AU2061900A - Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily - Google Patents

Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily Download PDF

Info

Publication number
AU2061900A
AU2061900A AU20619/00A AU2061900A AU2061900A AU 2061900 A AU2061900 A AU 2061900A AU 20619/00 A AU20619/00 A AU 20619/00A AU 2061900 A AU2061900 A AU 2061900A AU 2061900 A AU2061900 A AU 2061900A
Authority
AU
Australia
Prior art keywords
rank
seq
ser
amino acid
leu
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.)
Granted
Application number
AU20619/00A
Other versions
AU741504B2 (en
Inventor
Dirk M. Anderson
Laurent J. Galibert
Eugene Maraskovsky
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.)
Immunex Corp
Original Assignee
Immunex Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunex Corp filed Critical Immunex Corp
Priority to AU20619/00A priority Critical patent/AU741504B2/en
Publication of AU2061900A publication Critical patent/AU2061900A/en
Application granted granted Critical
Publication of AU741504B2 publication Critical patent/AU741504B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

AUSTRALIA
Patents Act COMPLETE SPECIFICATION
(ORIGINAL)
Class Int. Class Application Number: Lodged: Complete Specification Lodged: Accepted: Published: Priority Related Art: Name of Applicant: Immunex Corporation Actual Inventor(s): DIRK M ANDERSON, LAURENT J GALIBERT, EUGENE MARASKOVSKY Address for Service: PHILLIPS ORMONDE FITZPATRICK Patent and Trade Mark Attorneys 367 Collins Street Melbourne 3000 AUSTRALIA Invention Title: RECEPTOR ACTIVATOR OF NF-KAPPA B, RECEPTOR IS MEMBER OF TNF RECEPTOR
SUPERFAMILY
S Our Ref: 610431 POF Code: 44735/44735 The following statement is a full description of this invention, including the best method of performing it known to applicant(s): -1-
TITLE
RECEPTOR ACTIVATOR OF NF-KAPPA B, RECEPTOR IS MEMBER OF TNF RECEPTOR SUPERFAMILY The present application is a divisional from Australian patent application number 57184/98 the entire disclosure of which is incorporated herein by reference.
TECHNICAL FIELD OF THE INVENTION The present invention relates generally to the field of cytokine receptors, and more specifically to cytokine receptor/ligand pairs having immunoregulatory activity.
BACKGROUND OF THE INVENTION Efficient functioning of the immune system requires a fine balance between cell proliferation and differentiation and cell death, to ensure that the immune system is capable of reacting to foreign, but not self antigens. Integral to the process of regulating the immune and inflammatory response are various members of the Tumor Necrosis Factor (TNF) Receptor/Nerve Growth Factor Receptor superfamily (Smith et al., Science 248:1019; 1990). This family of receptors includes two different TNF receptors (Type I and Type II; Smith et al., supra; and Schall et al., Cell 61:361, 1990), nerve growth factor receptor (Johnson et al., Cell 47:545, 1986), B cell antigen CD40 (Stamenkovic et al., EMBO J. 8:1403, 1989), CD27 (Camerini et al., J. Immunol. 147:3165, 1991), (Durkop et al., Cell 68:421, 1992), T cell antigen OX40 (Mallett et al., EMBO J. 9:1063, 1990), human Fas antigen (Itoh et al., Cell 66:233, 1991), murine 4-1BB receptor (Kwon 20 et al., Proc. Natl. Acad. Sci. USA 86:1963, 1989) and a receptor referred to as Apoptosis- Inducing Receptor (AIR; USSN 08/720,864, filed October 4, 1996).
is a receptor present on B lymphocytes, epithelial cells and some carcinoma cell lines that interacts with a ligand found on activated T cells, CD40L (USSN 08/249,189, filed May 24, 1994). The interaction of this ligand/receptor pair is essential for both the cellular and humoral immune response. Signal transduction via CD40 is mediated through the association of the cytoplasmic domain of this molecule with members of the TNF receptor-associated factors (TRAFs; Baker and Reddy, Oncogene 12:1, 1996).
It has recently been found that mice that are defective in TRAF3 expression due to a S targeted disruption in the gene encoding TRAF3 appear normal at birth but develop progressive hypoglycemia and depletion of peripheral white cells, and die by about ten days S of age (Xu et al., Immunity 5:407, 1996). The immune responses of chimeric mice reconstituted with TRAF3 4 fetal liver cells resemble those of CD40-deficient mice, although TRAF3-/- B cells appear to be functionally normal.
The critical role of TRAF3 in signal transduction may be in its interaction with one of the other members of the TNF receptor superfamily, for example, CD30 or CD27, which are present on T cells. Alternatively, there may be other, as yet unidentified members of this family of receptors that interact with TRAF3 and play an important role in postnatal development as well as in the development of a competent immune system.
Identifying additional members of the TNF receptor superfamily would provide an additional means of regulating the immune and inflammatory response, as well as potentially providing further insight into post-natal development in mammals.
SUMMARY OF THE INVENTION The present invention provides a novel receptor, referred to as RANK (for receptor activator of NF-KB), that is a member of the TNF receptor superfamily. RANK is a Type I transmembrane protein having 616 amino acid residues that interacts with TRAF3.
Triggering of RANK by over-expression, co-expression of RANK and membrane bound RANK ligand (RANKL), and with addition of soluble RANKL or agonistic antibodies to RANK results in the upregulation of the transcription factor NF-KB, a ubiquitous transcription factor that is most extensively utilized in cells of the immune system.
Soluble forms of the receptor can be prepared and used to interfere with signal transduction through membrane-bound RANK, and hence upregulation of NF-KB; accordingly, pharmaceutical compositions comprising soluble forms of the novel receptor are also provided. Inhibition of NF-KB by RANK antagonists may be useful in ameliorating negative effects of an inflammatory response that result from triggering of RANK, for example in treating toxic shock or sepsis, graft-versus-host reactions, or acute inflammatory reactions. Soluble forms of the receptor will also be useful in vitro to screen for agonists or antagonists of RANK activity.
i* The cytoplasmic domain of RANK will be useful in developing assays for inhibitors of signal transduction, for example, for screening for molecules that inhibit 25 interaction of RANK with TRAF2 or TRAF3. Deleted forms and fusion proteins comprising the novel receptor are also disclosed.
The present invention also identifies a counterstructure, or ligand, for RANK, referred to as RANKL. RANKL is a Type 2 transmembrane protein with an intracellular domain of less than about 50 amino acids, a transmembrane domain and an extracellular domain of from about 240 to 250 amino acids. Similar to other members of the TNF *family to which it belongs, RANKL has a 'spacer' region between the transmembrane domain and the receptor binding domain that is not necessary for receptor binding.
Accordingly, soluble forms of RANKL can comprise the entire extracellular domain or fragments thereof that include the receptor binding region.
These and other aspects of the present invention will become evident upon reference to the following detailed description of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 demonstrates the influence of RANK.Fc and hRANKL on activated T cell growth. Human peripheral blood T cells were cultured as described in Example 12; viable T cell recovery was determined by triplicate trypan blue countings.
Figure 2 illustrates the ability of RANKL to induce human DC cluster formation.
Functionally mature dendritic cells (DC) were generated in vitro from CD34+ bone marrow (BM) progenitors and cultured as described in Example 13. CDla+ DC were cultured in a cytokine cocktail alone (Figure 2A), in cocktail plus CD40L (Figure 2B), RANKL (Figure 2C), or heat inactivated (AH) RANKL (Figure 2D), and then photographed using an inversion microscope.
Figure 3 demonstrates that RANKL enhances DC allo-stimulatory capacity.
Allogeneic T cells were incubated with varying numbers of irradiated DC cultured as described in Example 13. The cultures were pulsed with 3 H]-thymidine and the cells harvested onto glass fiber sheets for counting. Values represent the mean standard deviation (SD) of triplicate cultures.
Figure 4 presents an alignment of human RANK with other TNFR family members in the region of structurally conserved extracellular cysteine-rich pseudorepeats. Predicted disulfide linkages (DS 1-DS3) are indicated. RANK and CD40 contain identical amino acid substitutions CAG) eliminating DS2 in the second pseudorepeat.
Figure 5 presents an alignment of human RANKL with other TNF family members.
DETAILED DESCRIPTION OF THE INVENTION A novel partial cDNA insert with a predicted open reading frame having some S similarity to CD40 was identified in a database containing sequence information from 25 cDNAs generated from human bone marrow-derived dendritic cells The insert was used to hybridize to colony blots generated from a DC cDNA library containing full-length cDNAs. Several colony hybridizations were performed, and two clones (SEQ ID NOs:l and 3) were isolated. SEQ ID NO:5 shows the nucleotide and amino acid sequence of a predicted full-length protein based on alignment of the overlapping sequences of SEQ ID 30 NOs:l and 3.
RANK is a member of the TNF receptor superfamily; it most closely resembles in the extracellular region. Similar to CD40, RANK associates with TRAF2 and TRAF3 (as determined by co-immunoprecipitation assays substantially as described by Rothe et al., Cell 83:1243, 1995). TRAFs are critically important in the regulation of the immune and inflammatory response. Through their association with various members of the TNF receptor superfamily, a signal is transduced to a cell. That signal results in the proliferation, differentiation or apoptosis of the cell, depending on which receptor(s) is/are triggered and which TRAF(s) associate with the receptor(s); different signals can be transduced to a cell via coordination of various signaling events. Thus, a signal transduced through one member of this family may be proliferative, differentiative or apoptotc depending on other signals being transduced to the cell, and/or the state of differentiatio of the cell. Such exquisite regulation of this proliferative/apoptotic pathway is necessary to develop and maintain protection against pathogens; imbalances can result in autoimmune disease.
RANK is expressed on epithelial cells, some B cell lines, and on activated T cells.
However, its expression on activated T cells is late, about four days after activation. This time course of expression coincides with the expression of Fas, a known agent of apoptosis. RANK may act as an anti-apoptotic signal, rescuing cells that express
RANK
from apoptosis as CD40 is known to do. Alternatively, RANK may confirm an apoptotic signal under the appropriate circumstances, again similar to CD40. RANK and its ligand are likely to play an integral role in regulation of the immune and inflammatory response.
Moreover, the post-natal lethality of mice having a targeted disruption of the TRAF3 gene demonstrates the importance of this molecule not only in the immune response but in development. The isolation of RANK, as a protein that associates with TRAF3, and its ligand will allow further definition of this signaling pathway, and development of diagnostic and therapeutic modalities for use in the area of autoimmune and/or inflammatory disease.
DNAs. Proteins and Analogs The present invention provides isolated RANK polypeptides and analogs (or muteins) thereof having an activity exhibited by the native molecule RANK muteins that bind specifically to a RANK ligand expressed on cells or immobilized on a surface or to RANK-specific antibodies; soluble forms thereof that inhibit RANK ligand-induced signaling through RANK). Such proteins are substantially free of contaminating endogenous materials and, optionally, without associated native-pattern glycosylation.
Derivatives of RANK within the scope of the invention also include various structural S forms of the primary proteins which retain biological activity. Due to the presence of ionizable amino and carboxyl groups, for example, a RANK protein may be in the form of acidic or basic salts, or may be in neutral form. Individual amino acid residues may also be modified by oxidation or reduction. The primary amino acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creating amino acid sequence mutants. Covalent derivatives are prepared by linking particular functional groups to amino acid side chains or at the N- or C-termini.
Derivatives of RANK may also be obtained by the action of cross-linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccininide, at cysteine and lysine residues. The inventive proteins may also be covalently bound through reactive side groups to various insoluble substrates, such as cyanogen bromide-activated, bisoxiraneactivated, carbonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolefin surfaces (with or without glutaraldehyde cross-linking). Once bound to a substrate, the proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the proteins or against other proteins which are similar to RANK or RANKL, as well as other proteins that bind RANK or RANKL or homologs thereof.
Soluble forms of RANK are also within the scope of the invention. The nucleotide and predicted amino acid sequence of the RANK is shown in SEQ ID NOs:l through 6.
Computer analysis indicated that the protein has an N-terminal signal peptide; the predicted cleavage site follows residue 24. Those skilled in the art will recognize that the actual cleavage site may be different than that predicted by computer analysis. Thus, the Nterminal amino acid of the cleaved peptide is expected to be within about five amino acids on either side of the predicted, preferred cleavage site following residue 24. Moreover a soluble form beginning with amino acid 33 was prepared; this soluble form bound RANKL. The signal peptide is predicted to be followed by a 188 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 383 amino acid cytoplasmic tail.
Soluble RANK comprises the signal peptide and the extracellular domain (residues 1 to 213 of SEQ ID NO:6) or a fragment thereof. Alternatively, a different signal peptide can be substituted for the native leader, beginning with residue 1 and continuing through a residue selected from the group consisting of amino acids 24 through 33 (inclusive) of SEQ ID NO:6. Moreover, fragments of the extracellular domain will also provide soluble forms of RANK. Fragments can be prepared using known techniques to isolate a desired portion 25 of the extracellular region, and can be prepared, for example, by comparing the extracellular region with those of other members of the TNFR family and selecting forms similar to those prepared for other family members. Alternatively, unique restriction sites or PCR techniques that are known in the art can be used to prepare numerous truncated forms which can be expressed and analyzed for activity.
30 Fragments can be prepared using known techniques to isolate a desired portion of the extracellular region, and can be prepared, for example, by comparing the extracellular region with those of other members of the TNFR family (of which RANK is a member) and selecting forms similar to those prepared for other family members. Alternatively, unique restriction sites or PCR techniques that are known in the art can be used to prepare numerous truncated forms which can be expressed and analyzed for activity.
Other derivatives of the RANK proteins within the scope of this invention include covalent or aggregative conjugates of the proteins or their fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. For example, the conjugated peptide may be a signal (or leader) polypeptide sequence at the N-terminal region of the protein which co-translationally or posttranslationally directs transfer of the protein from its site of synthesis to its site of function inside or outside of the cell membrane or wall the yeast a-factor leader).
Protein fusions can comprise peptides added to facilitate purification or identification of RANK proteins and homologs poly-His). The amino acid sequence of the inventive proteins can also be linked to an identification peptide such as that described by Hopp et al., Bio/Technology 6:1204 (1988). Such a highly antigenic peptide provides an epitope reversibly bound by a specific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein. The sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation in E. coli.
Fusion proteins further comprise the amino acid sequence of a RANK linked to an immunoglobulin Fc region. An exemplary Fc region is a human IgG, having a nucleotide an amino acid sequence set forth in SEQ ID NO:8. Fragments of an Fe region may also be used, as can Fc muteins. For example, certain residues within the hinge region of an Fc region are critical for high affinity binding to FeyRI. Canfield and Morrison Exp. Med.
173:1483; 1991) reported that Leu( 2 34 and Leu(2 35 )were critical to high affinity binding of IgG 3 to FcyRI present on U937 cells. Similar results were obtained by Lund et al. (J.
Immunol. 147:2657, 1991; Molecular ,nmunol. 29:53, 1991). Such mutations, alone or in combination, can be made in an IgG, Fc region to decrease the affinity of IgG, for FcR.
Depending on the portion of the Fc region used, a fusion protein may be expressed as a dimer, through formation of interchain disulfide bonds. If the fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four RANK regions.
In another embodiment, RANK proteins further comprise an oligomerizing peptide such as a leucine zipper domain. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, 1988). Leucine zipper S 30 domain is a term used to refer to a conserved peptide domain present in these (and other) proteins, which is responsible for dimerization of the proteins. The leucine zipper domain (also referred to herein as an oligomerizing, or oligomer-forming, domain) comprises a repetitive heptad repeat, with four or five leucine residues interspersed with other amino acids. Examples of leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-binding protein found in rat liver (C/EBP; Landschulz et al., Science 243:1681, 1989). Two nuclear transforming proteins, fos and jun, also exhibit leucine zipper domains, as does the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240:1759, 1988). The products of the nuclear oncogenesfos andjun comprise leucine zipper domains preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989). The leucine zipper domain is necessary for biological activity (DNA binding) in these proteins.
The fusogenic proteins of several different viruses, including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547,1989; Britton, Nature 353:394, 1991; Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990). The leucine zipper domains in these fusogenic viral proteins are near the transmembrane region of the proteins; it has been suggested that the leucine zipper domains could contribute to the oligomeric structure of the fusogenic proteins. Oligomerization of fusogenic viral proteins is involved in fusion pore formation (Spruce et al, Proc. Natl. Acad. Sci. U.S.A. 88:3523, 1991).
Leucine zipper domains have also been recently reported to play a role in oligomerization of heat-shock transcription factors (Rabindran et al., Science 259:230, 1993).
Leucine zipper domains fold as short, parallel coiled coils. (O'Shea et al., Science 254:539; 1991) The general architecture of the parallel coiled coil has been well characterized, with a "knobs-into-holes" packing as proposed by Crick in 1953 (Acta Crystallogr. 6:689). The dimer formed by a leucine zipper domain is stabilized by the heptad repeat, designated (abcdefg)n according to the notation of McLachlan and Stewart Mol. Biol. 98:293; 1975), in which residues a and d are generally hydrophobic residues, with d being a leucine, which line up on the same face of a helix. Oppositelycharged residues coriimonly occur at positions g and e. Thus, in a parallel coiled coil formed from two helical leucine zipper domains, the "knobs" formed by the hydrophobic side chains of the first helix are packed into the "holes" formed between the side chains of the second helix.
The leucine residues at position d contribute large hydrophobic stabilization energies, and are important for dimer formation (Krystek et al., Int. J. Peptide Res.
