
AGAATTGATA
TTCAGTGTCT
GTAATAATAA
TCTAACTTAA
CTCTTCTGTA
ATCCAG
GTGCACGAGC
CTCTGAAGAT
TTTATTTAAC
AGCTGACTCC
CATTGGCTAG
TTGCCCCCAA
ACTGACTATG
TAATAGAAAT
GAAGCTTTGA
AAAGTTACTA
CTTATCCCAT
TTGATTCGAG
AGACTGCCAC
AAGTACATTT
CTTTGTTTCA
CGTTCAACAA
CGCCCTCTTA
AAAATCTAGA
GATTTCAGTT
ATATATCTGT
1856 1916 1976 2036 2096 2156 2216 2276 2336 2396 2432 INFORMATION FOR SEQ ID NO:i21: SEQUENCE CHARACTERISTICS: LENGTH: 401 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:i21:
S.
S S Met 1.
Asn Lys Trp Cys Cys Ala Leu Val Phe Leu Asp Ile Ile Giu Trp Thr Gin Giu Thr Phe Pro Lys Tyr Leu His Tyr Asp Pro Gly Thr S S
S.
SS
S. S S S
S.
Pro Giu Thr Giy Arg Gin Leu Leu Cys Asp Lys Cys Tyr Leu Lys Gin His Cys Thr Val Arg Arg Lys Thr Leu Cys Vai Pro Cys Pro Asp Tyr Ser Thr Asp Ser Trp Thr Ser Asp Giu Val Tyr Cys Ser Pro Vai Cys Lys Giu Leu Gin Thr Vai Lys Gin Giu Cys Asn Arg Thr His Asn Arg Vai Cys Giu Cys Glu Giu Giy Arg Tyr 1.00 105 110 Leu Giu Leu 115 Giu Phe Cys Leu Lys His Arg Ser Cys 120 Pro Pro Gly Leu 125 Gly Val Leu Gin Ala Gly 130 Pro Giu Arg Asn Thr Val Cys Lys Arg 140 Pro Asp Gly Phe Ser Gly Giu Thr Ser Lys Ala Pro Arg Lys His Thr Cys Ser Ser Leu Gly Leu Leu Leu Ile 170 Gin Lys 175 s0e *fee 0.0.
Gly Asn Ala His Asp Asn Val Cys Ser Gly Asn Arg 185 Giu Ala Thr 190
A$.
5 9 0
S.
S *504 5* *5 0
*SS.
S S 5* 4 S. S 55 'SaSS.
5
S*
*S S 4 0@ 4O Gin Asn Cys 195 Gly Ile Asp Val Leu Cys Glu Giu Ala. Phe Phe Arg 205 Ser Val Leu Val Phe Ala 210 Val Pro Thr Lys Ile Pro Asn Trp Asp 225 Ser Leu Pro Giy Thr Lys Val Asn Ala 230 Ser Val Giu Arg 25 Lys Arg Arg His Ser Gin Giu Gin Phe Gin Leu Leu Lys Leu 255 Trp Lys His Gin Asn Arg Asp Gin 260 Met Val Lys Lys Ile Ile Gin 270 Gly His Ala Asp Ile Asp 275 Leu Cys Glu Ser Ser Val Gin Arg His 280 Asn Leu 290 Thr Thr Giu Gin Arg Ile Leu Met Ser Leu Pro Gly Lys Lys Ile Ser Pro Asp Gu Ile Giu Arg Thr Arg Lys Thr Cys Lys Pro Ser Giu Gin Leu 325 Leu Lys Leu Leu Leu Trp Arg Ile Lys Asn 335 Gly Asp Gin Lys Ala Tyr Thr Leu Lys Giy Met Tyr Ala Leu Lys His Leu 350 Arg Lys Thr His Phe Pro Lys Thr 360 Val Thr His Ser Ile Arg 370 Phe Leu His Ser Thr Met Tyr Arg Leu 380 Tyr Gin Lys Leu Phe 385 Leu Glu Met Ile Asn Gin Val Gin Ser Val 395 Lys Ile Ser 0 0* 0 o.
INFORMATION FOR SEQ ID NO:i22: SEQUENCE CHARACTERISTICS: LENGTH: i324 base pairs TYPE: nucieic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: CDS LOCATION: 90.. 1292 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:122: 174 CCTTATATAA ACGTCATGAT TGCCTGGGCT GCAGAGACGC ACCTAGCACT GACCCAGCGG CTGCCTCCTG AGGTTTCCCG AGGACCACA ATG Met 1 AAC AAG TGG CTG TGC TGC GCA Asn Lys Trp, Leu Cys Cys Ala CTC CTG Leu Leu GTG CTC CTG GAC Val Leu Leu Asp ATT GAA TGG ACA ACC CAG GAA ACC CTT Ile Glu Trp Thr Thr Gin Glu Thr Leu CCT CCA AAG TAC TTG CAT TAT GAC CCA GAA ACT GGT CAT CAG CTC CTG Pro Pro Lys Tyr Leu His Tyr Asp Pro Glu Thr Gly His Gin Leu Leu 15 25 30 35 TGT GAC AAA TGT GCT CCT GGC ACC TAC CTA AAA CAG CAC TGC ACA GTG Cys Asp Lys Cys Ala Pro Gly Thr Tyr Leu Lys Gin His Cys Thr Val AGG AGG AAG ACA Arg Arg Lys Thr TTG TGT GTC CCT TGC Leu Cys Val Pro Cys 65 CCT GAC CAC TCT TAT ACG GAC Pro Asp His Ser Tyr Thr Asp 25 AGC TGG CAC ACC AGT GAT GAG TGT GTG TAT TGC AGC CCA GTG TGC AAG Ser Trp His Thr Ser Asp Glu Cys Val Tyr Cys Ser Pro Val Cys Lys GAA CTG Gfu Leu CAG TCC GTG AAG Gin Ser Val Lys GAG TGC AAC CGC ACC CAC AAC CGA GTG Giu Cys Asn Arg Thr His Asn Arg Val 100
TGT
Cys 105 GAG TGT GAG GAA GGG CGT TAC CTG GAG Giu Cys Giu Giu Gly Arg Tyr Leu Giu 110 GAA TTC TGC TTG Giu Phe Cys Leu AAG 449 Lys 120 CAC CGG AGC TGT CCC CCG GGC TCC GGC GTG GTG CAA GCT GGA ACC CCA 175 His Arg Ser Cys Pro Pro Gly Ser Gly Val Vai Gin Ala Gly Thr Pro GAG CGA AAC ACA GTT Giu Arg Asn Thr Val 140 TGC AAA AAA TGT Cys Lys Lys Cys 145 CCA GAT GGG TTC TTC TCA GGT Pro Asp Gly Phe Phe Ser Gly 150 GAG ACT TCA Giu Thr Ser 155 TCG AAA GCA CCC TGT ATA AAA CAC ACG AAC TGC AGC ACA Ser Lys Ala Pro Cys Ile Lys His Thr Asn Cys Ser Thr TTT GGC Phe Gly 170 CTC CTG CTA ATT CAG AAA Leu Leu Leu Ile Gin Lys 175 GGA AAT GCA Gly Asn Aia ACA CAT GAC AAC GTG Thr His Asp Asn Val TGT TCC GGA AAC AGA Cys Ser Giy Asn Arg 185 CTG TGT GAA GAG GCC Leu Cys Giu Giu Ala 205 GAA GCC ACG CAA AAG TGT GGA ATA GAT GTC Giu Ala Thr Gin Lys Cys Gly Ile Asp Val 689 TTC TTC AGG TTT GCT Phe Phe Arg Phe Aia 210 GTT CCT ACC AAG ATT ATA Val Pro Thr Lys Ile Ile 215 CCA AAT TGG CTG AGT GTT TTG GTG GAC Pro Asn Trp Leu Ser Val Leu Val Asp 220 225 AGT TTG CCT GGG ACC AAA GTG Ser Leu Pro Gly Thr Lys Val 230 AAT GCC GAG Asn Ala Giu 235 AGT GTA GAG AGG ATA Ser Val Giu Arg Ile 240 AAA CGG AGA CAC AGC TCA CAA GAG Lys Arg Arg His Ser Ser Gin Giu 245 CAA ACC TTC CAG CTG CTG Gin Thr Phe Gin Leu Leu 250 CTG TGG AAA CAT Leu Trp Lys His
CAA
Gin 260 AAC AGA GAC CAG Asn Arg Asp Gin GAA ATG GTG AAG AAG ATC ATC CAA GAC ATT Giu Met Val Lys Lys Ile Ile Gin Asp Ile 265 270 GAC CTC TGT GAA Asp Leu Cys Giu 275 AGC AGC Ser Ser 280 CTT CTT Leu Leu 295 GTG CAG CGG CAT CTC GGC CAC TCG AAC CTC ACC ACA GAG CAG Vai Gin Arg His Leu Gly His Ser Asn Leu Thr Thr Giu Gin GCC TTG ATG Aia Leu Met GAG AGA ACG Giu Arg Thr 315 GAG AGC CTG CCT GGG Giu Ser Leu Pro Giy 300 AGA AAG ACC TGC AAA Arg Lys Thr Cys Lys 320 AAG AAG Lys Lys 305 ATC AGC Ile Ser CCA GAA GAG ATT Pro Giu Giu Ile 310 1025 1073 TCG AGC GAG CAG CTC CTG AAG CTA Ser Ser Giu Gin Leu Leu Lys Leu 325 CTC AGT Leu Ser 330 TTA TGG AGG ATC AAA Leu Trp Arg Ile Lys 335 AAT GGT GAC Asn Giy Asp CAA GAC Gin Asp 340 ACC TTG, AAG Thr Leu Lys 1121
CTG