38:229, 1991). Lovejoy et al. recently reported the synthesis of a triple-stranded a-helical S bundle in which the helices run up-up-down (Science 259:1288, 1993). Their studies confirmed that hydrophobic stabilization energy provides the main driving force for the formation of coiled coils from helical monomers. These studies also indicate that Selectrostatic interactions contribute to the stoichiometry and geometry of coiled coils.
Several studies have indicated that conservative amino acids may be substituted for individual leucine residues with minimal decrease in the ability to dimerize; multiple changes, however, usually result in loss of this ability (Landschulz et al., Science 243:1681, 1989; Turner and Tjian, Science 243:1689, 1989; Hu et al., Science 250:1400, 1990). van Heekeren et al. reported that a number of different amino residues can be substituted for the leucine residues in the leucine zipper domain of GCN4, and further found that some GCN4 proteins containing two leucine substitutions were weakly active (Nucl. Acids Res. 20:3721, 1992). Mutation of the first and second heptadic leucines of the leucine zipper domain of the measles virus fusion protein (MVF) did not affect syncytium formation (a measure of virally-induced cell fusion); however, mutation of all four leucine residues prevented fusion completely (Buckland et al., J. Gen. Virol. 73:1703 1992). None of the mutations affected the ability of MVF to form a tetramer.
Amino acid substitutions in the a and d residues of a synthetic peptide representing the GCN4 leucine zipper domain have been found to change the oligomerization properties of the leucine zipper domain (Alber, Sixth Symposium of the Protein Society, San Diego, CA). When all residues at position a are changed to isoleucine, the leucine zipper still forms a parallel dimer. When, in addition to this change, all leucine residues at position d are also changed to isoleucine, the resultant peptide spontaneously forms a trimeric parallel' coiled coil in solution. Substituting all amino acids at position d with isoleucine and at position a with leucine results in a peptide that tetramerizes. Peptides containing these substitutions are still referred to as leucine zipper domains.
Also included within the scope of the invention are fragments or derivatives of the intracellular domain of RANK. Such fragments are prepared by any of the hereinmentioned techniques, and include peptides that are identical to the cytoplasmic domain of RANK as shown in SEQ ID NO:5, or of murine RANK as shown in SEQ ID NO: 15, and those that comprise a portion of the cytoplasmic region. All techniques used in preparing soluble forms may also be used in preparing fragments or analogs of the cytoplasmic domain RT-PCR techniques or use of selected restriction enzymes to prepare truncations). DNAs encoding all or a fragment of the intracytoplasmic domain will be :i useful in identifying other proteins that are associated with RANK signalling, for example using the immunoprecipitation techniques described herein, or another technique such as a 25 yeast two-hybrid system (Rothe et al., supra).
The present invention also includes RANK with or without associated native-pattern glycosylation. Proteins expressed in yeast or mammalian expression systems, COS-7 cells, may be similar or slightly different in molecular weight and glycosylation pattern than the native molecules, depending upon the expression system. Expression of DNAs encoding the inventive proteins in bacteria such as E. coli provides non-glycosylated molecules. Functional mutant analogs of RANK protein having inactivated
N-
glycosylation sites can be produced by oligonucleotide synthesis and ligation or by sitespecific mutagenesis techniques. These analog proteins can be produced in a homogeneous, reduced-carbohydrate form in good yield using yeast expression systems.
N-glycosylation sites in eukaryotic proteins are characterized by the amino acid triplet Asnwhere A, is any amino acid except Pro, and Z is Ser or Thr. In this sequence, asparagine provides a side chain amino group for covalent attachment of carbohydrate.
Such a site can be eliminated by substituting another amino acid for Asn or for residue
Z,
deleting Asn or Z, or inserting a non-Z amino acid between Al and Z, or an amino acid other than Asn between Asn and A,.
RANK protein derivatives may also be obtained by mutations of the native RANK or subunits thereof. A RANK mutated protein, as referred to herein, is a polypeptide homologous to a native RANK protein, respectively, but which has an amino acid sequence different from the native protein because of one or a plurality of deletions, insertions or substitutions. The effect of any mutation made in a DNA encoding a mutated peptide may be easily determined by analyzing the ability of the mutated peptide to bind its counterstructure in a specific manner. Moreover, activity of RANK analogs, muteins or derivatives can be determined by any of the assays described herein (for example, inhibition of the ability of RANK to activate transcription).
Analogs of the inventive proteins may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity. For example, cysteine residues can be deleted or replaced with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation. Other approaches to mutagenesis involve modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
When a deletion or insertion strategy is adopted, the potential effect of the deletion or insertion on biological activity should be considered. Subunits of the inventive proteins may be constructed by deleting terminal or intemrnal residues or sequences. Soluble forms of RANK can be readily prepared and tested for their ability to inhibit RANK-induced NFriKB activation. Polypeptides corresponding to the cytoplasmic regions, and fragments thereof (for example, a death domain) can be prepared by similar techniques. Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of RANK to proteins that have similar structures, as well as by performing structural analysis of the inventive RANK proteins.
Generally, substitutions should be made conservatively; the most preferred substitute amino acids are those which do not affect the biological activity of RANK ability of the inventive proteins to bind antibodies to the corresponding native protein in substantially equivalent a manner, the ability to bind the counterstructure in substantially the same manner as the native protein, the ability to transduce a RANK signal, or ability to induce NF-KB activation upon overexpression in transient transfection systems, for example). Examples of conservative substitutions include substitution of amino acids outside of the binding domain(s) (either ligand/receptor or antibody binding areas for the extracellular domain, or regions that interact with other, intracellular proteins for the cytoplasmic domain), and substitution of amino acids that do not alter the secondary and/or tertiary structure of the native protein. Additional examples include substituting one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gin and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known.
Mutations in nucleotide sequences constructed for expression of analog proteins or fragments thereof must, of course, preserve the reading frame phase of the coding sequences and preferably will not create complementary regions that could hybridize to produce secondary mRNA structures such as loops or hairpins which would adversely affect translation of the mRNA.
Not all mutations in the nucleotide sequence which encodes a RANK protein or fragments thereof will be expressed in the final product, for example, nucleotide substitutions may be made to enhance expression, primarily to avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A, incorporated herein by reference), or to provide codons that are more readily translated by the selected host, the well-known E. coli preference codons for E. coli expression.
Although a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants, random mutagenesis may be conducted and the expressed mutated proteins screened for the desired activity. Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required. Exemplary methods of making the alterations set forth above are disclosed by Walder et al. (Gene 42:133, 1986); Bauer et al. (Gene 37:73, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic '30 Engineering: Principles and Methods, Plenum Press, 1981); and U.S. Patent NOs.
4,518,584 and 4,737,462 disclose suitable techniques, and are incorporated by reference S* herein.
Other embodiments of the inventive proteins include RANK polypeptides encoded by DNAs capable of hybridizing to the DNA of SEQ ID NO:6 under moderately stringent '-35 conditions (prewashing solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50C, 5 X SSC, overnight) to the DNA sequences encoding RANK, or more preferably under stringent conditions (for example, hybridization in 6 X SSC at 63'C overnight; washing in 3 X SSC at 55C), and other sequences which are degenerate to those which encode the RANK. In one embodiment, RANK polypeptides are at least about 70% identical in amino acid sequence to the amino acid sequence of native RANK protein as set forth in SEQ ID NO:5. In a preferred embodiment, RANK polypeptides are at least about 80% identical in amino acid sequence to the native form of RANK; most preferred polypeptides are those that are at least about 90% identical to native
RANK.
Percent identity may be determined using a computer program, for example, the GAP computer program described by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). For fragments derived from the RANK protein, the identity is calculated based on that portion of the RANK protein that is present in the fragment The biological activity of RANK analogs or muteins can be determined by testing the ability of the analogs or muteins to inhibit activation of transcription, for example as described in the Examples herein. Alternatively, suitable assays, for example, an enzyme immunoassay or a dot blot, employing an antibody that binds native RANK, or a soluble form of RANKL, can be used to assess the activity of RANK analogs or muteins, as can assays that employ cells expressing RANKL. Suitable assays also include, for example, signal transduction assays and methods that evaluate the ability of the cytoplasmic region of RANK to associate with other intracellular proteins TRAFs 2 and 3) involved in signal transduction will also be useful to assess the activity of RANK analogs or muteins.
Such methods are well known in the art.
Fragments of the RANK nucleotide sequences are also useful. In one embodiment, such fragments comprise at least about 17 consecutive nucleotides, preferably at least about 25 nucleotides, more preferably at least 30 consecutive nucleotides, of the RANK DNA disclosed herein. DNA and RNA complements of such fragments are provided herein, along with both single-stranded and double-stranded forms of the RANK DNA of SEQ ID and those encoding the aforementioned polypeptides. A fragment of RANK DNA S generally comprises at least about 17 nucleotides, preferably from about 17 to about nucleotides. Such nucleic acid fragments (for example, a probe corresponding to the extracellular domain of RANK) are used as a probe or as primers in a polymerase chain reaction (PCR).
The probes also find use in detecting the presence of RANK nucleic acids in in vitro assays and in such procedures as Northern and Southern blots. Cell types expressing RANK can be identified as well. Such procedures are well known, and the skilled artisan can choose a probe of suitable length, depending on the particular intended application. For PCR, 5' and 3' primers corresponding to the termini of a desired RANK DNA sequence are employed to amplify that sequence, using conventional techniques.
Other useful fragments of the RANK nucleic acids are antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target RANK mRNA (sense) or RANK DNA (antisense) sequences.
The ability to create an antisense or a sense oligonucleotide, based upon a cDNA sequence for a given protein is described in, for example, Stein and Cohen, Cancer Res. 48:2659, 1988 and van der Krol et al., BioTechniques 6:958, 1988.
Uses of DNAs, Proteins and Analogs The RANK DNAs, proteins and analogs described herein will have numerous uses, including the preparation of pharmaceutical compositions. For example, soluble forms of RANK will be useful as antagonists of RANK-mediated NF-KB activation, as well as to inhibit transduction of a signal via RANK. RANK compositions (both protein and DNAs) will also be useful in development of both agonistic and antagonistic antibodies to RANK.
The inventive DNAs are useful for the expression of recombinant proteins, and as probes for analysis (either quantitative or qualitative) of the presence or distribution of RANK transcripts.
The inventive proteins will also be useful in preparing kits that are used to detect soluble RANK or RANKL, or monitor RANK-related activity, for example, in patient specimens. RANK proteins will also find uses in monitoring RANK-related activity in other samples or compositions, as is necessary when screening for antagonists or mimetics of this activity (for example, peptides or small molecules that inhibit or mimic, respectively, the interaction). A variety of assay formats are useful in such kits, including (but not limited to) ELISA, dot blot, solid phase binding assays (such as those using a biosensor), rapid format assays and bioassays.
The purified RANK according to the invention will facilitate the discovery of inhibitors of RANK, and thus, inhibitors of an inflammatory response (via inhibition of NF-KB activation). The use of a purified RANK polypeptide in the screening for potential inhibitors is important and can virtually eliminate the possibility of interfering reactions with contaminants. Such a screening assay can utilize either the extracellular domain of RANK, the intracellular domain, or a fragment of either of these polypeptides. Detecting the inhibiting activity of a molecule would typically involve use of a soluble form of RANK derived from the extracellular domain in a screening assay to detect molecules capable of binding RANK and inhibiting binding of, for example, an agonistic antibody or RANKL, or using a polypeptide derived from the intracellular domain in an assay to detect inhibition of the interaction of RANK and other, intracellular proteins involved in signal transduction.
Moreover, in vitro systems can be used to ascertain the ability of molecules to antagonize or agonize RANK activity. Included in such methods are uses of RANK chimeras, for example, a chimera of the RANK intracellular domain and an extracellular domain derived from a protein having a known ligand. The effects on signal transduction of various molecule can then be monitored by utilizing the known ligand to transduce a signal.
In addition, RANK polypeptides can also be used for structure-based design of RANK-inhibitors. Such structure-based design is also known as "rational drug design." The RANK polypeptides can be three-dimensionally analyzed by, for example, X-ray crystallography, nuclear magnetic resonance or homology modeling, all of which are wellknown methods. The use of RANK structural information in molecular modeling software systems to assist in inhibitor design is also encompassed by the invention. Such computerassisted modeling and drug design may utilize information such as chemical conformational analysis, electrostatic potential of the molecules, protein folding, etc. A particular method of the invention comprises analyzing the three dimensional structure of RANK for likely binding sites of substrates, synthesizing a new molecule that incorporates a predictive reactive site, and assaying the new molecule as described above.
Expression of Recombinant RANK The proteins of the present invention are preferably produced by recombinant DNA methods by inserting a DNA sequence encoding RANK protein or an analog thereof into a recombinant expression vector and expressing the DNA sequence in a recombinant expression system under conditions promoting expression. DNA sequences encoding the proteins provided by this invention can be assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene which is capable of being inserted in a recombinant expression vector and expressed in a recombinant transcriptional unit.
Recombinant expression vectors include synthetic or cDNA-derived DNA S fragments encoding RANK, or homologs, muteins or bioequivalent analogs thereof, operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes. Such regulatory elements include a S" transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation, as described in detail below. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants may additionally be incorporated.
DNA regions are operably linked when they are functionally related to each other.
For example, DNA for a signal peptide (secretory leader) is operably linked to DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide; a promoter is operably linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to permit translation. Generally, operably linked means contiguous and, in the case of secretory leaders, contiguous and in reading frame. DNA sequences encoding RANK, or homologs or analogs thereof which are to be expressed in a microorganism will preferably contain no introns that could prematurely terminate transcription of DNA into mRNA.
Useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEMI (Promega Biotec, Madison, WI, USA). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed. E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species (Bolivar et al., Gene 2:95, 1977). pBR322 contains genes for ampicillin and tetracycline resistance and thus provides simple means for identifying transformed cells.
Promoters commonly used in recombinant microbial expression vectors include the P-lactamase (penicillinase) and lactose promoter system (Chang et al., Nature 275:615, 1978; and Goeddel et al., Nature 281:544, 1979), the tryptophan (trp) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, 1980; and EPA 36,776) and tac promoter (Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, p.
412, 1982). A particularly useful bacterial expression system employs the phage X PL promoter and c1857ts thermolabile repressor. Plasmid vectors available from the American Type Culture Collection which incorporate derivatives of the X PL promoter include plasmid pHUB2, resident in E. coli strain JMB9 (ATCC 37092) and pPLc28, resident in E. coli RR1 (ATCC 53082).
Suitable promoter sequences in yeast vectors include the promoters for metallothionein, 3 -phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2073, 1980) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 17:4900, 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6phosphate isomerase, 3 -phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Suitable vectors and promoters S for use in yeast expression are further described in R. Hitzeman et al., EPA 73,657.
Preferred yeast vectors can be assembled using DNA sequences from pBR322 for 35 selection and replication in E. coli (Ampr gene and origin of replication) and yeast DNA sequences including a glucose-repressible ADH2 promoter and a-factor secretion leader.
The ADH2 promoter has been described by Russell et al. Biol. Chem. 258:2674. 1982) and Beier et al. (Nature 300:724, 1982). The yeast or-factor leader, which directs secretion of heterologous proteins, can be inserted between the promoter and the structural gene to be expressed. See, Kurjan et al., Cell 30:933, 1982; and Bitter et al., Proc. Natl. Acad.
Sci. USA 81:5330, 1984. The leader sequence may be modified to contain, near its 3' end, one or more useful restriction sites to facilitate fusion of the leader sequence to foreign genes.
The transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources. For example, commonly used promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence. The early and late promoters are particularly useful because both are obtained easily from the virus as a fragment which also contains the viral origin of replication (Fiers et al., Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 bp sequence extending from the Hind III site toward the BglI site located in the viral origin of replication is included. Further, viral genomic promoter, control and/or signal sequences may be utilized, provided such control sequences are compatible with the host cell chosen.
Exemplary vectors can be constructed as disclosed by Okayama and Berg (Mol. Cell. Biol.
3:280, 1983).
A useful system for stable high level expression of mammalian receptor cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. (Mol. Immunol. 23:935, 1986). A preferred eukaryotic vector for expression of RANK DNA is referred to as pDC406 (McMahan et al., EMBO J. 10:2821, 1991), and includes regulatory sequences derived from SV40, human immunodeficiency virus (HIV), and Epstein-Barr virus (EBV). Other preferred vectors include pDC409 and pDC410, which are derived from pDC406. pDC410 was derived from pDC406 by substituting the EBV origin of replication with sequences encoding the SV40 large T antigen. pDC409 differs from pDC406 in that a Bgl II restriction site outside of the multiple cloning site has been deleted, making the Bgl II site within the multiple cloning site unique.
A useful cell line that allows for episomal replication of expression vectors, such as pDC406 and pDC409, which contain the EBV origin of replication, is CV- I/EBNA (ATCC CRL 10478). The CV-1/EBNA cell line was derived by transfection of the CV-I cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-1) and constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter.
Host Cells Transformed host cells are cells which have been transformed or transfected with expression vectors constructed using recombinant DNA techniques and which contain sequences encoding the proteins of the pres-nt invention. Transformed host cells may express the desired protein (RANK, or homologs or analogs thereof), but host cells transformed for purposes of cloning or amplifying the inventive DNA do not need to express the protein. Expressed proteins will preferably be secreted into the culture supernatant, depending on the DNA selected, but may be deposited in the cell membrane.
Suitable host cells for expression of proteins include prokaryotes, yeast or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram negative or gram positive organisms, for example E. coli or Bacillus spp. Higher eukaryotic cells include established cell lines of mammalian origin as described below.
Cell-free translation systems could also be employed to produce proteins using RNAs derived from the DNA constructs disclosed herein. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, New York, 1985), the relevant disclosure of which is hereby incorporated by reference.