Leu 345 ATG TAT GCC CTC Met Tyr Aia Leu CAC TTG AAA ACA TCC His Leu Lys Thr Ser 355 CAC TTT CCC AAA His Phe Pro Lys
ACT
Thr 360 1169 GTC ACC CAC AGT Val Thr His Ser AGG AAG ACC ATG AGG Arg Lys Thr Met Arg 370 TTC CTG CAC AGC TTC ACA Phe Leu His Ser Phe Thr 375 1217 ATG TAC AGA CTG TAT CAG AAG CTC Met Tyr Arg Leu Tyr Gin Lys Leu 380 TTA GAP. ATG ATA Leu Giu Met Ile GGG AAT CAG Gly Asn Gin 390 1265 GTT CAA TCC Val Gin Ser 395 GTG AAA ATA AGC TGC TTA Val Lys Ile Ser Cys Leu 400 TAACTAGGAA TGGTCACTGG 1312 GCTGTTTCTT CA 12 1324 INFORMATION FOR SEQ ID NO:123: SEQUENCE CHARACTERISTICS: LENGTH: 401 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein Met 1 Glu (xi) SEQUENCE Asn Lys Trp Leu 5 Trp Thr Thr Gln DESCRIPTION: SEQ ID NO:123: Cys Cys Ala Leu Leu Val Leu Leu Asp Ile Ile Glu Thr Leu Lys Tyr Leu Pro Glu Thr Tyr Leu Lys Gly His Gln Leu Asp Lys Cys His Tyr Asp Pro Gly Thr Cys Val Pro Gln His Cys Arg Arg Lys Thr Thr Cys Val Pro Asp His Ser Asp Ser Trp Ser Asp Glu Tyr Cys Ser Cys Lys Glu Ser Val Lys Gln Glu Arg Tyr Cys Asn ArgAnArVaCyGuGl Asn Arg Val Cys Glu Glu Leu Giu Ile 115 Giu Phe Cys Leu His Arg Ser Cys Pro Giy Ser Giy Val Val Gin Aia Giy Pro Giu Arg Asn Thr 140 Val Cys Lys Lys Pro Asp Gly Phe Ser Gly Giu Thr Ser Lys Aia Pro Ile Lys His Thr Cys Ser Thr Phe Leu Leu Leu Ile Gin Lys 175 1.: Gly Asn Ala His Asp Asn Val Ser Gly Asn Arg Giu Ala Thr 190 Phe Phe Arg Gin Lys Cys 195 Gly Ile Asp Val Thr 200 Leu Cys Giu Giu Phe Ala 210 Val Pro Thr Lys Ile Pro Asn Trp Ser Val Leu Val Asp 225 Ser Leu Pro Giy Lys Val Asn Ala Ser Val Giu Arg Lys Arg Arg His Ser Gin Giu Gin Thr 250 Giu Met 265 Phe Gin Leu Leu Lys Leu 255 Trp Lys His Asp Ile Asp 275 Gin 260 Asn Arg Asp Gin Val Lys Lys Ile Ile Gin 270 Gly His Ser Leu Cys Giu Ser Ser 280 Val Gin Arg His Asn Leu 290 Thr Thr Giu Gin Leu 295 Leu Ala Leu Met Ser Leu Pro Giy Lys Lys Ile Ser Pro Giu Giu Ile Giu Arg Thr Arg Lys Thr Cys Lys 310 Ser Ser Giu Gin Leu Lys Leu Leu Ser 330 Leu Trp Arg Ile Lys Asn 335 Gly Asp Gin Thr Leu Lys Gly Leu 345 Met Tyr Ala Leu Lys His Leu 350 Lys Thr Ser 355 His Phe Pro Lys Vai Thr His Ser Leu Arg Lys Thr 365 Met Arg 370 Phe Leu His Ser Thr Met Tyr Arg Tyr Gin Lys Leu 15 Phe 385 Leu Giu Met Ile Asn Gin Val Gin Vai Lys Ile Ser Cys 400 INFORMATION FOR SEQ ID NO:124: SEQUENCE CHARACTERISTICS: LENGTH: 1355 base pairs TYPE: nucleic acid STRAN'DEDNESS: single TOPOLOGY: iinear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: CDS LOCATION: 94. .1296 180 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:124: GTATATATAA CGTGATGAGC GTACGGGTGC GGAGACGCAC CGGAGCGCTC GCCCAGCCGC CGCTCCAAGC CCCTGAGGTT TCCGGGGACC ACA ATG APAC AAG, TTG CTG TGC TGC Met Asn Lys Leu Leu Cys Cys GCG CTC GTG, TTT CTG GAC ATC TCC ATT Ala Leu Val Phe Leu Asp Ile Ser Ile
AAG
Lys TGG ACC Trp Thr ACC CAG, GAA ACG Thr Gin Glu Thr 4 9 *9@g *9 4 p TTT CCT Phe Pro 15 25 CCA AAG, TAC CTT CAT Pro Lys Tyr Leu His 30 TAT GAC GAA GAA ACC TCT CAT CAG CTG Tyr Asp Giu Giu Thr Ser His Gin Leu
TTG
Leu TGT GAC AAA TGT CCT Cys Asp Lys Cys Pro CCT GGT ACC TAC Pro Gly Thr Tyr AAA CAA CAC TGT Lys Gin His Cys .4 4 9 4.
9 .4 .4 9. GCA AAG TGG AAG ACC Ala Lys Trp Lys Thr GTG TGC GCC CCT TGC CCT GAC CAC TAC TAC ACA Val Cys Ala Pro Cys Pro Asp His Tyr Tyr Thr 306 25 GAC AGC TGG Asp Ser Trp ACC AGT GAC GAG Thr Ser Asp Glu CTA TAC TGC AGC Leu Tyr Cys Ser CCC GTG TGC Pro Vai Cys AAG GAG CTG Lys Glu Leu CAG TAC GTC AAG Gin Tyr Val Lys GAG TGC AAT CGC ACC CAC AAC CGC Glu Cys Asn Arg Thr His Asn Arg 100 GTG, TGC Val Cys 105 GAA TGC Glu Cys AAG GAA GGG CGC Lys Glu Gly Arg 110 TAC CTT GAG ATA GAG Tyr Leu Glu Ile Glu 115 TTC TGC TTG, Phe Cys Leu AAA CAT AGG AGC TGC CCT CCT GGA TTT GGA GTG GTG CAA GCT GGA ACC 181 His Arg Ser Cys Pro Pro Gly Phe Gly Val Val Gin Ala Gly Lys 120 CCA GAG CGA AAT ACA Pro Giu Arg Asn Thr 140 GTT TGC AAA AGA TGT Vai Cys Lys Arg Cys 145 CCA GAT Pro Asp GGG TTC TTC TCA Giy Phe Phe Ser 150 AAT GAG ACG TCA Asn Giu Thr Ser 155 TCT AAA GCA CCC TGT AGA AAA CAC ACA AAT TGC AGT Ser Lys Ala Pro Cys Arg Lys His Thr Asn Cys Ser 9@ GTC TTT GGT CTC CTG CTA ACT CAG AAA GGA AAT GCA ACA CAC GAC AAC Val Phe Gly Leu Leu Leu Thr Gin Lys Gly Asn Ala Thr His Asp Asn 175 ATA TGT TCC GGA AAC AGT GAA TCA ACT CAA AAA TGT GGA ATA GAT GTT 20 Ile Cys Ser Gly Asn Ser Giu Ser Thr Gin Lys Cys Gly Ile Asp Vai
ACC
Thr 25 200 CTG TGT GAG GAG GCA Leu Cys Giu Giu Ala 205 TTC TTC AGG TTT GOT GTT CCT ACA AAG Phe Phe Arg Phe Ala Val Pro Thr Lys 210 TTT 738 Phe 215 ACG CCT AAC TGG Thr Pro Asn Trp AGT GTC TTG GTA Ser Val Leu Val GAC AAT TTG COT GGC ACC AAA Asp Asn Leu Pro Gly Thr Lys 225 230 AAA CGG CAA CAC AGC TCA CAA Lys Arg Gin His Ser Ser Gin 245 *GTA AAC GCA Val Asn Ala AGT GTA GAG AGG Ser Vai Giu Arg
ATA
Ile 240 3 5 GAA CAG ACT TTC CAG OTG Giu Gin Thr Phe Gin Leu 250 CTG AAG TTA TGG Leu Lys Leu Trp 255 AAA OAT Lys His CAA AAC AAA GC Gin Asn Lys Ala 260 182 CAA GAT ATA GTC AAG AAG ATC ATC CAA GAT ATT GAC CTC TGT GAA AAC Gin Asp 265 Ile Val Lys Lys Ile Ile Gin Asp Ilie Asp Leu Cys Giu Asn GTG CAG CGG CAC ATT Vai Gin Arg His Ile 285 GGA CAT GCT AAC CTC Giy His Ala Asn Leu 290 ACC TTC GAG CAG CTT Thr Phe Giu Gin Leu 295 GTG GGA GCA GAA GAC Val Giy Aia Giu Asp 310 CGT AGC TTG ATG Arg Ser Leu Met ATT GAA AAA ACA Ile Giu Lys Thr 315
GAA
Giu 300 AGC TTA CCG GGA AAG AAA Ser Leu Pro Giy Lys Lys 305 1026 ATA AAG GCA TGC AAA Ile Lys Ala Cys Lys CCC AGT GAC Pro Ser Asp CAG ATC CTG AAG Gin Ile Leu Lys 1074 CTG CTC AGT Leu Leu Ser 330 TTG TGG CGA ATA Leu Trp Arg Ile AAT GGC GAC CAA GAC ACC TTG AAG Asn Gly Asp Gin Asp Thr Leu Lys 340 1122 GGC CTA Gly Leu 345 ATG CAC GCA CTA AAG CAC Met His Ala Leu Lys His 350 TCA AAG ACG TAC CAC Ser Lys Thr Tyr His 355 TTT CCC AAA Phe Pro Lys 1170 1218