Prokaryotic expression hosts may be used for expression of RANK, or homologs or analogs thereof that do not require extensive proteolytic and disulfide processing.
Prokaryotic expression vectors generally comprise one or more phenotypic selectable markers, for example a gene encoding proteins conferring antibiotic resistance or supplying an autotrophic requirement, and an origin of replication recognized by the host to ensure amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium, and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
Recombinant RANK may also be expressed in yeast hosts, preferably from the Saccharomyces species, such as S. cerevisiae. Yeast of other genera, such as Pichia or Kluyveromyces may also be employed. Yeast vectors will generally contain an origin of 30 replication from the 2t yeast plasmid or an autonomously replicating sequence
(ARS),
S promoter, DNA encoding the protein, sequences for polyadenylation and transcription termination and a selection gene. Preferably, yeast vectors will include an origin of replication and selectable marker permitting transformation of both yeast and E. coli, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae trpi gene, which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, and a promoter derived from a highly expressed yeast gene to induce transcription of a structural sequence downstream. The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
Suitable yeast transformation protocols are known to those of skill in the art; an exemplary technique is described by Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929, 1978, selecting for Trp transformants in a selective medium consisting of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose, 10 u.g/ml adenine and 20 u.g/ml uracil.
Host strains transformed by vectors comprising the ADH2 promoter may be grown for expression in a rich medium consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 gg/ml adenine and 80 g.g/ml uracil. Derepression of the ADH2 promoter occurs upon exhaustion of medium glucose. Crude yeast supernatants are harvested by filtration and held at 4*C prior to further purification.
Various mammalian or insect cell culture systems can be employed to express recombinant protein. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988). Examples of suitable mammalian host cell lines include the COS-7 lines of monkey kidney cells, described by Gluzman (Cell 23:175, 1981), and other cell lines capable of expressing an appropriate vector including, for example, CV-1/EBNA (ATCC CRL 10478), L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines. Mammalian expression vectors may comprise nontranscribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking nontranscribed sequences, and 5' or 3' nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
°25 Purification of Recombinant RANK Purified RANK, and homologs or analogs thereof are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNAs of the present invention, which are then purified from culture media or cell extracts. For example, supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
Following the concentration step, the concentrate can be applied to a suitable purification matrix. For example, a suitable affinity matrix can comprise a counter structure protein or lectin or antibody molecule bound to a suitable support. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Gel filtration chromatography also provides a means of purifying the inventive proteins.
Affinity chromatography is a particularly preferred method of purifying RANK and homologs thereof. For example, a RANK expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography. Moreover, a RANK protein comprising an oligomerizing zipper domain may be purified on a resin comprising an antibody specific to the oligomerizing zipper domain. Monoclonal antibodies against the RANK protein may also be useful in affinity chromatography purification, by utilizing methods that are well-known in the art. A ligand may also be used to prepare an affinity matrix for affinity purification of RANK.
Finally, one or more reversed-phase high performance liquid chromatography
(RP-
HPLC) steps employing hydrophobic RP-HPLC media, silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a RANK composition.
Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
Recombinant protein produced in bacterial culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
Fermentation of yeast which express the inventive protein as a secreted protein greatly simplifies purification. Secreted recombinant protein resulting from a large-scale fermentation can be purified by methods analogous to those disclosed by Urdal et al. (J.
Chromatog. 296:171, 1984). This reference describes two sequential, reversed-phase HPLC steps for purification of recombinant human GM-CSF on a preparative
HPLC
column.
Protein synthesized in recombinant culture is characterized by the presence of cell components, including proteins, in amounts and of a character which depend upon the purification steps taken to recover the inventive protein from the culture. These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic origin and preferably are present in innocuous contaminant quantities, on the order of less than :35 about 1 percent by weight. Further, recombinant cell culture enables the production of the inventive proteins free of other proteins which may be normally associated with the proteins as they are found in nature in the species of origin.
Uses and Administration of RANK Compositions The present invention provides methods of using therapeutic compositions comprising an effective amount of a protein and a suitable diluent and carrier, and methods for regulating an immune or inflammatory response. The use of RANK in conjunction with soluble cytokine receptors or cytokines, or other immunoregulatory molecules is also contemplated.
For therapeutic use, purified protein is administered to a patient, preferably a human, for treatment in a manner appropriate to the indication. Thus, for example, RANK protein compositions administered to regulate immune function can be given by bolus injection, continuous infusion, sustained release from implants, or other suitable technique.
Typically, a therapeutic agent will be administered in the form of a composition comprising purified RANK, in conjunction with physiologically acceptable carriers, excipients or diluents. Such carriers will be nontoxic to recipients at the dosages and concentrations employed.
Ordinarily, the preparation of such protein compositions entails combining the inventive protein with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients. Neutral buffered saline or saline mixed with conspecific serum albumin are exemplary appropriate diluents. Preferably, product is formulated as a lyophilizate using appropriate excipient solutions sucrose) as diluents. Appropriate S dosages can be determined in trials. The amount and frequency of administration will S depend, of course, on such factors as the nature and severity of the indication being treated, the desired response, the condition of the patient, and so forth.
25 Soluble forms of RANK and other RANK antagonists such as antagonistic monoclonal antibodies can be administered for the purpose of inhibiting RANK-induced induction of NF-KB activity. NF-KB is a transcription factor that is utilized extensively by cells of the immune system, and plays a role in the inflammatory response. Thus, inhibitors of RANK signalling will be useful in treating conditions in which signalling through RANK has given rise to negative consequences, for example, toxic or septic shock, or graft-versus-host reactions. They may also be useful in interfering with the role of NF-KB in cellular transformation. Tumor cells are more responsive to radiation when their NF-KB is blocked; thus, soluble RANK (or other antagonists of RANK signalling) will be useful as an adjunct therapy for disease characterized by neoplastic cells that express
RANK.
Throughout the description and claims of this specification, the word "comprise" and variations of the word, such as "comprising" and comprises", is not intended to exclude other additives, components, integers or steps".
"he lollowging examples arc offered by way of illustration, and not by way of limitation. Those skilled in the art wIill recognize that variations of the invention lembod(ied in the examples can be made, esl)pcially in light of the teachings of the various references cited herein, the disclosures of which are incorporated by reference.
EXAMPLE I The example describes the identification and isolation of a DNA encoding a novel member of the TNF receptor superfamily. A partial cDNA insert with a predicted open reading frame having some similarity to CD40 (a cell-surface antigen present on the surface of both normal and neoplastic human B cells that has been shown to play an important role in B-cell proliferation and differentiation; Stamenkovic et al., EMBO J. 8:1403, 1989), was identified in a database containing sequence information from cDNAs generated from human bone marrow-derived dendritic cells The insert was excised from the vector by restriction endonuclease digestion, gel purified, labeled with 32 p, and used to hybridize to colony blots generated from a DC cDNA library containing larger cDNA inserts using high stringency hybridization and washing techniques (hybridization in 5xSSC, formamide at 42°C overnight, washing in 0.5xSSC at 63°C); other suitable high stringency conditions are disclosed in Sambrook et al. in Molecular Cloning: A Laboratory Manual, 2nd ed. (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY; 1989), 9.52-9.55.
Initial experiments yielded a clone referred to as 9D-8A (SEQ ID NO:1); subsequent analysis indicated that this clone contained all but the extreme 5' end of a novel cDNA, with predicted intron sequence at the extreme 5' end (nucleotides 1-92 of SEQ ID NO:1).
S Additional colony hybridizations were performed, and a second clone was isolated. The S" second clone, referred to as 9D-15C (SEQ ID NO:3), contained the 5' end without intron interruption but not the full 3'end. SEQ ID NO:5 shows the nucleotide and amino acid sequence of a predicted full-length protein based on alignment of the overlapping sequences S" of SEQ ID NOs:1 and 3.
The encoded protein was designated RANK, for receptor activator of NF-KB. The cDNA encodes a predicted Type I transmembrane protein having 616 amino acid residues, with a predicted 24 amino acid signal sequence (the computer predicted cleavage site is after Leu24), a 188 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 383 amino acid cytoplasmic tail. The extracellular region of RANK displayed significant amino acid homology (38.5% identity, 52.3% similarity) to CD40. A cloning vector (pBluescriptSK-) containing human RANK sequence, designated pBluescript:huRANK (in E. coli DHIOB), was deposited with the American Type Culture Collection, Rockville, MD (ATCC) on December 20, 1996, under terms of the Budapest Treaty, and given accession number 98285.
EXAMPLE 2 This example describes construction of a RANK DNA construct to express a RANK/Fc fusion protein. A soluble form of RANK fused to the Fc region of human IgG, was constructed in the mammalian expression vector pDC409 (USSN 08/571,579). This expression vector encodes the leader sequence of the Cytomegalovirus (CMV) open reading frame R27080 (SEQ ID NO:9), followed by amino acids 33-213 of RANK, followed by a mutated form of the constant domain of human IgG, that exhibits reduced affinity for Fc receptors (SEQ ID NO:8; for the fusion protein, the Fc portion of the construct consisted of Arg3 through Lys232). An alternative expression vector encompassing amino acids 1-213 of RANK (using the native leader sequence) followed by the IgG, mutein was also prepared. Both expression vectors were found to induce high levels of expression of the RANK/Fc fusion protein in transfected cells.
To obtain RANK/Fc protein, a RANK/Fc expression plasmid is transfected into CV-1/EBNA cells, and supernatants are collected for about one week. The RANK/Fc fusion protein is purified by means well-known in the art for purification of Fc fusion proteins, for example, by protein A sepharose column chromatography according to manufacturer's recommendations Pharmacia, Uppsala, Sweden). SDSpolyacrylamide gel electrophoresis analysis indicted that the purified RANK/Fc protein migrated with a molecular weight of -55kDa in the presence of a reducing agent, and at a molecular weight of -11OkDa in the absence of a reducing agent.
N-terminal amino acid sequencing of the purified protein made using the CMV R27080 leader showed 60% cleavage after Ala20, 20% cleavage after Pro22 and cleavage after Arg28 (which is the Furin cleavage site; amino acid residues are relative to SEQ ID NO:9); N-terminal amino acid analysis of the fusion protein expressed with the native leader showed cleavage predominantly after Gln25 (80% after Gln25 and 20% after Arg23; amino acid residues are relative to SEQ ID NO:6, full-length RANK). Both fusion proteins were able to bind a ligand for RANK is a specific manner they bound to the surface of various cell lines such as a murine thymoma cell line, EL4), indicating that the presence of additional amino acids at the N-terminus of RANK does not interfere with its ability to bind RANKL. Moreover, the construct comprising the CMV leader encoded RANK beginning at amino acid 33; thus, a RANK peptide having an N-terminus at an amino acid between Arg23 and Pro33, inclusive, is expected to be able to bind a ligand for RANK in a specific manner.
Other members of the TNF receptor superfamily have a region of amino acids between the transmembrane domain and the ligand binding domain that is referred to as a 'spacer' region, which is not necessary for ligand binding. In RANK, the amino acids between 196 and 213 are predicted to form such a spacer region. Accordingly, a soluble form of RANK that terminates with an amino acid in this region is expected to retain the ability to bind a ligand for RANK in a specific manner. Preferred C-terminal amino acids for soluble RANK peptides are selected from the group consisting of amino acids 213 and 196 of SEQ ID NO:6, although other amino acids in the spacer region may be utilized as a C-terminus.
EXAMPLE 3 This example illustrates the preparation of monoclonal antibodies against RANK.
Preparations of purified recombinant RANK, for example, or transfected cells expressing high levels of RANK, are employed to generate monoclonal antibodies against RANK using conventional techniques, such as those disclosed in U.S. Patent 4,411,993.
DNA
encoding RANK can also be used as an immunogen, for example, as reviewed by Pardoll and Beckerleg in Immunity 3:165, 1995. Such antibodies are likely to be useful in interfering with RANK-induced signaling (antagonistic or blocking antibodies) or in inducing a signal by cross-linking RANK (agonistic antibodies), as components of diagnostic or research assays for RANK or RANK activity, or in affinity purification of
RANK.
To immunize rodents, RANK immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, MT), and injected in amounts ranging from 10-100 tig subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats. DNA may be given intradermally (Raz et al., Proc. Natl. Acad. Sci. USA 91:9519, 1994) or intamuscularly (Wang et al., Proc. Natl. Acad. Sci. USA 90:4156, 1993); saline has been S found to be a suitable diluent for DNA-based antigens. Ten days to three weeks days later, the immunized animals are boosted with additional immunogen and periodically boosted thereafter on a weekly, biweekly or every third week immunization schedule.
Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS analysis.
S Following detection of an appropriate antibody titer, positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are 30 sacrificed, splenocytes harvested, and fused to a murine myeloma cell line NS 1 or preferably Ag 8.653 [ATCC CRL 1580]). Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
Hybridoma clones thus generated can be screened by ELISA for reactivity with RANK, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in U.S. Patent 4,703,004. A preferred screening technique is the antibody capture technique described by Beckman et al., J. Immunol.
144:4212 (1990). Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations mg/ml) of anti-RANK monoclonal antibody. The resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to RANK protein.
Monoclonal antibodies were generated using RANK/Fc fusion protein as the immunogen. These reagents were screened to confirm reactivity against the RANK protein. Using the methods described herein to monitor the activity of the mAbs, both blocking antibodies that bind RANK and inhibit binding of a ligand to RANK) and non-blocking antibodies that bind RANK and do not inhibit ligand binding) were isolated.
EXAMPLE 4 This example illustrates the induction of NF-KB activity by RANK in 293/EBNA cells (cell line was derived by transfection of the 293 cell line with a gene encoding Epstein- Barr virus nuclear antigen-1 (EBNA-1) that constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter). Activation of NF-KB activity was measured in 293/EBNA cells essentially as described by Yao et al. (Immunity 3:811, 1995). Nuclear extracts were prepared and analyzed for NF-KB activity by a gel retardation assay using a 25 base pair oligonucleotide spanning the NF-KB binding sites. Two million ~cells were seeded into 10 cm dishes two days prior to DNA transfection and cultured in DMEM-F12 media containing 2.5% FBS (fetal bovine serum). DNA transfections were performed as described herein for the IL-8 promoter/reporter assays.
**25 Nuclear extracts were prepared by solubilization of isolated nuclei with 400 mM NaCI (Yao et al., supra). Oligonucleotides containing an NF-KB binding site were annealed and endlabeled with 32 P using T4 DNA polynucleotide kinase. Mobility shift reactions contained 10 gig of nuclear extract, 4 gtg of poly(dI-dC) and 15,000 cpm labeled double-stranded oligonucleotide and incubated at room temperature for 20 minutes.
30 Resulting protein-DNA complexes were resolved on a 6% native polyacrylamide gel in Po* 0.25 X Tris-borate-EDTA buffer.
Overexpression of RANK resulted in induction of NF-KB activity as shown by an appropriate shift in the mobility of the radioactive probe on the gel. Similar results were observed when RANK was triggered by a ligand that binds RANK and transduces a signal to cells expressing the receptor by co-transfecting cells with human RANK and murine RANKL DNA; see Example 7 below), and would be expected to occur when triggering is done with agonistic antibodies.
EXAMPLE This example describes a gene promoter/reporter system based on the human Interleukin-8 (IL-8) promoter used to analyze the activation of gene transcription in vivo.
The induction of human IL-8 gene transcription by the cytokines Interleukin-1 (IL-I) or tumor necrosis factor-alpha (TNF-a) is known to be dependent upon intact NF-KB and NF-IL-6 transcription factor binding sites. Fusion of the cytokine-responsive IL-8 promoter with a cDNA encoding the murine IL-4 receptor (mIL-4R) allows measurement of promoter activation by detection of the heterologous reporter protein (mlL-4R) on the cell surface of transfected cells.
Human kidney epithelial cells (293/EBNA) are transfected (via the DEAE/DEXTRAN method) with plasmids encoding: the reporter/promoter construct (referred to as pIL-8rep), and the cDNA(s) of interest. DNA concentrations are always kept constant by the addition of empty vector DNA. The 293/EBNA cells are plated at a density of 2.5 x 104 cells/ml (3 ml/ well) in a 6 well plate and incubated for two days prior to transfection. Two days after transfection, the mIL-4 receptor is detected by a radioimmunoassay (RIA) described below.
In one such experiment, the 293/EBNA cells were co-transfected with DNA encoding RANK and with DNA encoding RANKL (see Example 7 below). Co-expression of this receptor and its counterstructure by cells results in activation of the signaling process .20 of RANK. For such co-transfection studies, the DNA concentration/well for the DEAE transfection were as follows: 40 ng of pIL-8rep [pBluescriptSK- vector (Stratagene)]; 0.4 ng CD40 (DNA encoding CD40, a control receptor; pCDM8 vector); 0.4 ng RANK (DNA encoding RANK; pDC409 vector), and either 1-50 ng CD40L (DNA encoding the ligand for CD40, which acts as a positive control when co-transfected with CD40 and as a negative control when co-transfected with RANK; in pDC304) or RANKL (DNA encoding a ligand for RANK; in pDC406). Similar experiments can be done using soluble RANKL or agonistic antibodies to RANK to trigger cells transfected with RANK.
For the mlL-4R-specific RIA, a monoclonal antibody reactive with mIL-4R is labeled with '25I via a Chloramine T conjugation method; the resulting specific activity is .30 typically 1.5 x 1016 cpm/nmol. After 48 hours, transfected cells are washed once with media (DMEM/F12 5% FBS). Non-specific binding sites are blocked by the addition of pre-warmed binding media containing 5% non-fat dry milk and incubation at 37°C/5%
CO
2 in a tissue culture incubator for one hour. The blocking media is decanted and binding buffer containing anti-mIL-4R (clone Ml; rat IgG1) is added to the cells and incubated with rocking at room temperature for 1 hour. After incubation of the cells with the radiolabeled antibody, cells are washed extensively with binding buffer (2X) and twice with phosphate-buffered saline (PBS). Cells are lysed in 1 ml of 0.5M NaOH, and total radioactivity is measured with a gamma counter.