ACT
Thr 360 GTC ACT CAG AGT CTA Val Thr Gin Ser Leu 365 AAG AAG ACC ATC AGG Lys Lys Thr Ile Arg 370 TTC CTT CAC AGC Phe Leu His Ser ACA ATG TAC AAA TTG TAT CAG AAG TTA TTT TTA GAA ATG Thr Met Tyr Lys Leu Tyr Gin Lys Leu Phe Leu Giu Met ATA GGT AAC Ile Gly Asn 390 1266 CAG GTC CAA Gin Val Gin GTA AAA ATA AGC Val Lys Ile Ser TTA TAACTGGAAA TGGCCATTGA Leu 1316 183 GCTGTTTCCT CACAATTGGC GAGATCCCAT GGATGATAA 1355 INFORMATION FOR SEQ ID NO:125: SEQUENCE CHARACTERISTICS: LENGTH: 401 amino acids TYPE: amino acid .0 TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE Asn Lys Leu Leu 5 Trp Thr Thr Gin Giu Thr Ser His DESCRIPTION: SEQ ID NO:125: Cys Cys Ala Leu Val Phe Leu Asp Ile Ser Ile Glu Thr Phe Lys Tyr Leu Giu Gin Leu Asp Lys Cys His Tyr Asp Pro Gly Thr Cys Ala Pro Tyr Leu Cvs Pro Lys Gin His Cys Lys Trp Lys Thr Asp His Tyr Asp Ser Trp, Thr Ser Asp Glu Leu Tyr Cys Ser Val Cys Lys Giu Tyr Val Lys Cys Asn Arg Asn Arg Val Giu Cys Lys Glu Gly Arg Tyr 110 Leu Giu Ile Giu Phe Cys Leu Lys His Arg Ser Cys Pro Pro Giy Phe 125 Gly Val Val Gin Ala Gly Thr Pro Glu Arg Asn Val Cys Lys Arg Cys Pro Asp Gly Phe Phe Ser Asn Glu Thr Ser Lys Ala Pro Arg Lys His Thr Cys Ser Val Phe Leu Leu Leu Thr Gin Lys 175 Gly Asn Ala His Asp Asn Ile Cys 185 Ser Gly Asn Ser Glu Ser Thr 190 Gin Lys Cys 195 Gly Ile Asp Val Leu Cys Giu Giu Phe Phe Arg Phe Ala 210 Val Pro Thr Lys Phe Thr Pro Asn Trp 215 Ser Val Leu Val Asp Asn Leu Pro Giy 225 Lys Val Asn Ala Ser Val Giu Arg Lys Arg Gin His Ser 245 Ser Gin Giu Gin Phe Gin Leu Leu Lys Leu 255 Trp Lys His Asp Ile Asp 275 Asn Lys Ala Gin Asp 265 Ile Val Lys Lys Ile Ile Gin 270 Gly His Ala Leu Cys Giu Asn Val Gin Arg His Asn Leu Thr Phe Giu 290 Gin Leu 295 Giu Asp 310 Arg Ser Leu Met Giu Ser Leu Pro Giy 300 Lys Val Gly Ala Ile Giu Lys Thr 315 Ile Lys Ala Cys Lys Pro Ser Asp Gin Ile 325 Thr Leu Lys Leu Leu Leu Trp Arg Ile Lys Asn 335 Gly Asp Gin Leu Lys Giy Leu 345 Val His Ala Leu Lys Thr Tyr 355 Ile Arg Phe 370 Phe Pro Lys Thr Gin Ser Lys His Ser 350 Lys Lys Thr Gin Lys Leu Leu His Ser Met Tyr Lys Phe 385 Leu Leu Giu Met Ile Giy 390 Gin Val Gin Ser 395 Lys Ile Ser 20 INFORMATION FOR SEQ ID NO:126: SEQUENCE CHARACTERISTICS: LENGTH: 139 amino acids TYPE: amino acid 25 STRANDEDNESS: singie TOPOLOGY: iinear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:126: Cys Pro Gin Giy Lys Tyr Ile His Pro Gin Asn Asn Ser Ile Cys Cys 1 5 10 Thr Lys Cys His Lys Gly Thr Tyr Leu Tyr Asn Asp cys 25 Pro Gly Pro Gly Gin Asp Thr Asp Cys Arg Glu Cys Glu Ser Gly Ser Phe Thr Ala Ser Glu Asn His Leu Arg His Cys Leu Ser Cys Lys Cys Arg Lys Glu Met Gly Gin Val Giu Ile Ser Ser Cys Vai Asp Arg Asp Thr Val Cys Giy Cys Lys Asn Gin Tyr Arg His Tyr Trp Ser Giu Asn Leu Phe Gin Cys 100 Phe Asn Cys Ser Leu Cys Leu Asn 105 Gly Thr Val His 110 Leu Ser Cys Gin Glu Lys Gin Asn Thr Val Cys Thr Cys His Ala Gly 115 120 125 Phe Phe 130 Leu Arg Giu Asn Cys Val Ser Cys INFORMATION FOR SEQ ID NO:127: SEQUENCE CHARACTERISTICS: LENGTH: 48 base pairs TYPE: nucleic acid STRMAJDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 187 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:127: CCGGCGGACA TTTATCACAC AGCAGCTGAT GAGAAGTTTC TTCATCCA INFORMATION FOR SEQ ID NO:128: SEQUENCE CHARACTERISTICS: LENGTH: 219 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) Met 1 Arg SEQUENCE DESCRIPTION: SEQ II Leu Gly Ile Trp Thr Leu Leu 5 Leu Ser Ser Lys Ser Val Asn )NO: 128: Pro Leu Val Leu Thr Ser Val Ala 10 Ala Gln Val Thr Asp Ile Asn Ser Val Thr Thr Val Glu Thr Gin Asn Gin Phe Cys His Lys Pro Cys Pro Lys Gly Leu Leu Glu Gly Leu Arg Lys Leu His His Pro Gly Glu Arg Lys Ala Arg Asp Cys Thr Vai Asn Gly Asp Giu Pro Tyr Thr Asp Lys Ala His Asp Cys Val Pro Cys Gin Giu Gly Lys Phe Ser Ser Leu Giu Val 115 Cys Arg Arg Cys Arg 105 Leu Cys Asp Giu Gly His Gly 110 Lys Cys Arg 9.
9* 9* 9 9* Giu Ile Asn Cys Arg Thr Gin Asn Cys Lys 130 Pro Asn Phe Phe Asn Ser Thr Val Cys Giu His Cys Asp 140 Cys Thr Lys Cys Giu His Giy Ile Ilie 150 Lys 155 Giu Cys Thr Leu Thr 160 Gly Trp 175 Ser Asn Thr Lys Lys Glu Glu Gly Arg Ser Asn Leu Leu Cys Leu Leu Leu Leu Pro Ile Pro Leu Ile Val Trp Val Lys Arg 190 Lys Giu Val 195 Gin Lys Thr Cys Lys His Arg Lys Giu Asn Gin Gly 205 Ser His Giu Ser Pro Thr 210 Asn Pro Giu Thr INFORMATION FOR SEQ ID NO:129: SEQUENCE CHARACTERISTICS: LENGTH: 280 amino acids TYPE: amino acid STRANDfEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:129: Met Gly Leu Ser Thr Val Pro Asp Leu Leu Leu Pro Leu Val Leu Leu Leu Leu Val 20 Leu Gly Asp Gly Ile Tyr Pro Ser Gly 25 Asp Ser His Arg Giu Lys Val Ile Gly Val Cys Pro Cys Thr Lys Leu Val Pro Gin Giy Lys Cys His Lys 35 Tyr Ile His Pro Gin Asn Ile Cys Giv Thr Tyr Leu Tyr Asn 70 Cys Pro Gly Pro Gin Asp Thr Asp His Leu Arg Giu Cys Giu Ser Ser Giy Ser Cys Ser Lys Arg His Cys Phe Thr 90 Cys Arg 105 Arg Asp Ala Ser Giu Asn Giu Ile Ser 115 Lys Asn Gin 130 Cys Thr Vai Lys Giu Met Thr Val Cys 125 Asn Leu Phe Giy Gin Val 110 Gly Cys Arg Gin Cys Phe Tyr Arg His Ser Giu Asn Cys Ser Leu Cys Leu Asn Giy Thr Vai His Leu Ser Cys Gin Giu 145 145 150 Lys Gin Asn Thr Cys Thr Cys His Ala Giy Phe Phe Leu Arg Giu 170 175 Asn Giu Cys Val Ser Cys Ser Asn 180 Cys 185 Lys Lys Ser Leu Giu Cys Thr 190 @0g@ So..
0 9SS@ 9 @9 00 S Lys Leu Cys 195 Leu Pro Gin Ile Giu Asn Val Lys Gly Thr Giu Asp Ser Gly Thr 210 Thr Val Leu Leu Leu Val Ile Phe Phe Giy Leu Cys Leu 220 Leu 225 Ser Leu Leu Phe Ile 230 Gly Leu Met Tyr Tyr Gin Arg Tr~p Ser Lys Leu Tyr Ile Val Cys Gly Ser Thr Pro Giu Lys Giu 255 SC' 0 9*
S.
05 G 0@
*S
4 0 5*
S.
Giy Giu Leu Phe Ser Pro 275 Giy Thr Thr Thr Pro Leu Ala Pro Asn Pro Ser 270 Thr Pro Gly Phe INFORMATION FOR SEQ ID NO:130: SEQUENCE CHARACTERISTICS: LENGTH: 207 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:130: Met Leu Arg Leu Ile Ala Leu Leu Val q.
Val Val Tyr Lys Cys Gly Asp Asp Val Pro 20 Tyr Giu Lys Asp Ala Ser Arg Leu Tyr Ser Ser Asn Gin 25 Al a Vai Tyr Giy Gly His Asp Giy Phe Tyr Ser Pro Cys Gly Leu Cys Ser Cys His Cys Giy 50 Giu Asp Pro Ala Giy Ser Asn Thr Giy Thr Phe Ser Thr Asn Pro Aia Cys Cys Arg Giy Pro Thr Giy His Glu Ser Gin Pro Cys Asp Arg Thr His 100 Gly Asp Arg Vai Cys Cys Ser Thr Giy Asn Tyr Cys 110 Gin Thr Lys Leu Leu Lys 115 Gin Asn Giy Ilie Cys Ala Cys Pro Aia Giy Tyr Gly Vai Ser Gly His Thr Arg Aia Giy Asp Thr 130 135 140 Leu 145 Thr Cys Glu Lys Cys Pro 150 Thr 192 Pro His Thr Ser Phe Asn Tyr Ser 155 Tyr Ile Asp Ser Leu Ser Val Gly Glu Arg Cys Ser Pro 160 Phe Asn 175 His Asn Leu Tyr Pro Asn Glu Thr Ser Thr Thr Ala Gly 190 Tyr Thr r Glu Val Ile Lys Thr Lys Glu P) 195 2' INFORMATION FOR SEQ ID NO:131: he 00 Val Thr Leu Asn 205 SEQUENCE CHARACTERISTICS: LENGTH: 227 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:131: Met Ala Pro Val Ala Val Trp Ala Ala Leu Ala 1 5 10 Trp Ala Ala Ala His Ala Leu Pro Ala Gin Val 25 Ala Pro Glu Pro Gly Ser Thr Cys Arg Leu Arg 40 Thr Ala Gin Met Cys Cys Ser Lys Cys Ser Pro Val Gly Leu Glu Leu Ala Phe Thr Pro Tyr Glu Tyr Tyr Asp Gin Gly Gln His Ala Lys 193 55 Val Phe Cys Thr Lys Thr Ser Asp Thr Val Cys Asp Ser Cys Glu Asp 70 75 Ser Thr Tyr Thr Gin Leu Trp Asn Trp Vai Pro Giu Cys Leu Ser Cys 90 Giy Ser Arg Cys Ser Ser Asp Gin Val Giu Thr Gin Ala Cys Thr Arg 100 105 110 Giu Gin Asn Arg Ile Cys Thr Cys Arg Pro Gly Trp Tyr Cys Ala Leu 115 120 125 Ser Lys Gin Giu Gly Cys Arg Leu Cys Ala Pro Leu Arg Lys Cys Arg 130 135 140 *.Pro Gly Phe Gly Vai Ala Arg Pro Gly Thr Glu Thr Ser Asp Val Val 145 150 155 160 Cys Lys Pro Cys Ala Pro Gly Thr Phe Ser Asn Thr Thr Ser Ser Thr 165 170 175 :25 Asp Ile Cys Arg Pro His Gin Ile Cys Asn Val Val Ala Ile Pro Gly 180 185 190 Asn Ala Ser Arg Asp Ala Val Cys Thr Ser Thr Ser Pro Thr Arg Ser 195 200 205 Met Ala Pro Gly Ala Val His Leu Pro Gin Pro Val Ser Thr Arg Ser 210 215 220 Gin His Thr 225 INFORMATION FOR SEQ ID NO:132: SEQUENCE CHARACTERISTICS: LENGTH: 197 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:132: r Met Val Ser Leu Pro Arg Leu Cys Ala Leu Trp Gly Cys Leu Leu Thr Ala Val His Leu Gly Gin Cys Val Cys Ser Asp Lys Gin Tyr Leu Arg Leu Thr His Asp Gly Gin Cys Cys Asp Leu Cys Gin Pro Gly Ser His Cys Thr Ala Leu Lys Thr Gin Cys Pro Cys Asp Ser Gly Glu Phe Ser Ala Trp Asn Arg Glu Arg Cys His Gin Arg His Cys Glu Pro Asn Gin Gly Leu Val Lys Lys Glu Gly Thr Ala Glu Ser Asp Thr Val Cys Thr Cys Lys Glu Gly Gin His Cys Thr Ser Lys Asp 115 Cys Glu Ala Cys Ala Gin His Thr Pro Cys Ile Pro Gly Phe Gly 130 Pro Cys Val Met Glu Met Ala 135 Phe Thr Glu Thr Ser Asn Gin Pro Val Gly 145 Cys Thr Asp Thr Val Cys His 140 Ser Ser Leu Phe Glu Lys 155 160 Asn Leu Glu Val Leu Gin 175 Gly Leu Lys Ser Arg Met 190 Tyr Pro Trp Thr 165 Ser Gin 180 Cys Glu Asp Lys Gly Thr Thr Asn Val Arg Ala Leu Leu Val 195 INFORMATION FOR SEQ ID NO:133: SEQUENCE CHARACTERISTICS: LENGTH: 208 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) Met 1 Glu SEQUENCE DESCRIPTION: SEQ ID NO:133: Asn Lys Trp Leu Cys Cys Ala Leu Leu Val Phe Leu Asp Ile Ile 5 10 Trp Thr Thr Gin Glu Thr Phe Pro Pro Lys Tyr Leu His Tyr Asp 25 196 Pro Giu Thr Gly Arg Gin Leu Leu Cys Asp Lys Cys Ala Pro Gly Thr Tyr Leu Lys Gin His Cys Val Arg Arg Lys Leu Cys Val Pro Pro Asp Tyr Ser Thr Asp Ser Trp Thr Ser Asp Glu Val Tyr Cys Ser Val Cys Lys Giu Gin Thr Val Lys Gin Glu 0.
Cys Asn Arg Leu Glu Leu 115 His Asn Arg Val Glu Cys Giu Glu Gly Arg Tyr 110 Pro Gly Leu Giu Phe Cys Leu His Arg Ser Cys Gly Val 130 Leu Gin Ala Gly Pro Giu Arg Asn Val Cys Lys Arg Pro Asp Gly Phe Ser Gly Giu Thr Ser Lys Ala Pro Arg Lys His Thr Cys Ser Ser Leu Gly Leu Leu Leu Ilie 170 Gin Lys 175 Gly Asn Ala His Asp Asn Val Cys Ser Gly Asn Arg 185 Giu Ala Thr 190 Phe Phe Arg Gin Asn Cys 195 Gly Ile Asp Val Thr Leu Cys Glu Giu Ala 200 205 INFORMATION FOR SEQ ID NO:134: SEQUENCE CHARACTERISTICS: LENGTH: 224 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:134: Met Gly Ala Gly Ala Thr Gly Arg Ala Met Asp Gly Pro Arg Leu Leu Leu Leu Leu Leu Leu Gly Val Ser Gly Gly Ala Lys Glu Ala Cys Pro Thr Gly Leu Tyr Thr His Ser Gly Glu Cys Cys Lys Ala Cys Asn Leu Gly Glu Gly Val Ala Gln Pro Cys Gly Ala Gln Thr Val Cys Glu Pro Cys Leu Asp Val Thr Phe Ser Val Val Ser Ala Thr s0 Glu Pro Cys Lys Pro Cys Thr Glu Cys Gly Leu Gln Ser Met Ser Ala Pro Cys Val Glu Ala Asp Asp Val Cys Arg Cys Ala Tyr Gly 110 Tyr Tyr Gin Asp Glu Thr Thr Gly Arg Cys Glu Ala Cys Arg Val Cys 125 Lys Gin Asn Thr Giu Ala Gly 130 Ser Gly Leu Phe Ser Cys Gin Val Cys Glu Glu Cys 145 Asp Gly Thr Tyr Ser Asp Glu Ala Asn His I155C 160 Val Asp Pro Cys Leu 165 Pro Cys Thr Val Giu Asp Thr Glu Arg Gin 175 Leu Arg Giu Thr Arg Trp Ala Asp 185 Ala Glu Cys Glu Glu Ile Pro 190 Gly Arg Trp 195 Ile Thr Arg Ser Pro Pro Glu Gly Ser 205 Glu Gin 220 Asp Ser Thr Asp Leu Ile Ala Pro 210 Ser Thr Gin Glu Glu Ala Pro Pro INFORMATION FOR SEQ ID NO:135: SEQUENCE CHARACTERISTICS: LENGTH: 205 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:135: 