Using this assay, 293/EBNA co-transfected with DNAs encoding RANK demonstrated transcriptional activation, as shown by detection of mulL-4R on the cell surface. Overexpression of RANK resulted in transcription of muIL-4R, as did triggering of the RANK by RANKL. Similar results are observed when RANK is triggered by agonistic antibodies.
EXAMPLE 6 This example illustrates the association of RANK with TRAF proteins. Interaction of RANK with cytoplasmic TRAF proteins was demonstrated by co-immunoprecipitation assays essentially as described by Hsu et al. (Cell 84:299; 1996). Briefly, 293/EBNA cells were co-transfected with plasmids that direct the synthesis of RANK and epitope-tagged (FLAG®; SEQ ID NO:7) TRAF2 or TRAF3. Two days after transfection, surface proteins were labeled with biotin-ester, and cells were lysed in a buffer containing 0.5% RANK and proteins associated with this receptor were immunoprecipitated with anti- RANK, washed extensively, resolved by electrophoretic separation on a 6-10% SDS polyacrylamide gel and electrophoretically transferred to a nitrocellulose membrane for Western blotting. The association of TRAF2 and TRAF3 proteins with RANK was visualized by probing the membrane with an antibody that specifically recognizes the FLAG® epitope. TRAFs 2 and 3 did not immunopreciptitate with anti-RANK in the absence of RANK expression.
EXAMPLE 7 This example describes isolation of a ligand for RANK, referred to as RANKL, by direct expression cloning. The ligand was cloned essentially as described in USSN 08/249,189, filed May 24, 1994 (the relevant disclosure of which is incorporated by reference herein), for CD40L. Briefly, a library was prepared from a clone of a mouse thymoma cell line EL-4 (ATCC TIB 39), called EL-40.5, derived by sorting five times with biotinylated CD40/Fc fusion protein in a FACS (fluorescence activated cell sorter). The cDNA library was made using standard methodology; the plasmid DNA was isolated and transfected into sub-confluent CV1-EBNA cells using a DEAE-dextran method.
Transfectants were screened by slide autoradiography for expression of RANKL using a two-step binding method with RANK/Fc fusion protein as prepared in Example 2 followed by radioiodinated goat anti-human IgG antibody.
A clone encoding a protein that specifically bound RANK was isolated and sequenced; the clone was referred to as 11H. An expression vector containing murine RANKL sequence, designated pDC406:muRANK-L (in E. coli DHIOB), was deposited with the American Type Culture Collection, Rockville, MD (ATCC) on December 1996, under terms of the Budapest Treaty, and given accession number 98284. The nucleotide sequence and predicted amino acid sequence of this clone are illustrated in SEQ ID NO: 10. This clone did not contain an initiator methionine; additional, full-length clones were obtained from a 7B9 library (prepared substantially as described in US patent 5,599,905, issued February 4, 1997); the 5' region was found to be identical to that of human RANKL as shown in SEQ ID NO: 12, amino acids 1 through 22, except for substitution of a Gly for a Thr at residue 9.
This ligand is useful for assessing the ability of RANK to bind RANKL by a number of different assays. For example, transfected cells expressing RANKL can be used in a FACS assay (or similar assay) to evaluate the ability of soluble RANK to bind RANKL. Moreover, soluble forms of RANKL can be prepared and used in assays that are known in the art ELISA or BIAcore assays essentially as described in USSN 08/249,189, filed May 24, 1994). RANKL is also useful in affinity purification of RANK, and as a reagent in methods to measure the levels of RANK in a sample. Soluble RANKL is also useful in inducing NF-KB activation and thus protecting cells that express RANK from apoptosis.
EXAMPLE 8 SThis example describes the isolation of a human RANK ligand (RANKL) using a PCR-based technique. Murine RANK ligand-specific oligonucleotide primers were used in PCR reactions using human cell line-derived first strand cDNAs as templates. Primers corresponded to nucleotides 478-497 and to the complement of nucleotides 858-878 of murine RANK ligand (SEQ ID NO:10). An amplified band approximately 400 bp in length from one reaction using the human epidermoid cell line KB (ATCC CCL-17) was gel purified, and its nucleotide sequence determined; the sequence was 85% identical to the corresponding region of murine RANK ligand, confirming that the fragment was from Shuman
RANKL.
To obtain full-length human RANKL cDNAs, two human RANKL-specific oligonucleotides derived from the KB PCR product nucleotide sequence were radiolabeled 0 and used as hybridization probes to screen a human PBL cDNA library prepared in lambda gtlO (Stratagene, La Jolla, CA), substantially as described in US patent 5,599,905, issued February 4, 1997 Several positive hybridizing plaques were identified and purified, their inserts subcloned into pBluescript SK- (Stratagene, La Jolla, CA), and their nucleotide sequence determined. One isolate, PBL3, was found to encode most of the predicted human RANKL, but appeared to be missing approximately 200 bp of 5' coding region. A second isolate, PBL5 was found to encode much of the predicted human RANKL, including the entire 5' end and an additional 200 bp of 5' untranslated sequence.
The 5' end of PBL5 and the 3' end of PBL3 were ligated together to form a full length cDNA encoding human RANKL. The nucleotide and predicted amino acid sequence of the full-length human RANK ligand is shown in SEQ ID NO: 12. Human RANK ligand shares 83% nucleotide and 84% amino acid identity with murine RANK ligand. A plasmid vector containing human RANKL sequence, designated pBluescript:huRANK-L (in E. coli DHIOB), was deposited with the American Type Culture Collection, Rockville, MD (ATCC) on March 11, 1997 under terms of the Budapest Treaty, and given accession number 98354.
Murine and human RANKL are Type 2 transmembrane proteins. Murine RANKL contains a predicted 48 amino acid intracellular domain, 21 amino acid transmembrane domain and 247 amino acid extracellular domain. Human RANKL contains a predicted 47 amino acid intracellular domain, 21 amino acid transmembrane domain and 249 amino acid extracellular domain.
EXAMPLE 9 This example describes the chromosomal mapping of human RANK using PCRbased mapping strategies. Initial human chromosomal assignments were made using RANK and RANKL-specific PCR primers and a BIOS Somatic Cell Hybrid PCRable DNA kit from BIOS Laboratories (New Haven, CT), following the manufacturer's instructions. RANK mapped to human chromosome 18; RANK ligand mapped to human .20 chromosome 13. More detailed mapping was performed using a radiation hybrid mapping panel Genebridge 4 Radiation Hybrid Panel (Research Genetics, Huntsville, AL; described in Walter, MA et al., Nature Genetics 7:22-28, 1994). Data from this analysis was then submitted electronically to the MIT Radiation Hybrid Mapper (URL: http://wwwgenome.wi.mit.edu/cgi-bin/contig/rhmapper.pl) following the instructions contained therein. This analysis yielded specific genetic marker names which, when submitted :electronically to the NCBI Entrez browser (URL: http://www3.ncbi.nlm.nih.gov/htbinpost/Entrez/query?db=c&form--), yielded the specific map locations. RANK mapped to chromosome 18q22.1, and RANKL mapped to chromosome 13q 14.
EXAMPLE This example illustrates the preparation of monoclonal antibodies against RANKL.
Preparations of purified recombinant RANKL, for example, or transfixed cells expressing high levels of RANKL, are employed to generate monoclonal antibodies against RANKL using conventional techniques, such as those disclosed in US Patent 4,411,993. DNA encoding RANKL can also be used as an immunogen, for example, as reviewed by Pardoll and Beckerleg in Immunity 3:165, 1995. Such antibodies are likely to be useful in interfering with RANKL signaling (antagonistic or blocking antibodies), as components of diagnostic or research assays for RANKL or RANKL activity, or in affinity purification of
RANKL.
To immunize rodents, RANKL immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, MT), and injected in amounts ranging from 10-100 g subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats. DNA may be given intradermally (Raz et al., Proc. Natl. Acad. Sci. USA 91:9519, 1994) or intamuscularly (Wang et al., Proc. Natl. Acad. Sci. USA 90:4156, 1993); saline has been found to be a suitable diluent for DNA-based antigens. Ten days to three weeks days later, the immunized animals are boosted with additional immunogen and periodically boosted thereafter on a weekly, biweekly or every third week immunization schedule.
Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS analysis.
Following detection of an appropriate antibody titer, positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are sacrificed, splenocytes harvested, and fused to a murine myeloma cell line NS 1 or preferably 'Ag 8.653 [ATCC CRL 1580]). Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one 0 containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
Hybridoma clones thus generated can be screened by ELISA for reactivity with RANKL, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in US Patent 4,703,004. A preferred screening technique 25 is the antibody capture technique described by Beckman et al., J. Immunol. 144:4212 (1990). Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations mg/ml) of anti-RANK monoclonal S. antibody. The resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to RANKL protein. Using the methods described herein to monitor the activity of the mAbs, both blocking antibodies that bind RANKL and inhibit binding to RANK) and non-blocking antibodies that bind RANKL and do not inhibit binding) are isolated.
EXAMPLE 11 This example demonstrates that RANK expression can be up-regulated. Human peripheral blood T cells were purified by flow cytometry sorting or by negative selection using antibody coated beads, and activated with anti-CD3 (OKT3, Dako) coated plates or phytohemagglutinin in the presence or absence of various cytokines, including Interleukin- 4 (IL-4),Transforming Growth Factor-B (TGF-B) and other commercially available cytokines ILl-a, IL-2, IL-3, IL-6, IL-7, IL-8, IL-10, IL-12, IL-15, IFN-y, TNF- a).
Expression of RANK was evaluated by FACS in a time course experiment for day 2 to day 8, using a mouse monoclonal antibody mAbl44 (prepared as described in Example as shown in the table below. Results are expressed as to referring to the relative increase in intensity of staining with anti-RANK. Double labeling experiments using both anti-RANK and anti-CD8 or anti-CD4 antibodies were also performed.
Table 1: Upregulation of RANK by Cytokines **o *o oooe rr oe oro o Cytokine (concentration) Results: IL-4 (50 ng/ml) TGF-B (5 ng/ml) to IL-4 (50 ng/ml) +TGF-B (5 ng/ml) IL -a IL-2 IL-3 IL-7 IL-8 (lOng/ml) IL-10 IL-12 IFN-y (100U/ml) TNF-a (10ng/ml) Of the cytokines tested, IL-4 and TGF-B increased the level of RANK expression on both CD8+ cytotoxic and CD4+ helper T cells from day 4 to day 8. The combination of IL-4 and TGF-B acted synergistically to upregulate expression of this receptor on activated T cells. This particular combination of cytokines is secreted by suppresser T cells, and is believed to be important in the generation of tolerance (reviewed in Mitchison and Sieper, Z. Rheumatol. 54:141, 1995), implicating the interaction of RANK in regulation of an immune response towards either tolerance or induction of an active immune response.
EXAMPLE 12 This example illustrates the influence of RANK.Fc and hRANKL on activated
T
cell growth. The addition of TGFB to anti-CD3 activated human peripheral blood T lymphocytes induces proliferation arrest and ultimately death of most lymphocytes within the first few days of culture. We tested the effect of RANK:RANKL interactions on TGFBtreated T cells by adding RANK.Fc or soluble human RANKL to T cell cultures.
Human peripheral blood T cells (7 x 10I PBT) were cultured for six days on anti- CD3 (OKT3, 5pg/ml) and anti-Flag (M I, 5pg/ml) coated 24 well plates in the presence of TGFB (Ing/ml) and IL-4 (10ng/ml), with or without recombinant FLAG-tagged soluble hRANKL (lCg/ml) or RANK.Fc (10pg/ml). Viable T cell recovery was determined by triplicate trypan blue countings.
The addition of RANK.Fc significantly reduced the number of viable T cells recovered after six days, whereas soluble RANKL greatly increased the recovery of viable T cells (Figure Thus, endogenous or exogenous RANKL enhances the number of viable T cells generated in the presence of TGFB. TGF8, along with IL-4, has been implicated in immune response regulation when secreted by the TH 3 /regulatory T cell subset. These T cells are believed to mediate bystander suppression of effector T cells.
Accordingly, RANK and its ligand may act in an auto/paracrine fashion to influence T cell tolerance. Moreover, TGFB is known to play a role in the evasion of the immune system effected by certain pathogenic or opportunistic organisms. In addition to playing a role in the development of tolerance, RANK may also play a role in immune system evasion by pathogens.
25 EXAMPLE 13 S This example illustrates the influence of the interaction of RANK on CDIa dendritic cells Functionally mature dendritic cells (DC) were generated in vitro from S CD34+ bone marrow (BM) progenitors. Briefly, human BM cells from normal healthy volunteers were density fractionated using Ficoll medium and CD34+ cells immunoaffinity isolated using an anti-CD34 matrix column (Ceprate, CellPro). The CD34+ BM cells were then cultured in human GM-CSF (20 ng/ml), human IL-4 (20 ng/ml), human TNF-a ng/ml), human CHO-derived Flt3L (FL; 100 ng/ml) in Super McCoy's medium supplemented with 10% fetal calf serum in a fully humidified 37 0 C incubator
CO
2 for 14 days. CDla+, HLA-DR+ DC were then sorted using a FACStar PlusTM, and used for biological evaluation of RANK On human CDla+ DC derived from CD34+ bone marrow cells, only a subset of CDla+ DC expressed RANK at the cell surface as assessed by flow cytometric analysis. However, addition of CD40L to the DC cultures resulted in RANK surface expression on the majority of CDIa+ DC. CD40L has been shown to activate DC by enhancing in vitro cluster formation, inducing DC morphological changes and upregulating HLA-DR, CD54, CD58, CD80 and CD86 expression Addition of RANKL to DC cultures significantly increased the degree of DC aggregation and cluster formation above control cultures, similar to the effects seen with (Figure Sorted human CDla+ DC were cultured in a cytokine cocktail (GM- CSF, IL-4, TNF-a and FL) (upper left panel), in cocktail plus CD40L (lpg/ml) (upper right), in cocktail plus RANKL (lug/ml) (lower left), or in cocktail plus heat inactivated (AH) RANKL (lg/ml) (lower right) in 24-well flat bottomed culture plates in 1 ml culture media for 48-72 hours and then photographed using an inversion microscope. An increase in DC aggregation and cluster formation above control cultures was not evident when heat inactivated RANKL was used, indicating that this effect was dependent on biologically active protein. However, initial phenotypic analysis of adhesion molecule expression indicated that RANKL-induced clustering was not due to increased levels of CD2, CD la, CD54 or CD58.
The addition of RANKL to CDla DC enhanced their allo-stimulatory capacity in a mixed lymphocyte reaction (MLR) by at least 3- to 10-fold, comparable to S DC (Figure Allogeneic T cells (1x10 5 were incubated with varying numbers of irradiated (2000 rad) DC cultured as indicated above for Figure 2 in 96-well round bottomed culture plates in 0.2 ml culture medium for four days. The cultures were pulsed with 0.5 mCi 3 H]-thymidine for eight hours and the cells harvested onto glass fiber sheets for counting on a gas phase P counter. The background counts for either T cells or DC cultured alone were <100 cpm. Values represent the mean SD of triplicate cultures. Heat inactivated RANKL had no effect. DC allo-stimulatory activity was not further enhanced when RANKL and CD40L were used in combination, possibly due to DC functional capacity having reached a maximal level with either cytokine alone. Neither RANKL nor enhanced the in vitro growth of DC over the three day culture period. Unlike CD40L, RANKL did not significantly increase the levels of HLA-DR expression nor the expression of CD80 or CD86.
RANKL can enhance DC cluster formation and functional capacity without modulating known molecules involved in cell adhesion (CD18, CD54), antigen presentation (HLA-DR) or costimulation (CD86), all of which are regulated by CD40/CD40L signaling. The lack of an effect-on the expression of these molecules suggests that RANKL may regulate DC function via an alternate pathway(s) distinct from CD40/CD40L. Given that CD40L regulates RANK surface expression on in vitrogenerated DC and that CD40L is upregulated on activated T cells during DC-T cell interactions, RANK and its ligand may form an important part of the activation cascade that is induced during DC-mediated T cell expansion. Furthermore, culture of DC in RANKL results in decreased levels of CDlb/c expression, and increased levels of CD83. Both of these molecules are similarly modulated during DC maturation by CD40L (Caux et al. J.
Exp. Med. 180:1263; 1994), indicating that RANKL induces DC maturation.
Dendritic cells are referred to as "professional" antigen presenting cells, and have a high capacity for sensitizing MHC-restricted T cells. There is growing interest in using dendritic cells ex vivo as tumor or infectious disease vaccine adjuvants (see, for example, Romani, et al., J. Exp. Med., 180:83, 1994). Therefore, an agent such as RANKL that induces DC maturation and enhances the ability of dendritic cells to stimulate an immune response is likely to be useful in immunotherapy of various diseases.
EXAMPLE 14 This example describes the isolation of the murine homolog of RANK, referred to as muRANK. MuRANK was isolated by a combination of cross-species PCR and colony hybridization. The conservation of Cys residues in the Cys-rich pseudorepeats of the extracellular domains of TNFR superfamily member proteins was exploited to design human RANK-based PCR primers to be used on murine first strand cDNAs from various sources. Both the sense upstream primer and the antisense downstream primer were designed to have their 3' ends terminate within Cys residues.