199 Met Tyr Val Trp, Val Gin Gin Pro Thr Ala Phe Leu Leu Leu Gly Leu Cys Val Lys Asp Leu Gly Val Gly His Lys Thr Val Lys Leu Asn 25 Cys Thr Tyr Pro Gly Met Val Cys Glu Pro Ser Cys Cys Arg Gin Pro Gly Ser Arg Cys Asp His Thr Thr Val Cys Glv Phe Tyr Asn Giu Asn Tyr Asp Lys Gin Cys Cys Asn His Ser Gly Ser Giu Gin Asn Cys Thr Pro Thr Giu Asp Cys Gin Cys Giy Thr Gin Asp Ser Ser 115 His Phe Ser Lys Leu Giy Cys Val Pro Pro Arg Gin 110 Pro Pro Gly Thr Asn Cys Pro Gly Ser Ala Cys Lys 130 Thr Leu Ser Gly Lys Arg His Pro Ala 155 Thr Asn Ser Leu Val Cys Giu Ser Leu Leu Leu Leu Trp Giu Thr 175 Ser Thr Thr Val Trp 190 Gin Arg Thr Thr 180 Arg Pro Thr Pro Pro Arg Thr Ser Gin Leu Pro Ser Thr Pro Thr Leu Val 205 INFORMATION FOR SEQ ID NO:136: SEQUENCE CHARACTERISTICS: LENGTH: 191 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear 0O** *4
S
9*
S
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:136: Gly Asn Asn Cys Tyr Asn Val Val 5 Ile Val Leu Leu Leu Val Gly Cys Glu Lys Val Gly Ala Val Gln Asn Ser Cys Asp Asn Cys Gln S. 9
SS
OS
5* 4
*S
S*
Pro Gly Thr 35 Phe Cys Arg Lys Tyr Asn Pro Val Cys Ser Cys Pro Pro Ser Thr Phe Ser Ser Gly Gly Gln Pro Cys Asn Ile Cys Arg Val Cys Ala Gly Phe Arg Phe Lys Lys Phe Cys Ser Ser His Asn Ala Glu Cys Glu Cys Ile Glu Gly Phe His Cys Leu Gly Pro Gln Cys Thr Cys Glu Lys Asp Cys Arg Pro Gly Gln Glu Leu Thr 110 Lys Gin Gly 115 Cys Lys Thr Cys Leu Gly Thr Phe Asp Gin Asn Asp Gly Arg Gly Thr 13 0 Ser Val 145 Pro Val Gly Val Cys Arg Thr Asn Cys 4.9 *00 Leu Lys Thr Val Ser Phe 165 Thr Glu Lys Val Cys Gly Pro Ser Thr Ser Val Thr Pro 175 Phe Leu 190 Gly Gly Pro Gly Gly His Ser Lei 180 INFORMATION FOR SEQ ID NO:137: SEQUENCE CHARACTERISTICS: LENGTH: 54 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA u1 Leu Thr Leu (xi) SEQUENCE DESCRIPTION: SEQ ID NO:137: TATGGATGAA GAAACTTCTC ATCAGCTGCT GTGTGATAAA TGTCCGCCGG GTAC INFORMATION FOR SEQ ID NO:138: SEQUENCE CHARACTERISTICS: 202 LENGTH: 380 amino acids TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:138: a a. a Glu Thr Leu Pro Pro Lys Tyr Leu His Tyr Asp Pro Glu Leu Leu Cys 20 Thr Val Arg Lys Cys Ala Gly Thr Tyr Leu Thr Gly His Lys Gin His Asp His Ser Cys Ser Pro Cys Arg Lys Thr 35 Tyr Thr Asp Leu 40 Ser Val Pro Cys Pro Tyr Ser Trp His Asp Glu Cys Val Cys Lys Glu Leu Val Lys Gin Asn Arg Thr Arg Val Cys Glu Arg Cys Glu Glu Ser Cys Pro Gly Arg 90 Tyr Leu Glu Ile Giu Phe Cys Leu Lys Gly Thr Pro 115 Phe Ser Gly 130 Pro 105 Cys Gly Ser Gly Val Arg Asn Thr Lys Lys Cys Val Gin Ala 110 Asp Gly Phe His Thr Asn Glu Thr Ser Ala Pro Cys Cys Ser Thr Phe Gly 145 Leu Leu Ile Gin Gly Asn Ala Thr Asp Asn Val Cys Gly Asn Arg Giu Thr Gin Lys Cys Gly Ile 175 Asp Val Thr Leu Cys Giu Giu Aia 180 Phe Arg Phe Ala Val Pro Thr 190 Leu Pro Gly Lys Ilie Ile 195 Pro Asn Trp Leu Ser Vai Leu Val Asp 200 0 Thr Lys 210 Val Asn Ala Glu Vai Giu Arg Ile Arg Arg His Ser Gin Giu Gin Thr Phe 230 Gin Leu Leu Lys Trp Lys His Gin Arg Asp Gin Giu Val Lys Lys Ile Gin Asp Ile Asp Leu Cys 255 Giu Ser Ser Gin Arg His Leu His Ser Asn Leu Thr Thr Giu 270 Ile Ser Pro Gin Leu Leu 275 Ala Leu Met Giu Leu Pro Gly Lys Glu Giu 290 Ile Giu Arg Thr Lys Thr Cys Lys Ser Giu Gin Leu Leu Lys Leu Leu Ser 305 Leu Lys Gly Leu Met Trp Arg Ile Lys Asn Gly Asp Gin Asp 315 Tyr Ala Leu Lys Leu Lys Thr Ser His Phe 335 204 Pro Lys Thr Val Thr His Ser Leu Arg Lys Thr Met Arg Phe Leu His 340 345 350 Ser Phe Thr Met Tyr Arg Leu Tyr Gin Lys Leu Phe Leu Glu Met Ile 355 360 365 Gly Asn Gin Val Gin Ser Val Lys Ile Ser Cys Leu 370 375 380 INFORMATION FOR SEQ ID NO:139: SEQUENCE CHARACTERISTICS: LENGTH: 380 amino acids 15 25 TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:139: Glu Thr Phe Pro Pro Lys Tyr Leu His Tyr Asp Glu Glu Thr Ser His 1 5 10 Gin Leu Leu Cys Asp Lys Cys Pro Pro Gly Thr Tyr Leu Lys Gin His 25 Cys Thr Ala Lys Trp Lys Thr Val Cys Ala Pro Cys Pro Asp His Tyr 40 Tyr Thr Asp Ser Trp His Thr Ser Asp Glu Cys Leu Tyr Cys Ser Pro 55 205 Val Cys Lys Giu Leu Gin Tyr Val Lys Gin Glu Cys Asn Arg Thr Asn Arg Val Cys Giu Cys Lys Giu Gly Arg Tyr Leu Giu Ile 90 Giu Phe Cys Leu Lys His Arg Ser Cys Pro 100 Gly Phe Gly Val Vai Gin Ala 110 Gly Thr Pro 115 Giu Arg Asn Thr Cys Lys Arg Cys Asp Giy Phe Phe Ser Asn Giu Thr Ser Ser Lys Ala Pro Cys Arg Lys His Thr Asn is 130 135 140 Cys 5 145 Ser Vai Phe Gly Leu Leu Thr Gin Lys 155 Gly Asn Aia Thr Asp Asn Ile Cys Ser Giy Asn Ser Giu 165 Thr Gin Lys Cys Giy Ile 175
S
S
Asp Val Thr Lys Phe Thr 195 Cys Giu Giu Ala Phe Arg Phe Ala Val Pro Thr 190 Pro Asn Trp Leu Val Leu Val Asp Leu Pro Gly Thr Lys 210 Val Asn Ala Giu Val Giu Arg Ile Arg Gin His Ser Ser Gin Giu Gin Thr 225 Gin Leu Leu Lys Leu Trp 235 Lys His Gin Lys Ala Gin Asp Ile 245 Val Lys Lys Ile Ile Gin Asp Ile Asp Leu Cys 250 255 Glu Asn Ser Val 260 Gin Arg His Ile His Ala Asn Leu Thr Phe Glu 270 Val Gly Ala Gin Leu Arg Ser Leu Met Glu 275 Ser 280 Leu Pro Gly Lys Glu Asp 290 Ile Glu Lys Thr Ile Lys Ala Cys Lys 295 Ser Asp Gin Ile r Leu Lys Leu Leu Ser 305 Trp Arg Ile Lys Asn 315 Gly Asp Gin Asp Leu Lys Gly Leu Met His Ala Leu Lys His Ser Lys Thr Tyr His Phe S* 15 325 330 335 Pro Lys Thr Val 340 Thr Gin Ser Leu Lys Thr Ile Arg Phe Leu His 350 Ser Phe Thr 355 Met Tyr Lys Leu Gin Lys Leu Phe Leu Glu Met Ile 365 Gly Asn 370 Gin Val Gin Ser Lys Ile Ser Cys INFORMATION FOR SEQ ID NO:140: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 207 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:140: TGGACCACCC AGAAGTACCT TCATTATGAC INFORMATION FOR SEQ ID