The upstream sense primer encoded nucleotides 272-295 of SEQ ID NO:5 (region encoding amino acids 79-86); the downstream antisense primer encoded the complement of nucleotides 409-427 (region encoding amino acids 124-130). Standard PCR reactions were set up and run, using these primers and first strand cDNAs from various murine cell line or tissue sources. Thirty reaction cycles of 94°C for 30 seconds, 50 0 C for 30 seconds, and 72 0 C for 20 seconds were run. PCR products were anlyzed by electrophoresis, and specific bands were seen in several samples. The band from one sample was gel purified and DNA sequencing revealed that the sequence between the primers was approximately identical to the corresponding human RANK nucleotide sequence.
30 A plasmid based cDNA library prepared from the murine fetal liver epithelium line FLE18 (one of the cell lines identified as positive in the PCR screen) was screened for fulllength RANK cDNAs using murine RANK-specific oligonucleotide probes derived from the murine RANK sequence determined from sequencing the PCR product. Two cDNAs, one encoding the 5' end and one encoding the 3' end of full-length murine RANK (based on sequence comparison with the full-length human RANK) were recombined to generate a full-length murine RANK cDNA. The nucleotide and amino acid seqeunce of muRANK are shown in SEQ ID Nos:14 and The cDNA encodes a predicted Type 1 transmembrane protein having 625 amino acid residues, with a predicted 30 amino acid signal sequence, a 184 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 390 amino acid cytoplasmic tail. The extracellular region of muRANK displayed significant amino acid homology (69.7% identity, 80.8% similarity) to huRANK. Those of skill in the art will recognize that the actual cleavage site can be different from that predicted by computer; accordingly, the N-terminal of RANK may be from amino acid 25 to amino acid Other members of the TNF receptor superfamily have a region of amino acids between the transmembrane domain and the ligand binding domain that is referred to as a 'spacer' region, which is not necessary for ligand binding. In muRANK, the amino acids between 197 and 214 are predicted to form such a spacer region. Accordingly, a soluble form of RANK that terminates with an amino acid in this region is expected to retain the ability to bind a ligand for RANK in a specific manner. Preferred C-terminal amino acids for soluble RANK peptides are selected from the group consisting of amino acids 214, and 197 of SEQ ID NO: 14, although other amino acids in the spacer region may be utilized as a C-terminus.
EXAMPLE :This example illustrates the preparation of several different soluble forms of RANK .20 and RANKL. Standard techniques of restriction enzyme cutting and ligation, in combination with PCR-based isolation of fragments for which no convenient restriction sites existed, were used. When PCR was utilized, PCR products were sequenced to ascertain whether any mutations had been introduced; no such mutations were found.
In addition to the huRANK/Fc described in Example 2, another RANK/Fc fusion 5 protein was prepared by ligating DNA encoding amino acids 1-213 of SEQ ID NO:6, to DNA encoding amino acids 3-232 of the Fc mutein described previously (SEQ ID NO:8).
A similar construct was prepared for murine RANK, ligating DNA encoding amino acids 1- S 213 of full-length murine RANK (SEQ ID NO: 15) to DNA encoding amino acids 3-232 of the Fc mutein (SEQ ID NO:8).
A soluble, tagged, poly-His version of huRANKL was prepared by ligating DNA encoding the leader peptide from the immunoglobulin kappa chain (SEQ ID NO:16) to DNA encoding a short version of the FLAGTM tag (SEQ ID NO: 17), followed by codons encoding Gly Ser, then a poly-His tag (SEQ ID NO:18), followed by codons encoding Gly Thr Ser, and DNA encoding amino acids 138-317 of SEQ ID NO: 13. A soluble, poly-His tagged version of murine RANKL was prepared by ligating DNA encoding the CMV leader (SEQ ID NO:9) to codons encoding Arg Thr Ser, followed by DNA encoding poly-His (SEQ ID NO: 18) followed by DNA encoding amino acids 119-294 of SEQ ID NO: 11.
A soluble, oligomeric form of huRANKL was prepared by ligating DNA encoding the CMV leader (SEQ ID NO:9) to a codon encoding Asp followed by DNA ending a trimer-former "leucine" zipper (SEQ ID NO:19), then by codons encoding Thr Arg Ser followed by amino acids 138-317 of SEQ ID NO:13.
These and other constructs are prepared by routine experimentation. The various DNAs are then inserted into a suitable expression vector, and expressed. Particularly preferred expression vectors are those which can be used in mammalian cells. For example, pDC409 and pDC304, described herein, are useful for transient expression. For stable transfection, the use of CHO cells is preferred; several useful vectors are described in USSN 08/785,150, now allowed, for example, one of the 2A5-3 ,-derived expression vectors discussed therein.
EXAMPLE 16 This example demonstrates that RANKL expression can be up-regulated on murine T cells. Cells were obtained from mesenteric lymph nodes of C57BU6 mice, and activated with anti-CD3 coated plates, Concanavalin A (ConA) or phorbol myristate acetate in combination with ionomycin (anti-CD3: 500A2; Immunex Corporation, Seattle WA; ConA, PMA, ionomycin, Sigma, St. Louis, MO) substantially as described herein, and cultured from about 2 to 5 days. Expression of RANKL was evaluated in a three color analysis by 20 FACS, using antibodies to the T cell markers CD4, CD8 and CD45RB, and RANK/Fc, prepared as described herein.
o*o RANKL was not expressed on unstimulated murine T cells. T cells stimulated with either anti-CD3, ConA, or PMA/ionomycin, showed differential expression of RANKL: CD4 /CD45RBLo and CD4 /CD45RBHi cells were positive for RANKL, but CD8+ cells 25 were not. RANKL was not observed on B cells, similar to results observed with human cells.
EXAMPLE 17 This example illustrates the effects of murine RANKL on cell proliferation and :30 activation. Various cells or cell lines representative of cells that play a role in an immune response (murine spleen, thymus and lymphnode) were evaluated by culturing them under conditions promoting their viability, in the presence or absence of RANKL. RANKL did not stimulate any of the tested cells to proliferate. One cell line, a macrophage cell line referred to as RAW 264.7 (ATCC accession number TIB 71) exhibited some signs of activation.
RAW cells constitutively produce small amounts of TNF-a. Incubation with either human or murine RANKL enhanced production of TNF-oa by these cells in a dose dependent manner. The results were not due to contamination of RANKL preparations with endotoxin, since boiling RANKL for 10 minutes abrogated TNF-a production, whereas a similar treatment of purified endotoxin (LPS) did not affect the ability of the LPS to stimulate TNF-a production. Despite the fact that RANKL activated the macrophage cell line RAW T64.7 for TNF-a production, neither human RANKL nor murine RANKL stimulated nitric oxide production by these cells.
EXAMPLE 18 This example illustrates the effects of murine RANKL on growth and development of the thymus in fetal mice. Pregnant mice were injected with 1 mg of RANK/Fc or vehicle control protein (murine serum albumin; MSA) on days 13, 16 and 19 of gestation. After birth, the neonates continued to be injected with RANK/Fc intraperitoneally (IP) on a daily basis, beginning at a dose of 1 gg, and doubling the dose about every four days, for a final dosage of 4 gg. Neonates were taken at days 1, 8 and 15 post birth, their thymuses and spleens harvested and examined for size, cellularity and phenotypic composition.
A slight reduction in thymic size at day 1 was observed in the neonates born to the female injected with RANK/Fc; a similar decrease in size was not observed in the control S neonates. At day 8, thymic size and cellularity were reduced by about 50% in the S RANK/Fc-treated animals as compared to MSA treated mice. Phenotypic analysis demonstrated that the relative proportions of different T cell populations in the thymus were S the same in the RANK/Fc mice as the control mice, indicating that the decreased cellularity S was due to a global depression in the number of thymic T cells as opposed to a decrease in a specific population(s). The RANK/Fc-treated neonates were not significantly different from the control neonates at day 15 with respect to either size, cellularity or phenotype of '025 thymic cells. No significant differences were observed in spleen size, cellularity or composition at any of the time points evaluated. The difference in cellularity on day 8 and not on day 15 may suggest that RANK/Fc may assert its effect early in thymic development.
EXAMPLE 19 This example demonstrates that the C-terminal region of the cytoplasmic domain of RANK is important for binding of several different TRAF proteins. RANK contains at least two recognizable PXQX(X)T motifs that are likely TRAF docking sites. Accordingly, the importance of various regions of the cytoplasmic domain of RANK for TRAF binding was evaluated. A RANK/GST fusion protein was prepared substantially as described in Smith and Johnson, Gene 67:31 (1988), and used in the preparation of various truncations as described below.
Comparison of the nucleotide sequence of murine and human RANK indicated that there were several conserved regions that could be important for TRAF binding.
Accordingly, a PCR-based technique was developed to facilitate preparation of various Cterminal truncations that would retain the conserved regions. PCR primers were designed to introduce a stop codon and restriction enzyme site at selected points, yielding the truncations described in Table I below. Sequencing confirmed that no undesired mutations had been introduced in the constructs.
Radio-labeled 35 S-Met, Cys) TRAF proteins were prepared by in vitro translation using a commercially available reticulocyte lysate kit according to manufacturer's instructions (Promega). Truncated GST fusion proteins were purified substantially as described in Smith and Johnson (supra). Briefly, E. coli were transfected with an expression vector encoding a fusion protein, and induced to express the protein. The bacteria were lysed, insoluble material removed, and the fusion protein isolated by precipitation with glutathione-coated beads (Sepahrose 4B, Pharmacia, Uppsala Sweden) The beads were washed, and incubated with various radiolabeled TRAF proteins.
After incubation and wash steps, the fusion protein/TRAF complexes were removed from the beads by boiling in 0.1% SDS B-mercaptoethanol, and loaded onto 12% SDS gels (Novex). The gels were subjected to autoradiography, and the presence or absence of radiolabeled material recorded. The results are shown in Table 2 below.
Table 2: Binding of Various TRAF Proteins to the Cytoplasmic Domain of RANK a
S
*5S*
S
0 C terminal Truncations: E206-S339 E206-Y421 E206-M476 E206-G544 Full length TRAF1 TRAF2 TRAF3 TRAF4 TRAF6 These results indicate that TRAFI, TRAF2, TRAF3, TRAF 5 and TRAF6 bind to the most distal portion of the RANK cytoplasmic domain (between amino-acid G544 and A616). TRAF6 also has a binding site between S339 and Y421. In this experiment, also bound the cytoplasmic domain of RANK.
SEQUENCE LISTING GENERAL INFORMATION: APPLICANT: Immunex Corporation (ii) TITLE OF INVENTION: Receptor Activator of NF-kappaB (iii) NUMBER OF SEQUENCES: 19 (iv) CORRESPONDENCE ADDRESS: ADDRESSEE: Immunex Corporation, Law Department STREET: 51 University Street CITY: Seattle STATE: WA COUNTRY: USA ZIP: 98101 COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: Apple Power Macintosh OPERATING SYSTEM: Apple Operating System 7.5.5 SOFTWARE: Microsoft Word for Power Macintosh 6.0.1 (vi) CURRENT APPLICATION DATA: APPLICATION NUMBER: FILING DATE: 22 DECEMBER 1997
CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: APPLICATION NUMBER: USSN 60/064,671 FILING DATE: 14 OCTOBER 1997 *oo* CLASSIFICATION: a (vii) PRIOR APPLICATION DATA: APPLICATION NUMBER: USSN 08/813,509 FILING DATE: 07 MARCH 1997
CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: APPLICATION NUMBER: USSN 08/772,330 (60/064,671) FILING DATE: 23 DECEMBER 1996
CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION: NAME: Perkins, Patricia Anne REGISTRATION NUMBER: 34,693 REFERENCE/DOCKET NUMBER: 2851-WO (ix) TELECOMMUNICATION INFORMATION: TELEPHONE: (206)587-0430 TELEFAX: (206)233-0644 INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 3115 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: HOMO SAPIENS (vii) IMMEDIATE SOURCE: LIBRARY: BONE-MARROW DERIVED DENDRITIC CELLS CLONE: 9D-8A (ix) FEATURE: NAME/KEY: CDS LOCATION: 93..1868 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1: GCTGCTGCTG CTCTGCGCGC TGCTCGCCCG GCTGCAGTTT TATCCAGAAA GAGCTGTGTG GACTCTCTGC CTGACCTCAG TGTTCTTTTC AG GTG GCT TTG CAG ATC GCT CCT Val Ala Leu Gin Ile Ala Pro 0 9 0 *0*0 99 CCA TGT ACC Pro Cys Thr 10 AGT GAG AAG CAT Ser Glu Lys His
TAT
Tyr 15 GAG CAT CTG GGA Glu His Leu Gly
CGG
Arg TGC TGT AAC Cys Cys Asn AAA TGT Lys Cys 25 GAA CCA GGA AAG Glu Pro Gly Lys
TAC
Tyr ATG TCT TCT AAA Met Ser Ser Lys ACT ACT ACC TCT Thr Thr Thr Ser
GAC
Asp 40 AGT GTA TGT CTG Ser Val Cys Leu TGT GGC CCG GAT Cys Gly Pro Asp
GAA
Glu 50 TAC TTG GAT AGC Tyr Leu Asp Ser
TGG
Trp AAT GAA GAA GAT Asn Glu Glu Asp TGC TTG CTG CAT Cys Leu Leu His
AAA