NO:141: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA o (xi) SEQUENCE DESCRIPTION: SEQ ID NO:141: GTCATAATGA AGGTACTTCT GGGTGGTCCA a. 25 INFORMATION FOR SEQ ID NO:142: SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 208 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:142: GGACCACCCA GCTTCATTAT GACGAAGAAA C 31 INFORMATION FOR SEQ ID NO:143: SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:143: INFORMATION FOR SEQ ID NO:144: SEQUENCE CHARACTERISTICS: 25 LENGTH: 29 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 20 (ii)GTTTCTTCGT CATAATGAAGYPE: CTGGGTGGTC C 31 (xi) SEQUENCE DESCRIP(2) INFORMATION FOR SEQ ID NO:144: GTGGACCACC SEQUENCE CAGG ACGAAG AAACCTCTC 29 25 LENGTH: 29 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID N0:144: GTGGACCACC CAGGACGAAG AAACCTCTC 29 209 INFORMATION FOR SEQ ID NO:145: SEQUENCE CHARACTERISTICS: LENGTH: 29 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA GAGAGGTTTC TTCGTCCTGG GTGGTCCAC 29 (xi) SEQUENCE DESCRIPTION: SEQ ID N46:145: INFORMATION FOR SEQ ID Nacid:146: SEQUENCE CHARACTERISTICS: LENGTH: 29 base pairs TYPE: nucleic acidN (xi) SEQUENCE DESCRIPTION: SEQ ID NO:146: CGTTTCCTCC AAAGTTCCTT CATTATGAC 29 INFORMATION FOR SEQ ID NO:147: SEQUENCE CHARACTERISTICS: SEQUENCE CHARACTERISTICS 210 LENGTH: 29 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:147: se*@ GTCATAATGA AGGAACTTTG GAGGAAACG 29 *e INFORMATION FOR SEQ ID NO:148: 4 SEQUENCE CHARACTERISTICS: LENGTH: 32 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:148: GGAAACGTTT CCTGCAAAGT ACCTTCATTA TG 32 INFORMATION FOR SEQ ID NO:149: SEQUENCE CHARACTERISTICS: LENGTH: 32 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 211 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:149: CATAATGAAG GTACTTTGCA GGAAACGTTT CC 32 o. INFORMATION FOR SEQ ID NO:150: SEQUENCE CHARACTERISTICS: o0 LENGTH: 27 base pairs S15 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:150: 25 CACGCAAAAG TCGGGAATAG ATGTCAC 27 INFORMATION FOR SEQ ID NO:151: SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 212 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:151: GTGACATCTA TTCCCGACTT TTGCGTG 27 INFORMATION FOR SEQ ID NO:152: SEQUENCE CHARACTERISTICS: LENGTH: 25 base pairs TYPE: nucleic acid *o STRANDEDNESS: single TOPOLOGY: linear 15 (ii) MOLECULE TYPE: cDNA o e (xi) SEQUENCE DESCRIPTION: SEQ ID NO:152: CACCCTGTCG GAAGAGGCCT TCTTC INFORMATION FOR SEQ ID NO:153: S. SEQUENCE CHARACTERISTICS: LENGTH: 25 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 213 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:153: GAAGAAGGCC TCTTCCGACA GGGTG INFORMATION FOR SEQ ID NO:154: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single o TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:154: TGACCTCTCG GAAAGCAGCG TGCA 24 INFORMATION FOR SEQ ID NO:155: 25 SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:155: 214 TGCACGCTGC TTTCCGAGAG GTCA 24 INFORMATION FOR SEQ ID NO:156: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:156: e CCTCGAAATC GAGCGAGCAG CTCC 24 INFORMATION FOR SEQ ID NO:157: SEQUENCE CHARACTERISTICS: LENGTH: 25 base pairs TYPE: nucleic acid STRANDEDNESS: single 25 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:157: CGATTTCGAG GTCTTTCTCG TTCTC INFORMATION FOR SEQ ID NO:158: 215 SEQUENCE CHARACTERISTICS: LENGTH: 33 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:158: CCGTGAAAAT AAGCTCGTTA TAACTAGGAA TGG 33 INFORMATION FOR SEQ ID NO:159: SEQUENCE CHARACTERISTICS: LENGTH: 33 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:159: CCATTCCTAG TTATAACGAG CTTATTTTCA CGG 33 INFORMATION FOR SEQ ID NO:160: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single 216 TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:160: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 INFORMATION FOR SEQ ID NO:161: SEQUENCE CHARACTERISTICS: LENGTH: 44 base pairs TYPE: nucleic acid STRANDEDNESS: single S* TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 25 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:161: CCTCTCTCGA GTCAGGTGAC ATCTATTCCA CACTTTTGCG TGGC 44 INFORMATION FOR SEQ ID NO:162: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 217 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:162: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 INFORMATION FOR SEQ ID NO:163: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA S* (xi) SEQUENCE DESCRIPTION: SEQ ID NO:163: CCTCTCTCGA GTCAAGGAAC AGCAAACCTG AAGAAGGC 38 .e INFORMATION FOR SEQ ID NO:164: SEQUENCE CHARACTERISTICS: 25 LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:164: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 218 INFORMATION FOR SEQ ID NO:165: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:165: CCTCTCTCGA GTCACTCTGT GGTGAGGTTC GAGTGGCC 38 INFORMATION FOR SEQ ID NO:166: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 25 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:166: CCTCTGAGCT CAAGCTTCCG AGGACCACAA TGAACAAG 38 INFORMATION FOR SEQ ID NO:167: SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs 219 TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:167: CCTCTCTCGA GTCAGGATGT TTTCAAGTGC TTGAGGGC 38 INFORMATION FOR SEQ ID NO:168: SEQUENCE CHARACTERISTICS: LENGTH: 16 amino acids 0 TYPE: amino acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: protein g 25 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:168: Met Lys His His His His His His His Ala Ser Val Asn Ala Leu Glu 1 5 10

Claims (1)

  1. 220- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:- 1. An isolated nucleic acid encoding a polypeptide comprising at least one of the biological activities of OPG wherein the nucleic acid hybridises under stringent conditions with the complement of the polypeptide-encoding regions as shown in Figures 2B-2C (SEQ ID NO:120), 9A-9B (SEQ ID NO:122), and 9C-9D (SEQ ID NO: 124); with the proviso that the nucleic acid does not have the sequence as set forth in SEQ ID NO:6 of W096/26217. 2. The nucleic acid of claim 1 which is cDNA, genomic DNA, synthetic DNA or RNA. 3. A polypeptide encoded by the nucleic acid of claim 1. 4. A purified and isolated polypeptide encoded by the nucleic acid of claim 1 with the proviso that the polypeptide does not have the sequence as set forth in SEQ ID NO: 5 of WO96/26217. o 5. The polypeptide of claim 4 which is substantially free of other human proteins. 6. The polypeptide of claim 4 which is characterised by being a product of expression of an S. 15 exogenous DNA sequence. 7. The polypeptide of claim 6, wherein the DNA is a cDNA, genomic DNA or synthetic DNA. 8. The polypeptide of claim 4 which has been modified with a water-soluble polymer. o 9. The polypeptide of claim 8, wherein the water soluble polymer is polyethylene glycol. 20 10. The polypeptide of claim 4 further comprising an Fc region of human IgG1 extending from the carboxy terminus. 11. The nucleic acid of claim 1 including one or more codons preferred for Escherichia coli expression. 12. The nucleic acid of claim 1 having a detectable label attached thereto. 13. A expression vector comprising the nucleic acid of claim 1. 14. A transgenic mammal comprising the expression vector of claim 13. The transgenic mammal of claim 14 which is a rodent. S16. The transgenic mammal of claim 15 which is a mouse. -221- 17. A host cell transformed or transfected with the expression vector of claim 13. 18. The host cell of claim 17 which is a eukaryotic cell. 19. The host cell of claim 18 which is selected from the group consisting of CHO, COS, 293, 3T3, CV-1 and BHK cells. The host cell of claim 17 which is a prokaryotic cell. 21. The host cell of claim 20 which is Escherichia coli. 22. A process for the production of the polypeptide of claim 4 comprising: growing under suitable nutrient conditions host cells transformed or transfected with the nucleic acid of claim 1; and isolating the polypeptide products of the expression of the nuleicacid 23. An antibody or fragment thereof which specifically binds to the polypeptide of claim 3 or claim 4. 24. The antibody of claim 23 which is a monoclonal antibody. 25. A method for detecting the presence of the polypeptide of claim 3 in a biological sample comprising: incubating the sample with the antibody of claim 23 under conditions that allow binding of the antibody to the polypeptide of claim 3; and detecting the bound antibody. 26. A method to assess the ability of a candidate substance to bind to the polypeptide of claim 20 3 or claim 4 comprising; o* incubating the polypeptide of claim 3 or claim 4 with the candidate substance under conditions that allow binding; and measuring the bound substances. 27. A method of regulating the levels of the polypeptide of claim 3 in an animal comprising modifying the animal with the nucleic acid of claim 1. 28. The method of claim 27, wherein the nucleic acid promotes the increase in the tissue level of the polypeptide of claim 3. 29. The method of claim 28, wherein the animal is a human. -222- A pharmaceutical composition comprising a therapeutically effective amount of the polypeptide of any one of claims 4 to 7 in a pharmaceutically acceptable carrier, adjuvant, solubilizer, stabiliser and/or anti-oxidant. 31. A method of treating a bone disorder comprising administering a therapeutically effective amount of the polypeptide of any one of claims 4 to 7 or a pharmaceutical composition of claim 32. The method of claim 31, wherein the bone disorder is excessive bone loss. 33. The method of claim 32, wherein the bone disorder is selected from the group consisting of osteoporosis, Paget's disease of bone, hypercalcemia, hyperparathyroidism, steroid- ndudosteopenia, bone loss duc to rheumatoid arthritis, bne loss due to osteomyelitis, osteolytic metastasis, and periodontal bone loss. 34. The method of claim 31 further comprising administering a therapeutically effective amount of a substance selected from the group consisting of bone morphogenic proteins BMP-1 through BMP-12, TGF-P family members, IL-1 inhibitors, TNFc inhibitors, parathyroid hormone and analogs thereof, parathyroid hormone related protein and analogs thereof, E series prostaglandins, bisphosphonates, and bone-enhancing minerals. 35. The use of a polypeptide according to any one of claims 4 to 7 for the preparation of a medicament for the treatment of a bone disorder. 20 36. An isolated nucleic acid encoding a polypeptide comprising at least on of the biological activities of OPG according to claim 1 and substantially as herein described with reference to any one of the examples. 37. A polypeptide according to claim 3 and substantially as herein described with reference to any one of the examples. 38. A purified and isolated polypeptide according to claim 4 and substantially as herein rT SP defined with reference to any one of the examples. -223- 39. An expression vector according to claim 13 and substantially as herein defined with reference to any one of the examples A transgenic mammal according to claim 14 and substantially as herein defined with reference to any one of the examples. 41. A host cell according to claim 17 and substantially as herein defined with reference to any one of the examples. 42. A process for the production of the polypeptide of claim 4 substantially as herein defined with reference to any one of the examples. 43. An antibody or fragment thereof according to claim 23 and substantially as herein defined 4.6090W0II 1 UIIt IICUefice to an Foey-onl of the examples. 0* 44. A method for detecting the presence of the polypeptide of claim 3 substantially as herein defined with reference to any one of the examples. 45. A method to assess the ability of a candidate substance to bind to the polypeptide of claim 3 or claim 4 substantially as herein defined with reference to any one of the examples. 46. A method of regulating the levels of the polypeptide of claim 3 in an animal substantially as herein defined with reference to any one of the examples. 47. A pharmaceutical composition according to claim 30 and substantially as herein defined with reference to any one of the examples. .00 20 48. A method of treating a bone disorder according to claim 31 and substantially as herein 00 0 defined with reference to any one of the examples. 49. The use of a polypeptide according to claim 35 and substantially as herein defined with reference to any one of the examples. DATED this 28th day of January 2003 AMGEN INC. Attorney: IVAN A. RAJKOVIC R Fellow Institute of Patent Attorneys of Australia of BALDWIN SHELSTON WATERS I
AU65400/99A 1995-12-22 1999-12-22 Osteoprotegerin Ceased AU758672B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU65400/99A AU758672B2 (en) 1995-12-22 1999-12-22 Osteoprotegerin