Lys 65 GTT TGT GAT ACA Val Cys Asp Thr GGC AAG Gly Lys 161 209 257 305 353 401 449 497 GCC CTG GTG Ala Leu Val GCG TGC ACG Ala Cys Thr
GCC
Ala GTG GTC GCC GGC Val Val Ala Gly AGC ACG ACC CCC Ser Thr Thr Pro CGG CGC TGC Arg Arg Cys TGC TGC CGC Cys Cys Arg GCT GGG TAC CAC Ala Gly Tyr His
TGG
Trp 95 AGC CAG GAC TGC Ser Gin Asp Cys
GAG
Glu 100 CGC AAC Arg Asn 105 ACC GAG TGC GCG Thr Glu Cys Ala
CCG
Pro 110 GGC CTG GGC GCC Gly Leu Gly Ala
CAG
Gin 115 CAC CCG TTG CAG His Pro Leu Gin
CTC
Leu 120 AAC AAG GAC ACA Asn Lys Asp Thr TGC AAA CCT TGC Cys Lys Pro Cys GCA GGC TAC TTC Ala Gly Tyr Phe
TCT
Ser 135 GAT GCC TTT TCC TCC ACG GAC AAA TGC Asp Ala Phe Ser Ser Thr Asp Lys Cys 140 CCC TGG ACC AAC Pro Trp Thr Asn TGT ACC Cys Thr 150 545 593 TTC CTT GGA Phe Leu Gly GTT TGC AGT Val Cys Ser 170 AGA GTA GAA CAT Arg Val Glu His
CAT
His 160 GGG ACA GAG AAA TCC GAT GCG Gly Thr Glu Lys Ser Asp Ala 165 TCT TCT CTG CCA Ser Ser Leu Pro
GCT
Ala 175 AGA AAA CCA CCA Arg Lys Pro Pro GAA CCC CAT Glu Pro His GTT TAC Val Tyr 185 TTG CCC GGT TTA Leu Pro Gly Leu
ATA
Ile 190 ATT CTG CTT CTC TTC GCG TCT GTG GCC Ile Leu Leu Leu Phe Ala Ser Val Ala 195 689
CTG
Leu 200 GTG GCT GCC ATC Val Ala Ala Ile TTT GGC GTT TGC Phe Gly Val Cys AGO AAA AAA GGG Arg Lys Lys Giy
AAA
Lys 215 737 785 GCA CTC ACA GCT Ala Leu Thr Ala
AAT
Asn 220 TTG TOG CAC TG Leu Trp His Trp
ATC
Ile 225 AAT GAG OCT TOT Asn Glu Ala Cys GOC CGC Gly Arg 230 CTA AGT GGA Leu Ser Gly ACO GCA AAC Thr Ala Asn 250
GAT
Asp 235 AAG GAG TCC TCA Lys Giu Ser Ser
GOT
Gly 240 GAG AGT TOT GTC Asp Ser Cys Val AGT ACA CAC Ser Thr His 245 TTA CTG CTG Leu Leu Leu 6@@e 6 6O
S
0 *6 6 0
OSOO
*6S9
S
OSSS
S. S 0@
WS
*500 6 0 *5*S U0 @5 *0 6 0
*SSS
*6 S 6S 6@ TTT GOT CAG CAG Phe Gly Gin Gin
GGA
Gly 255 GCA TOT GAA GOT Ala Cys Giu Gly ACT CTG Thr Leu 265 GAG GAG AAG ACA Oiu Giu Lys Thr
TTT
Phe 270 CCA GAA GAT ATG Pro Glu Asp Met
TGC
Cys 275 TAC CCA GAT CAA Tyr Pro Asp Gin
GGT
Gly 280 GOT GTC TGT CAG Gly Val Cys Gin
GGC
Gly 285 ACG TGT OTA OGA Thr Cys Val Gly
GT
Gly 290 GOT CCC TAC GCA Gly Pro Tyr Ala
CAA
Gin 295 833 881 929 977 1025 1073 1121 GOC GAA GAT GCC Gly Giu Asp Ala ATG CTC TCA TTG Met Leu Ser Leu AGC AAG ACC GAG Ser Lys Thr Giu ATA GAG Ile Giu 310 GAA GAC AGC Glu Asp Ser CCC TCC CAG Pro Ser Gin 330
TTC
Phe 315 AGA CAG ATG CCC Arg Gin Met Pro
ACA
Thr 320 GAA GAT GAA TAC Oiu Asp Glu Tyr ATG GAC AGG Met Asp Arg 325 CCT GGA AGC Pro Gly Ser CCC ACA GAC CAG Pro Thr Asp Gin
TTA
Leu 335 CTG TTC CTC ACT Leu Phe Leu Thr AAA TCC Lys Ser 345 ACA CCT CCT TTC Thr Pro Pro Phe GAA CCC CTG GAG Glu Pro Leu Glu
GTO
Val1 355 000 GAG AAT GAC Gly Glu Asn Asp 1169
AOT
Ser 360 TTA AGO CAG TGC TTC ACO 000 ACA CAG Leu Ser Gin Cys Phe Thr Gly Thr Gin 365 ACA OTO GOT TCA Thr Val Gly Ser 1217 AGC TGC AAC TGC ACT GAG CCC CTG TGC AGG ACT Ser Cys Asn Cys Thr 380 Glu Pro Leu Cys Arg 385 Th r GAT TGG ACT Asp Trp Thr CCC ATG Pro Met 1265 1313 TCC TCT GALA Ser Ser Glu CAC TGG GCA His Trp Ala 410
AAC
Asn 395 TAC TTG CAA AAA GAG Tyr Leu Gin Lys Giu 400 GTG GAC AGT GGC CAT TGC CCG Val Asp Ser Gly His Cys Pro 405 GCC AGC CCC AGC Ala Ser Pro Ser
CCC
Pro 415 AAC TGG GCA GAT Asn Trp Ala Asp
GTC
Val 420 TGC ACA GGC Cys Thr Gly 1361 TGC CGG Cys Arg 425 AAC CCT CCT GGG Asn Pro Pro Gly
GAG
Glu 430 GAC TOT GAA CCC Asp Cys Olu Pro
CTC
Leu 435 GTG GGT TCC CCA Val Gly Ser Pro
AAA
Lys 440 COT GGA CCC TTG Arg Gly Pro Leu
CCC
Pro 445 CAG TGC GCC TAT Gin Cys Ala Tyr
GGC
Gly 450 ATG GOC CTT CCC Met Gly Leu Pro
CCT
Pro 1409 1457 1505 GAA GAA GAA GCC Giu Glu Giu Ala
AGC
Ser 460 AGG ACG GAG GCC Arg Thr Glu Ala
AGA
Arg 465 GAC CAG CCC GAG Asp Gin Pro Giu OAT 000 Asp Gly 470 GCT GAT 000 Ala Asp Gly AGC TCC CCT Ser Ser Pro 490 CTC CCA AGC TCA Leu Pro Ser Ser AGG GCA GGT 0CC Arg Ala Oly Ala GGG TCT OGA Gly Ser Oly 485 ACT GGA AAC Thr Gly Asn GOT GGC CAG TCC Gly Gly Gin Ser
CCT
Pro 495 GCA TCT GGA AAT Ala Ser Gly Asn
GTO
Val1 500 AGT AAC Ser Asn 505 TCC ACG TTC ATC Ser Thr Phe Ie
TCC
Ser 510 AGC GGG CAG GTG Ser Gly Gin Val
ATG
Met 515 A-AC TTC AAG GGC Asn Phe Lys Gly
GAC
Asp 520 ATC ATC GTG GTC Ile Ile Val Val
TAC
Tyr 525 GTC AGC CAG ACC Val Ser Gin Thr CAG GAG GGC GCG Gin Glu Gly Ala 1553 1601 1649 1697 1745 1793 1841 OCG OCT GCG GAG Ala Ala Ala .Olu
CCC
Pro 540 ATG OGC CGC CCG Met Gly Arg Pro
GTG
Val1 545 CAG GAG GAG ACC Gin Olu Glu Thr CTG OCO Leu Ala CGC CGA GAC Arg Arg Asp GOC GGC CCC Gly Gly Pro 570
TCC
Ser 555 TTC OCO GOG AAC Phe Ala Oly Asn
OGC
Gly 560 CCG CGC TTC CCG Pro Arg Phe Pro GAC CCG TGC Asp Pro Cys 565 AGO CCG OTO Arg Pro Val GAO 000 CTG CG Giu Oly Leu Arg
GAG
Glu 575 CCG GAG AAG 0CC Pro Olu Lys Ala
TCO
Ser 580 CAG GAO Gin Olu 585 CAA GGC 000 0CC Gin Gly Oly Ala AAG OCT Lys Ala 590 TGA OCGCCCCCCA
TGGCTGGGAG
1888 CCCGAJAGCTC OGAGCCAG CTCGCGAGGO CAGCACCOCA OCCTCTOCCC
CAGCCCCGGC
CACCCAGOOA TCOATCGGTA CAGTCOAOGA AGACCACCCG GCATTCTCTG
CCCACTTTGC
1948 CTTCCAGGAJA ATGGGCTTTT CAGGAAGTGA ATTGATGAGG ACTOTCCCCA TOCCCACGGA 2068
TGCTCAGCAG
AGTAATTTGT
CTATGTTTTC
TCTTTTTTAA
CTCTTTTCCT
TGGTGCGATT
TCCCACCTCA
CCCCGACTCC
CCCCAGCTAA
CCCCCACGCT
ATGGCTTTCC
CCTGGAGATA
GTTCTATATT
ATCTTTTTAA
CTAGGTGGTT
AAAACTCCAA
TGGAGAAAAT
GAATAAAGTT
CCCGCCGCAC
GGCACTATGA
CCCCCATATT
TGTAAAGGTT
TTTTTTTTTC
ATAGCCCGGT
ACCTTCGGAG
CCCCCCCCAG
AGCAGTCCTC
GGCCTGCTTT
CAGTGTGTGT
GTTGCTAAGT
CTCATTTTTC
GTTTGTGTCG
AATTTATCCA
GTTGCTGCAG
GAACAGGACA
GAAATTTTAA
TGGGGCAGAT
CAGCTATTTT
TGTATTCCTT
TTCTCAAAAA
TTTTTTrGGC
GCAGCCTCTA
TAGCTGGGAT
AGACACGGTC
CAGCCTCGGC
ACGTATTTTC
TCATTGTAAA
TGCTAGGAAC
TAAAAGAAAG
TTCCTTAAGC
TGCTGGCAGA
CTTGGCATTC
TGGGGCTCCT
AAAAAAA
GTCTCCCCTG
TATGACTATC
TTCATAACTT
TTCTCCTAAA
AACCTGGCTC
ACTCCTGGGC
CACAGCTGCA
CCACCATGTT
CTCCCAAAGT
TTTTGTGCCC
CACTTTTGGG
ATGTGGTGGG
AAAAAAGGAA
AGAACTAAGC
GGCACTCAGG
TTCTTATTCT
GGAALAGAAAG
CCACTCCTCA
CTGTTCTGTG
TTCTTGATAT
GGTGAGGGTC
TGGCCCAGGC
TCAAGCAATC
GGCCACGCCC
ACCCAGCCTG
ACTGGGATTA
CTGCTCACAG
AAAGGGCTAA
ACTTTCATAT
ACCCGATTTA
TCAGTATGTG
TACTTGGTAA
AGAGGTCTCT
GGCCCGGGAA.
AACTCGCAGC
GGGGGGGGGT
CTTTCCTCCC
TCTTTCTTTT
TAGAGTGCAG
CAAGTGATCC
AGCTTCCTCC
GTCTCAAACT
CAGGCGTGAG
TGTTTTAGAG
ACATGTGAGG
TCTGAAAAAT
TTTCTCCTGA
ACCTTACCCG
GCAAATTTCT
CTGGAAAAGA
GTTCAAGGAA
2128 2188 2248 2308 2368 2428 2488 2548 2608 2668 2728 2788 2848 2908 2968 3028 3088 3115 S. INFORMATION FOR SEQ ID NO:2: SEQUENCE CHARACTERISTICS: LENGTH: 591 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: Val Ala Leu Gin Ile Ala Pro Pro Cys Thr Ser Glu Lys His 1 5 10 His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr 25 Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys 40 Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu 55 Tyr Glu Met Ser Gly Pro Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn 75 Ser Gin Gly Cys Arg 14S Gly Lys Leu Cys Ile 225 Asp Cys Asp GlyC Val 305 Giu i Phe Li Leu G Th Asj Ai Let.
13( Prc Thr Pro Leu Tyr 210 ltsn Ser Glu ~er ~sp 4eu iu r Thr p Cys a. Gin 115 IAla Trp Glu Pro Phe 195 Arg Giu Cys Gly Cys 275 Gly Lys 'J Glu Thr G 3 Val G 355 Pr Git Hi3 Gi Thr Lys Asn 180 Ala Lys Al a Val1 Val1 260 P'yr ?ro Thr r 1lu 40 1 y *Arg .1 Cys 0 Pro Tyr Asn Ser 165 Glu Ser Lys Cys Ser 245 Leu Pro Tyr Giu Met 1Z 325 Pro C Glu A Ar Cy Let Ph( Cy.
i5C Asp Pro Val1 Gly Gly 230 Thr Leu Asp klia Ilie 1l0 \sp ;ly ~sn 9Cys Al sArg Ar -i Gin Le 12 Ser As 135 Thr Ph.
Ala Va.
His Va.
Ala Let 20( Lys Alz 215 Arg Lei.
His Thr Leu Thr Gin Gly 280 Gin Gly 295 Glu Giu Arg Pro Ser Lys Asp Ser 360
C
e 3.Cy Asr 10~ IAsr Ala Leu Cys Tyr 18S Val1 Leu Se r Ala Leu 265 Gly Giu Asp Ser Ser 345 Leu sTh 9, i Th: Ly PhE Glj Sex 170 Leu Ala Thr Gly Asn 250 Glu Val1 Asp Ser Gin 330 Thr Ser r Al~ 0 r Git *AsT SSex Lys 155 *Ser Pro Ala Ala Asp 235 Phe Glu Cys Ala Phe 315 Pro Pro Gin a GlI I Cys Thr -Sex 140 Arg Ser Gly Ile Asn 220 Lys Gly Lys Gin Arg 300 Arg Thr Pro Cys ~Tyr Ala -Val 125 *Thr Val Leu Leu Ile 205 Leu Glu Gin Thr Gly 285 Met Gin b' Asp G Phe S 3 Phe TI Hi~ Pr Cy Asr Glu Pro Ile 190 Phe 'Prp Ser Glm The 270 rhr ,eu let ln ;er 'hr Trp *Gly 3Lys Lys His Ala 175 Ile Gly His Ser Gly 255 Pro Cys Ser I Pro 'I Leu 1 335 dlu P Gly TI Ser Leu Pro Cys His 160 Arg Leu Val1 Trp, Gly 240 kia 3Mu /a 1 ~eu 'hr 4 eu ~ro 'hr Gin Ser 370 Thr Val Gly Ser diu Ser Cys Asn Cys Thr Giu Pro Leu Cys 375 380 Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu 385 390 395 400 Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn 405 410 415 Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro Gly Glu Asp Cys 420 425 430 Glu Pro Leu Val Gly Ser Pro Lys Arg Gly Pro Leu Pro Gin Cys Ala 435 440 445 Tyr Gly Met Gly Leu Pro Pro Glu Glu Glu Ala Ser Arg Thr Glu Ala 450 455 460 Arg Asp Gin Pro Glu Asp Gly Ala Asp Gly Arg Leu Pro Ser Ser Ala 465 470 475 480 Arg Ala Gly Ala Gly Ser Gly Ser Ser Pro Gly Gly Gin Ser Pro Ala 485 490 495 Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly 500 505 510 Gin Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gin 515 520 525 Thr Ser Gin Glu Gly Ala Ala Ala Ala Ala Glu Pro Met Gly Arg Pro 530 535 540 *"Val Gin Glu Glu Thr Leu Ala Arg Arg Asp Ser Phe Ala Gly Asn Gly S 545 550 555 560 Pro Arg Phe Pro Asp Pro Cys Gly Gly Pro Glu Gly Leu Arg Glu Pro 565 570 575 Glu Lys Ala Ser Arg Pro Val Gin Glu Gin Gly Gly Ala Lys Ala 580 585 590 INFORMATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: LENGTH: 1391 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear a a (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: HOMO SAPIENS (vii) IMMEDIATE SOURCE: LIBRARY: BONE-MARROW DERIVED DENDRITIC CELLS CLONE: 9D-15C (ix) FEATURE: NAME/KEY:
CDS
LOCATION: 39..1391 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: CCCCTGAGGC CGCGGCGCCC GCCACCCTCT CCCGCCCC ATG GCC CCG CCC GCC Met Ala Pro Arg Ala CGG CGG CCC CGC CCC CTG TTC GCG CTC CTG CTC CTC TGC GCG Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu Leu Cys Ala CTC CTC Leu Leu CCC CGG CTG Ala Arg Leu AAC CAT TAT Lys His Tyr
CAG
Cln GTG GCT TTC CAC Val Ala Leu Gin
ATC
Ile 30 OCT CCT CCA TGT Ala Pro Pro Cys ACC ACT GAG Thr Ser Clu CAA CCA GGA Clu Pro Oly GAG CAT CTG GGA Clu His Leu Cly
CGG
Arg 45 TGC TOT AAC AAA Cys Cys Asn Lys
TGT
Cys AAC TAC Lys Tyr ATC TCT TCT AAA Met Ser Ser Lys
TGC
Cys 60 ACT ACT ACC TCT Thr Thr Thr Ser
GAC
Asp AGT GTA TCT CTC Ser Val Cys Leu
CCC
Pro TGT GGC CCG GAT Cys Cly Pro Asp r r
CAA
Clu 75 TAC TTG GAT AGC Tyr Leu Asp Ser AAT AA GAA AT Asn Clu Clu Asp
AAA
Lys TGC TTG CTC CAT Cys Leu Leu His CTT TCT CAT ACA Val Cys Asp Thr
GGC
Cly AAG CCC CTC GTC Lys Ala Leu Val CCC GTC Ala Val 100 CTC CCC GGC Val Ala Cly TAC CAC TGG Tyr His Trp 120
AAC
Asn 105 AGC ACC ACC CCC Ser Thr Thr Pro CGC TCC GCC TGC Arg Cys Ala Cys ACG GCT GGG Thr Ala Cly 115 ACC GAG TGC Thr Clu Cys 149 197 245 293 341 389 437 485 533 581 629 AGC CAG GAC TGC Ser Cmn Asp Cys
GAG
Glu 125 TGC TGC CCC CGC Cys Cys Arg Arg
AAC
Asn 130 GCG CCC Ala Pro 135 GGC CTG GGC GCC Cly Leu Cly Ala
CAC
140 140 CAC CCC TTC CAC His Pro Leu Gin
CTC
Leu 145 AAC AAG GAC ACA Asn Lys Asp Thr
GTC
Va1 150 TGC AAA CCT TGC Cys Lys Pro Cys OCA GGC TAC TTC Ala Oly Tyr Phe OAT CCC TTT TCC Asp Ala Phe Ser
TCC
Ser 165 ACG GAC AAA TGC Thr Asp Lys Cys CCC TCG ACC AAC Pro Trp Thr Asn
TGT
Cys 175 ACC TTC CTT GGA Thr Phe Leu Gly AAC AGA Lys Arg 180 CTA CAA CAT Val Clu His
CAT
His 185 GGG ACA GAG AAA Cly Thr Glu Lys
TCC
Ser 190 GAT GCG GTT TGC Asp Ala Val Cys AGT TCT TCT Ser Ser Ser 195 CTG CCA GCT AGA Leu Pro Ala Arg 200 AAA CCA CCA Lys Pro Pro
PLAT
Asn 205 GAA CCC CAT GTT Glu Pro His Val TCT GTG 0CC CTG
TAC
Tyr 210 TTG CCC GGT Leu Pro Gly TTA ATA Leu Ile 215 ATT CTG CTT CTC Ile Leu Leu Leu TTC GCG Phe Ala 220 GTG GCT GCC ATC Val Ala Ala Ile Ser Val Ala Leu 225
ATC
Ile 230 TTT GGC GTT TGC Phe Gly Val Cys
TAT
Tyr 235 AGG AAA AAA Arg Lys Lys GGG AAA GCA Gly Lys Ala 240 CTC ACA GCT Leu Thr Ala TTG TGG CAC TGG Leu Trp His Trp AAT GAG GCT Asn Giu Ala TGT GGC CGC CTA AGT GGA GAT AAG Cys Gly Arg Leu Ser Gly Asp Ly's 255 260 773 821 869 GAG TCC TCA Glu Ser Ser CAG CAG GGA Gin Gin Gly 280
GOT
Gly 265 GAC AGT TGT GTC Asp Ser Cys Val
AGT
Ser 270 ACA CAC ACG GCA Thr His Thr Ala AAC TTT GOT Asn Phe Gly 275 GAG GAG AAG Giu Glu Lys OCA TGT GAA GOT Ala Cys Glu Gly TTA CTG CTG ACT Leu Leu Leu Thr
CTG
Leu 290 ACA TTT Thr Phe 295 CCA GAA GAT ATG Pro Glu Asp Met
TGC
Cys 300 TAC CCA GAT CAA Tyr Pro Asp Gin
GGT
Gly 305 GOT GTC TGT CAG Gly Val Cys Gin
S
65
S
55.5
GGC
Gly 310 ACG TGT GTA GGA Thr Cys Val Gly
GGT
Oly 315 GOT CCC TAC GCA Gly Pro Tyr Ala
CAA
Gin 320 GGC GAA OAT GCC Gly Glu Asp Ala ATG CTC TCA TTG Met Leu Ser Leu AGC AAG ACC GAG Ser Lys Thr Giu GAG GAA GAC AGC Glu Giu Asp Ser TTC AGA Phe Arg 340 CAG ATO CCC Gin Met Pro GAC CAG TTA Asp Gin Leu 360
ACA
Thr 345 GAA GAT GAA TAC Giu Asp Giu Tyr
ATG
Met 350 GAC AGG CCC TCC Asp Arg Pro Ser CAG CCC ACA Gin Pro Thr 355 ACA CCT CCT Thr Pro Pro 917 965 1013 1061 1109 1157 1205 1253 1301 1349 CTG TTC CTC ACT Leu Phe Leu Thr
GAG
Glu 365 CCT GGA AGC AAA Pro Gly Ser Lys
TCC
Ser 370 5**S TTC TCT Phe Ser 375 GAA CCC CTG GAG Giu Pro Leu Giu
GTG
Val1 380 GGG GAG AAT GAC Gly Giu Asn Asp
ACT
Ser 385 TTA AGC CAG TOC Leu Ser Gin Cys
TTC
Phe 390 ACG GGG ACA CAG Thr Giy Thr Gin ACA GTG GGT TCA Thr Val Gly Ser
GAA
Glu 400 AGC TGC MAC TGC Ser Cys Asn Cys
ACT
Thr 405 GAG CCC CTG TGC Glu Pro Leu Cys
AGO
Arg 410 ACT OAT TOG ACT Thr Asp Trp Thr
CCC
Pro 415 ATG TCC TCT GMA Met Ser Ser Glu AAC TAC Asn Tyr 420 TTG CMA AAA Leu Gin Lys GAG GTG Giu Val 425 GAC AGT GGC Asp Ser Giy
CAT
His 430 TGC CCG CAC TG Cys Pro His Trp GCA 0CC AGC Ala Ala Ser 435 CCC AGC CCC AAC TGG GCA GAT GTC TGC ACA GGC TGC CGG AAC Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn 440 445 450 INFORMATION FOR SEQ ID NO:4: SEQUENCE
CHARACTERISTICS:
LENGTH: 451 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: Met Ala Pro Arg Ala Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu 1 1391 f .00.