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/577788 1995-12-22
US08/706945 1996-09-03
AU14686/97A AU710587B2 (en) 1995-12-22 1996-12-20 Osteoprotegerin
AU65400/99A AU758672B2 (en) 1995-12-22 1999-12-22 Osteoprotegerin

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU14686/97A Division AU710587B2 (en) 1995-12-22 1996-12-20 Osteoprotegerin

Publications (2)

Publication Number Publication Date
AU6540099A AU6540099A (en) 2000-03-02
AU758672B2 true AU758672B2 (en) 2003-03-27

Family

ID=3704726

Family Applications (1)

Application Number Title Priority Date Filing Date
AU65400/99A Ceased AU758672B2 (en) 1995-12-22 1999-12-22 Osteoprotegerin

Country Status (1)

Country Link
AU (1) AU758672B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113621060B (en) * 2020-05-07 2023-07-04 浙江瑞硕生物技术有限公司 OPG antibody pair and application thereof

Also Published As

Publication number Publication date
AU6540099A (en) 2000-03-02

Similar Documents

Publication Publication Date Title
AU710587B2 (en) Osteoprotegerin
US6288032B1 (en) Osteoprotegerin
AU2005201799B2 (en) Osteoprotegerin binding proteins and receptors
US20030207827A1 (en) Osteoprotegerin
EP2311955A2 (en) Osteoprotegerin binding proteins and their receptors
WO2001003719A9 (en) Combination therapy for conditions leading to bone loss
US20100298229A1 (en) Osteoprotegerin
US7005413B1 (en) Combination therapy for conditions leading to bone loss
US20050147611A1 (en) Combination therapy for conditions leading to bone loss
AU758672B2 (en) Osteoprotegerin
TWI221482B (en) Osteoprotegerin
JP2004520083A (en) ATP binding cassette transporter-like molecules and uses thereof

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)