00* Leu Pro Lys Asp Asn Ala Ala Arg Leu 145 Asp Phe Val1 Val1 Cys Cys Cys Ser Glu Leu Cys Asn 130 Asn Ala Leu Cys Tyr 210 Ala Thr Glu Val1 Glu Val1 Thr 115 Thr Lys Phe Gly Ser 195 Leu Leu Ser Pro Cys Asp Ala 100 Ala Glu Asp Ser Lys 180 Ser Pro Leu Glu Gly Leu Lys Val1 Gly Cys Thr Ser 165 Arg Ser Gly Ala Lys Lys Pro 70 Cys Val1 Tyr Ala Val1 150 Thr Val1 Leu Leu Arg His Tyr 55 Cys Leu Ala His Pro 135 Cys Asp Giu Pro Ile 215 Leu Tyr 40 Met Gly Leu Gly Trp 120 Gly Lys Lys His Ala 200 Ile Gin 25 Glu Ser Pro His Asn 105 Ser Leu Pro Cys His 185 Arg Leu Val1 His Ser Asp Lys 90 Ser Gin Gly Cys Arg 170 Gly Lys Leu Ala Leu Lys Glu 75 Val1 Thr Asp Ala Leu 155 Pro Thr Pro Leu Leu Gly Cys Tyr Cys Thr Cys Gln 140 Ala T rp Glu Pro Phe 220 Gin Arg Thr Leu Asp Pro Glu 125 His Gly Thr Lys A~sn 205 Ala Ile Cys Thr Asp Thr Arg 110 Cys Pro Tyr Asn Ser 190 Glu Ser Ala Cys Thr Ser Gly Arg Cys Leu Phe Cys 175 Asp Pro Val Pro Asn Ser Trp Lys Cys Arg Gln Ser 160 Th r Ala His Ala Leu 225 Val Ala Ala Ile Ile 230 Phe Giy Val Cys Tyr Arg Lys Lys Gly 235 Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg 245 250 255 Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His 260 265 270 Thr Ala Asn Phe Gly Gin Gin Gly Ala Cys Glu Gly Val Leu Leu Leu 275 280 285 Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln 290 295 300 Gly Gly Val Cys Gin Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gin 305 310 315 320 Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu 325 330 335 Glu Asp Ser Phe Arg Gin Met Pro Thr Glu Asp Glu Tyr Met Asp Arg 340 345 350 Pro Ser Gin Pro Thr Asp Gin Leu Leu Phe Leu Thr Glu Pro Gly Ser 355 360 365 Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp 370 375 380 Ser Leu Ser Gin Cys Phe Thr Gly Thr Gin Ser Thr Val Gly Ser Glu 385 390 395 400 SSer Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met 405 410 415 i* Ser Ser Glu Asn Tyr Leu Gin Lys Glu Val Asp Ser Gly His Cys Pro 420 425 430 His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly 435 440 445 Cys Arg Asn 450 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 3136 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: HOMO SAPIENS (vii) IMMEDIATE SOURCE: LIBRARY: BONE-MARROW DERIVED DENDRITIC
CELLS
CLONE: FULL LENGTH RANK (ix) FEATURE: NAI'.E/KEY:
CDS
LOCATION: 39._1886 (xi) SEQUENCE DESCRIPTION: SEQ ID CCGCTGAGGC CGCGGCGCCC GCCAGCCTGT CCCGCGCC ATG GCC CCG CGC GCC Met Ala Pro Arg Ala CGG CGG CGC CGC CCG CTG TTC GCG CTG Arg Arg Arg Arg Pro Leu Phe Ala Leu CTG CTC TGC GCG Leu Leu Cys Ala CTG CTC Leu Leu GCC CGG CTG Ala Arg Leu AAG CAT TAT Lys His Tyr
CAG
Gln GTG GCT TTG CAG Val Ala Leu Gln GCT CCT CCA TGT Ala Pro Pro Cys ACC AGT GAG Thr Ser Glu GAA CCA GGA Glu Pro Gly GAG CAT CTG GGA Glu His Leu Gly
CGG
Arg 45 TGC TGT AAC AAA Cys Cys Asn Lys
TGT
Cys AAG TAC Lys Tyr 55 ATG TCT TCT AAA Met Ser Ser Lys
TGC
Cys 60 ACT ACT ACC TCT Thr Thr Thr Ser
GAC
Asp AGT OTA TGT CTG Ser Val Cys Leu a a TGT GGC CCG GAT Cys Gly Pro Asp
GAA
Glu 75 TAC TTG GAT AGC Tyr Leu Asp Ser
TGG
Trp 80 AAT GAA GAA GAT Asn Glu Glu Asp
AAA
Lys 245 293 341 TGC TTG CTG CAT Cys Leu Leu His
AAA
Lys OTT TGT GAT ACA Val Cys Asp Thr
GGC
Gly 95 AAG GCC CTG GTG Lys Ala Leu Val GCC GTG Ala Val 100 GTC GCC GGC Val Ala Gly TAC CAC TGG Tyr His Trp 120
AAC
Asn 105 AGC ACG ACC CCC Ser Thr Thr Pro CGC TGC GCG TGC Arg Cys Ala Cys ACG GCT GGG Thr Ala Gly 115 ACC GAG TGC Thr Glu Cys AGC CAG GAC TGC Ser Gln Asp Cys
GAG
Glu 125 TOC TGC CGC CGC Cys Cys Arg Arg
AAC
Asn 130 GCG CCG Ala Pro 135 GGC CTG GCC GCC Gly Leu Cly Ala
CAG
Gln 140 CAC CCG TTG CAG His Pro Leu Gin
CTC
Leu 145 AAC AAG GAC ACA Asn Lys Asp Thr
GTG
Val1 150 TGC AAA CCT TGC Cys Lys Pro Cys
CTT
Leu 155 GCA GGC TAC TTC Ala Cly Tyr Phe
TCT
Ser 160 CAT GCC TTT TCC Asp Ala Phe Ser
TCC
Ser 165 485 533 581 ACG GAC AAA TGC Thr Asp Lys Cys CCC TGG ACC AAC Pro Trp Thr Asn ACC TTC CTT GGA Thr Phe Leu Gly AAG AGA Lys Arg 180 GTA GAA CAT Val Glu His
CAT
His 185 GGG ACA GAG AAA Gly Thr Glu Lys
TCC
Ser 190 GAT 0CG OTT TGC Asp Ala Val Cys AGT TCT TCT Ser Ser Ser 195 629 CTC CCA GCT Leu Pro Ala 200 AGA AAA CCA CCA Arg Lys Pro Pro
AAT
Asn 205 GAA CCC CAT GTT Clu Pro His Val
TAC
Tyr 210 TTC CCC GGT Leu Pro Gly TTA ATA Leu Ile 215 ATT CTG CTT CTC Ile Leu Leu Leu
TTC
Phe 220 C TCT GTG GCC Ala Ser Val Ala
CTG
Leu 225 GTG CCT CCC ATC Val Ala Ala Ile
ATC
Ile 230 TTT CCC GTT TGC Phe Gly Val Cys
TAT
Tyr 235 AGG AAA AAA GCC Arg Lys Lys Cly
AAA
Lys 240 CCA CTC ACA CCT Ala Leu Thr Ala 725 773 821 TTC TCC CAC TCC Leu Trp His Trp AAT CAC CCT TCT Asn Ciu Ala Cys
GCC
Cly 255 CCC CTA AGT CCA Arg Leu Ser Cly CAT AAC Asp Lys 260 CAC TCC TCA Clu Ser Ser CAC CAC GGA Cmn Cn Cly 220
CCT
Cly 265 GAC ACT TCT GTC Asp Ser Cys Val ACA CAC ACC CCA Thr His Thr Ala AAC TTT CCT Asn Phe Cly 275 GAG GAC AAC Ciu Clu Lys CCA TGT GAA CCT Ala Cys Clu Cly
CTC
Val1 285; TTA CTG CTC ACT Leu Leu Leu Thr
CTG
Leu 290 ACA TTT Thr Phe 295 CCA CAA CAT ATC Pro Clu Asp Met TAC CCA CAT CAA Tyr Pro Asp Cmn
GGT
Cly 305 CGT CTC TGT CAC Cly Val Cys Cmn
S.
I S
S
S
S S
GCC
Cly 310 ACC TCT CTA CCA Thr Cys Val Cly CCT CCC TAC CCA Cly Pro Tyr Ala
CAA
Cln 320 CCC CAA CAT CC Cly Clu Asp Ala
AGG
Arg 325 965 1013 1061 ATC CTC TCA TTC Met Leu Ser Leu
CTC
Val1 330 ACC AAC, ACC CAC Ser Lys Thr Clu CAG GAA CAC AC Clu Clu Asp Ser TTC ACA Phe Arg 340 CAC ATC CCC Cln Met Pro CAC CAC TTA Asp Gin Leu 360
ACA
Thr 345 CAA CAT CAA TAC Clu Asp Ciu Tyr
ATG
Met 350 CAC ACC CCC TCC Asp Arg Pro Ser CAC CCC ACA Cmn Pro Thr 355 ACA CCT CCT Thr Pro Pro 1109 1157 .c
S
CTC TTC CTC ACT Leu Phe Leu Thr
GAC
Clu 365 CCT GGA ACC AAA Pro Cly Ser Lys
TCC
Ser 370 TTC TCT Phe Ser 375 CAA CCC CTC CAC Clu Pro Leu Clu
CTC
Val 380 CCC CAC AAT CAC Cly Clu Asn Asp
ACT
Ser 385 TTA ACC CAC TGC Leu Ser Cmn Cys
TTC
Phe 390 ACC CCC ACA CAC Thr Cly Thr Cmn ACA CTC CCT TCA Thr Val Cly Ser
CAA
Clu 400 ACC TCC AAC TC Ser Cys Asn Cys
ACT
Thr 405 1205 1253 1301 CAC CCC CTC TC Clu Pro Leu Cys ACT CAT TOG ACT Thr Asp Trp Thr ATG TCC TCT CAA Met Ser Ser Ciu AAC TAC Asn Tyr 420 TTC CAA AAA Leu Cmn Lys
CAC
Clu 425 CTG CAC ACT GC Val Asp Ser Cly TCC CCG CAC TCC Cys Pro His Trp CCA CCC AC Ala Ala Ser 435 1349 CCC AGC CCC AAC TOG GCA GAT GTC TGC ACA CCC TOC COG AAC CCT CCT Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro 1397 GGG GAG Gly Clii 455 CCC CAG Pro Gin 440
GAC
Asp TOT CAA CCC Cys Clii Pro 445
GTG
Val1
CTC
Leu 460
ATG
Met GOT TCC CCA Gly Ser Pro 450
COT
Arg
AAA
Lys 465 OGA CCC TTO Gly Pro Leu TGC GCC TAT Cys Ala Tyr 0CC CTT CCC Gly Leu Pro 470
AGO
Arg
CCT
Pro 480
CG
Cly GAA CAA GAA CC Ciu Glu Giu Ala
AGC
Ser 485 ACG GAG CC Thr Giu Ala
AGA
Arg 490
AGO
Arg CAG CCC GAO Gin Pro Oiu
OAT
Asp 495
TCT
Ser GCT CAT CCC Ala Asp Gly AGO CTC Arg Leu 500 CCA AOC TCA Pro Ser Ser CAG TCC CCT Gin Ser Pro 520 ATC TCC AC Ile Ser Ser
GC
Ala 505
OCA
Ala OCA COT GCC Ala Cly Ala
GOO
Gly 510
ACT
Thr OCA AGC TC Cly Ser Ser TCT OGA AAT Ser Cly Asn
GTO
Val1 525
AAC
Asn OGA AAC ACT Gly Asn Ser
AAC
Asn 530
ATC
Ile CCT GGT GGC Pro Gly Gly 515 TCC ACG TTC Ser Thr Phe ATC OTO GTC Ile Val Val CCC CAG GTG Gly Gin Val 535 TAC GTC Tyr Val
ATO
Met 540
CAG
Gin TTC AAO CGC Phe Lys Gly
GAC
Asp 545
GCG
Ala AOC CAC ACC Ser Gin Thr 550
ATO
Met
TCO
Se r 555
CAG
Gin GAG CCC GCO Glu Gly Aia
CC
Ala 560
C
Ala OCT 0CC GAO Ala Ala Giu
CCC
Pro 565 CCC CCC CCC Oly Arg Pro
OTO
Val1 570
CCC
Pro GAO GAO ACC Clu Ohu Thr
CTO
Leu 575
CCC
Pro COC CCA CAC Arg Arg Asp TCC TTC Ser Phe 580 1445 1493 1541 1589 1637 1685 1733 1781 1829 1877 1926 1986 2046 2106 2166 2226 2286 2346 C COG AA( Ala Oly Asi CTO CCC GAC Leu Arg Ohi CCC AAC GC Ala Lys A1z 615
GGCCTCCCA.
GTACACTCCA
TTTCAGGAAG
CACTGCOOCA
TOACACCTAT
ATTTOTATTC
GTTTTCTCAA
GCC
Cly 585
CCO
Pro CCC TTC CCC Arg Phe Pro
GAC
Asp 590
AGO
Arg TGC GOC GCC Cys Oly Gly CCC GAG CCC Pro Ciii Gly 595 Gin Oly Cly GAO AAO CCC TCC Giu Lys Ala Ser CCO OTO CAG Pro Val Gin
GAG
610 TOAOCGCCCC CCATGOCTCG GAGCCCGAAG
CTCGOAOCCA
GOCAOCACC
GCAAOACCAC
TGM-TTGATG
CATGTCTCCC
TTTTATGACT
CTTTTCATAA
AAATTCTCCT
OCAOCCTCTG
CCGGCATTCT
AGGACTGTCC
CTGCCACTCC
ATCCTGTTCT
CTTTTCTTGA
AAAGGTGAGG
CCCCACCCCC
CTGCCCACTT
CCATCCCCAC
TCAAACTCC
OTOOCOOCCO
TATCTTTCCT
GTCTCTTTCT
GCCACCCAC
TCCCTTCCAC
GGATOCTCAC
AGCAGTAATT
OCTCTATCTT
CCCTCTTTTT
TTTCTCTTTT
COATCCATCO
GAAATGGOCT
CAOCCCGCCG
TOTGCCACTA
TTCCCCCCAT
TAATGTAAAG
CCTTTTTTTT
TTCTTTTTTT
GGTGCAGCCT
GAGTAGCTGG
CAGAGACACG
CTCCAGCCTC
TTTACGTATT
TGTTCATTGT
AGTTGCTAGG
TTCTAAAAGA
TCGTTCCTTA
CCATGCTGGC
CAGCTTGGCA
GGCAACCTGG
CTAACTCCTG
GATCACAGCT
GTCCCACCAT
GGCCTCCCAA
TTCTTTTGTG
AAACACTTTT
AACATGTGGT
AAGAAAAAAG
AGCAGAACTA
AGAGGCACTC
TTCTTCTTAT
CTCTGGCCCA
GGCTCAAGCA
GCAGGCCACG
GTTACCCAGC
AGTACTGGGA
CCCCTGCTCA
GGGAAAGGGC
GGGACTTTCA
GAAACCCGAT
AGCTCAGTAT
AGGTACTTGG
TCTAGAGGTC
GGCTAGAGTG
ATCCAAGTGA
CCCAGCTTCC
CTGGTCTCAA
TTACAGGCGT
CAGTGTTTTA
TAAACATGTG
TATTCTGAAA
TTATTTCTCC
GTGACCTTAC
TAAGCAAkATT
TCTCTGGAAA
CAGTGGTGCG
TCCTCCCACC
TCCCCCCGAC
ACTCCCCAGC
GAGCCCCCAC
GAGATGGCTT
AGGCCTGGAG
AATGTTCTAT
TGAATCTTTT
CCGCTAGGTG
TCTAAAACTC
AGATGGAGAA
ATTATAGCCC
TCAACCTTCG
TCCCCCCCCC
TAAAGCAGTC
GCTGGCCTGC
TCCCAGTGTG
ATAGTTGCTA
ATTCTCATTT
TAAGTTTGTG
GTTAATTTAT
CAAGTTGCTG
AATGAACAGG
2406 2466 2526 2586 2646 2706 2766 2826 2886 2946 3006 3066 ACATGGGGCT CCTGGAAAGA AAGGGCCCGG GAAGTTCAAG GAAGAATAAA GTTGAAATTT 3126 TAAAAAAAAA 3136 INFORMATION FOR SEQ ID NO:6: SEQUENCE CHARACTERISTICS: LENGTH: 616 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: Met Ala Pro Arg Ala Arg Arg Arg Arg Pro 1 5 10 Leu Cys Ala Leu Leu Ala Arg Leu Gin Val 25 Pro Cys Thr Ser Giu Lys His Tyr Glu His 40 Lys Cys Giu Pro Gly Lys Tyr Met Ser Ser 55 Asp Ser Val Cys Leu Pro Cys Gly Pro Asp 70 Asn Giu Glu Asp Lys Cys Leu Leu His Lys 90 Ala Leu Val Ala Val Val Ala Gly Asn Ser Leu Phe Ala Leu Leu Lou Ala Leu Gin Leu Gly Arg Lys Cys Thr Ile Ala Pro Cys Cys Agn Thr Thr Ser Tyr Leu Asp Ser Trp Cys Asp Thr Gly Lys Arg Arg Cys 110 Thr Thr Pro 100 105 Ala Cys Thr Ala Gly TPyr His Trp Ser Gin Asp Cys Glu Cys Cys Arg 115 120 125 Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gin His Pro Leu Gin 130 135 140 Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser 145 150 155 160 Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr 165 170 175 Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala 180 185 190 Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His 195 200 205 Val Tyr Leu Pro Gly Leu Ilie Ile Leu Leu Leu Phe Ala Ser Val Ala 210 215 220 Leu Val Al~a Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys 225 230 235 240 Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg 245 250 255 Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His 260 265 270 Thr Ala Asn Phe Gly Gin Gin Gly Ala Cys Glu Gly Val Leu Leu Leu *275 280 285 Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gin .290 295 300 Gly Gly Val Cys Gin Gly Thr Cys Val Giy Gly Gly Pro Tyr Ala Gin 305 310 315 320 Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Giu 325 330 335 Glu Asp Ser Phe Arg Gin Met Pro Thr Glu Asp Glu Tyr Met Asp Arg 340 345 350 *.Pro Ser Gin Pro Thr Asp Gin Leu Leu Phe Leu Thr Giu Pro Gly Ser *355 360 365 Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp 370 375 380 Ser Leu Ser Gin Cys Phe Thr Gly Thr Gin Ser Thr Val Gly Ser Glu 385 390 395 400 Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met 405 410 415 Ser Ser Glu Asn Tyr Leu Gin Lys Glu Val Asp Ser Gly His Cys Pro 420 425 430 His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly 435 Cys Lys 465 Glu Ala Ser Ser Asp 545 Ala Arg Gly Gin Arg 450 Arg Glu Asp Ser Asn 530 Ile Ala Arg Gly Glu 610 Asn Pro Gly Pro Glu Ala Gly Arg 500 Pro Gly 515 Ser Thr Ile Val Ala Glu Asp Ser 580 Pro Glu 595.
Gin Gly Pro Leu Ser 485 Leu Gly Phe Val Pro 565 Phe Gly Gly Gly Pro 470 Arg Pro Gin Ile Tyr 550 Met Ala Leu Ala Glu 455 Gin Thr Ser Ser Ser 535 Val Gly Gly Arg Lys 615 440 Asp Cys Glu Ser Pro 520 Ser Ser Arg Asn Glu 600 Ala Cys Ala Ala Ala 505 Ala Gly Gin Pro Gly 585 Pro Glu Tyr Arg 490 Arg Ser Gin Thr Val 570 Pro Glu Pro Gly 475 Asp Ala Gly Val Ser 555 Gin Arg Lys 445 Leu 460 Met Gin Gly Asn Met 540 Gin Glu Phe Ala Val Gly Pro Ala Val 525 Asn Glu Glu Pro Ser 605 Gly Leu Glu Gly 510 Thr Phe Gly Thr Asp 590 Arg Ser Pro Asp 495 Ser Gly Lys Ala Leu 575 Pro Pro Pro Pro 480 Gly Gly Asn Gly Ala 560 Ala Cys Val INFORMATION FOR SEQ ID NO:7: SEQUENCE CHARACTERISTICS: LENGTH: 8 amino acids TYPE: amino acid STRANDEDNESS: not relevant TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (vii) IMMEDIATE SOURCE: CLONE: FLAG® peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: Asp Tyr Lys Asp Asp Asp Asp Lys 1 INFORMATION FOR SEQ ID NO:8: SEQUENCE CHARACTERISTICS: LENGTH: 232 amino acids TYPE: amino acid STRANDEDNESS: not relevant TOPOLOGY: linear (ii) MOLECULE TYPE: protein (vi) ORIGINAL SOURCE: ORGANISM: Human (vii) IMMEDIATE
SOURCE:
CLONE: IgG1 Fc mutein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: Glu Pro Arg Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala 1 5 10 Pro Glu Ala Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro 25 Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val 40 Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val 55 Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin 70 75 Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin 90 Asp Trp Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala 00 105 110 Leu Pro Ala Pro Met Gin Lys Thr Ile Ser Lys Ala Lys Gly Gin Pro 115 120 125 125 Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr 130 135 140 Lys Asn Gin Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg 145 150 155 160 His Ile Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn Tyr 165 170 175 Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr 180 185 190 Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly Asn Val Phe 195 200 205 Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gin Lys 210 215 220 Ser Leu Ser Leu Ser Pro Gly Lys 225 230 INFORMATION FOR SEQ ID NO:9: SEQUENCE CHARACTERISTICS: LENGTH: 31 amino acids TYPE: amino acid STRANDEDNESS: not relevant TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: CMV (R2780 Leader) (ix) FEATURE: OTHER INFORMATION: Metl-Arg28 is the actual leader peptide; Arg29 strengthens the furin cleavage site; nucleotides encoding Thr30 and Ser31 add a Spel site.
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: Met Ala Arg Arg Leu Trp Ile Leu Ser Leu Leu Ala Val Thr Leu Thr 1 5 10 Val Ala Leu Ala Ala Pro Ser Gin Lys Ser Lys Arg Arg Thr Ser 25 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 1630 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: Mus musculus (vii) IMMEDIATE SOURCE:
LIBRARY:
CLONE: RANKL (ix) FEATURE: NAME/KEY: CDS LOCATION: 3..884 (xi) SEQUENCE DESCRIPTION: SEQ ID CC GGC GTC CCA CAC GAG GGT CCG CTG CAC CCC GCG CCT TCT GCA CCG 47 Gly Val Pro His Glu Gly Pro Leu His Pro Ala Pro Ser Ala Pro 1 5 10 OCT CCG GC CCG CCA CCC Ala Pro Ala Pro Pro Pro GCC CCC TCC COC TCC ATG TTC Ala Ala Ser Arg 25 Ser Met Phe AGC ATC OCT Ser Ile Ala CTG GGG CTG Leu Gly Leu TAC TTT CGA Tyr Phe Arg
GGA
Gly CTG GGC CAG GTG Leu Gly Gin Val GTC TOC Val Cys 40 CTG GCC CTC Leu Ala Leu CTO TTC CTG Leu Phe Leu GAC AGC ACT Asp Ser Thr GCG CAG ATG GAT Ala Gin Met Asp
CCT
Pro 55 AAC AGA ATA TCA Asn Arg Ile Ser
GAA
Giu CAC TGC His Cys 6S TTT TAT AGA ATC Phe Tyr Arg Ile
CTG
Leu 70 AGA CTC CAT GAA Arg Leu His Giu
AAC
Asn GCA OAT TTG CAC Ala Asp Leu Gin
GAC
Asp TCO ACT CTO GAG Ser Thr Leu Glu GAA GAC ACA CTA Glu Asp Thr Leu
CCT
Pro GAC TCC TGC AGO Asp Ser Cys Arg
AGO
Arg 239 287 335 ATO AAA CAA 0CC Met Lys Gin Ala
TTT
Phe 100 CAG 000 0CC OTG Gin Gly Ala Val
CAG
Gin 105 AAO GAA CTO CAA CAC ATT Lys Giu Leu Gin His Ile 110 GTO 000 CCA Val Gly Pro TOO TTG OAT Trp Leu Asp 130
CAG
Gin 115 COC TTC TCA GGA Arg Phe Ser Gly
OCT
Ala 120 CCA OCT ATG ATG Pro Ala Met Met OAA GOC TCA Giu Gly Ser 125 CCA TTT GCA Pro Phe Ala OTO 0CC CAG CGA Vai Ala Gin Arg
GGC
Gly 135 AAO CCT GAO 0CC Lys Pro Giu Ala CAC CTC His Leu 145 ACC ATC AAT OCT Thr Ile Asn Ala
CC
Ala 150 AGC ATC CCA TCO Ser Ile Pro Ser
GT
Gly 155 TCC CAT AAA GTC Ser His Lys Val 383 431 479 527 575
ACT
Th r 160 CTG TCC TCT TG Leu Ser Ser Trp
TAC
Tyr 165 CAC OAT CGA C His Asp Arg Gly 0CC AAG ATC TCT Ala Lys Ile Ser
AAC
Asn 175 ATO ACO TTA AOC Met Thr Leu Ser OGA AAA CTA AGO Gly Lys Leu Arg
OTT
Val1 185 AAC CAA OAT C Asn Gin Asp Gly TTC TAT Phe Tyr 190 TAC CTG TAC Tyr Leu Tyr GTA CCT ACA Val Pro Thr 210 0CC Ala 195 AAC ATT TGC TTT Asn Ile Cys Phe
CG
Arg 200 CAT CAT GAA ACA His His Glu Thr TCG GGA AGC Ser Gly Ser 205 AAA ACC AOC Lys Thr Ser GAC TAT CTT CAG Asp Tyr Leu Gin
CTG
Leu 215 ATG GTO TAT GTC Met Val Tyr Val ATC AAA Ile Lys 225 ATC CCA ACT TCT Ile Pro Ser Ser
CAT
His 230 AAC CTC ATO AAA Asn Leu Met Lys
GGA
Gly 235 000 AOC ACO AAA Gly Ser Thr Lys 719
AAC
Asn 240 TGG TCG 0CC AAT TCT OAA TTC CAC TTT Trp Ser Gly Asn Ser Giu Phe His Phe 245 TCC ATA AAT OTT Ser Ile Asn Val
GG
Gly 255 767 GGA TTT TTG AAG CTC GGA GCT GGT GAA GAA ATT AGG ATT CAG GTG TC Gly Phe Phe Lys Leu Arg Ala Gly Glu Glu Ile Ser Ile Gin Val Ser 260 265 270 AAC CCT TCC CTG CTG GAT GGG GAT CA.A GAT GGG AGG TAG TTT GGG GGT Asn Pro Ser Leu Leu Asp Pro Asp Gin Asp Ala Thr Tyr Phe Gly Ala 275 280 285 TTC AAA GTT CAG GAG ATA GAG TGAGACTGAT TTGGTGGAAG ATTAGCATGG Phe Lys Val Gin Asp Ile Asp 290
ATGTCGTAGA
AGTAAGAGAC
GTTGTGTATA
AGAATTTTGT
GAAAAAGTTA
TGTGCCACTG
TGAAGGGTTA
TTTGTAATGA
GTGTAATGTT
AATATTTAAA
AGTGGTGGAG
CTGGTGAGGA
TGTTTGGAAA
ATGGGGGAG
TGTAAAGTC
AATGATTTGG
CACGTGAGGT
AGAAGCTTGA
AGTTCTTTTG
GGAGAGAAAA
TTGTGTGGAA
AATGTCTCAG
TTTGTAATTG
GATGTAGTTT
GTTGTTAAA.A
GTGTATGAAA
ATAGGTGATG
TAGAATTGAA
ATGGAAGGGG
AATTAAGAGG
AATTGTTACA
ATATATGTAT
AGTTTTGTAA
TGTTGAGATA
GGGTGAAGGT
ATTTCTTTAT
AATGGATGAT
GTGACAGGGG
TTAGATTCAT
CCAGATTGGG
GTCACAGTCT
ATGCGATGTG
TTGC!GCTGGG
TTTTATATAA
ATTATATTTG
TTTAATGTTT
AGTGGTAGGT
TCTTTTTAAG
GTGTATAGAT
TGTGTGTTGA
GGTGATTAGA
AGAGGTATTG
GTGGGTGTAA.
ATTGCAAAGA
ACCTGCAAAT
TGTCTAAAGT
TGGTATAGTA
TAAATGTAGA
AAGGGGGGAG
TTAATAGAGT
GTGTAAGAGT
GGGTGTACAG
GAAGGGTTTT
CGATGGTTAT
CCCCTGGACA
AATGATAGTG
AAGTTCTTTT
TATATTTGAG
TTTGATTGAA
GATGTATTTA
A.ATAGTGTTT
GTTGAG
974 1034 1094 1154 1214 1274 1334 1394 1454 1S14 1574 1630 a. a INFORMATION FOR SEQ ID NO:li: SEQUENCE CHARAGTERISTIGS: LENGTH: 294 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLEGULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:ll: Gly Val Pro His Giu Gly Pro Leu His Pro Ala Pro 1 5 10 Pro Ala Pro Pro Pro Ala Ala Ser Arg Ser Met Phe 25 Gly Leu Gly Leu Gly Gin Val Val Gys Ser Ile Ala 40 Phe Arg Ala Gin Met Asp Pro Asn Arg Ile Ser Glu 55 Ser Ala Pro Ala Leu Ala Leu Leu Leu Phe Leu Tyr Asp Ser Thr His Cys Phe Tyr Arg Ile Leu Arg Leu His Glu Asn Ala Asp Leu Gin Asp 70 75 Ser Thr Leu Glu Ser Glu Asp Thr Leu Pro Asp Ser Cys Arg Arg Met 90 Lys Gin Ala Phe Gin Gly Ala Val Gin Lys Glu Leu Gin His Ile Val 100 105 110 Gly Pro Gin Arg Phe Ser Gly Ala Pro Ala Met Met Glu Gly Ser Trp 115 120 125 Leu Asp Val Ala Gin Arg Gly Lys Pro Glu Ala Gin Pro Phe Ala His 130 135 140 Leu Thr Ile Asn Ala Ala Ser Ile Pro Ser Gly Ser His Lys Val Thr 145 150 155 160 Leu Ser Ser Trp Tyr His Asp Arg Gly Trp Ala Lys Ile Ser Asn Met 165 170 175 Thr Leu Ser Asn Gly Lys Leu Arg Val Asn Gin Asp Gly Phe Tyr Tyr 180 185 190 Leu Tyr Ala Asn Ile Cys Phe Arg His His Glu Thr Ser Gly Ser Val 195 200 205 Pro Thr Asp Tyr Leu Gin Leu Met Val Tyr Val Val Lys Thr Ser Ile 210 215 220 Lys Ile Pro Ser Ser His Asn Leu Met Lys Gly Gly Ser Thr Lys Asn 225 230 235 240 Trp Ser Gly Asn Ser Glu Phe His Phe Tyr Ser Ile Asn Val Gly Gly S* 245 250 255 Phe Phe Lys Leu Arg Ala Gly Glu Glu Ile Ser Ile Gin Val Ser Asn 260 265 270 Pro Ser Leu Leu Asp Pro Asp Gin Asp Ala Thr Tyr Phe Gly Ala Phe 275 280 285 Lys Val Gin Asp Ile Asp 290 2) INFORMATION FOR SEQ ID NO:12: *00 SEQUENCE CHARACTERISTICS: LENGTH: 954 base pairs TYPE: nucleic acid STRANDEDNESS: single S(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL:
NO
(iv) ANTI-SENSE:
NO
(vi) ORIGINAL SOURCE: ORGANISM: H-omo sapiens (vii) IMMEDIATE SOURCE:
LIBRARY:
CLONE: huRANKL (full length) (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .951 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:i2: ATG CGC CGC GCC AGC AGA GAC TAC ACC AAG TAC CTG CGT GGC TCG GAG Met Arg Arg Ala Ser Arg Asp Tyr Thr Lys Tyr Leu Arg Gly Ser Glu 9* *9 9* *9 1
GAG
Glu
CCG
Pro
TTC
Phe
GCC
Ala
GAA
Glu
GCA
Ala
CCT
Pro
AAG
Lys
GCG
Ala 145
GAA
Glu
A~TG
Met
CCG
Pro
GTG
Val1
CTG
Leu
GAT
Asp
GAT
Asp
GAT
Asp
GAA
Glu 130
ATG
Met
GCT
Ala
GGC
Gly
CCG
Pro
GCC
Ala
TTC
Phe
GGC
Gly
TTT
Phe
TCA
Ser 115
TTA
Leu
GTG
Val1
CAG
Gin
GGC
Gly
CCT
Pro
CTC
Leu
TTC
Phe
ACT
Thr
CAA
Gin 100
TGT
Cys
CAA
Gin
GAT
Asp
CCT
Pro 5
GGC
Gly
GCG
Ala
CTG
Leu
TAT
Tyr
CAC
His
GAC
Asp
AGG
Arg
CAT
His
GGC
Gly
TTT
Phe 165
CCC
Pro
CCG
Pro
GGG
Gly
TTC
Phe 70
TGC
Cys
ACA
Thr
AGA
Arg
ATC
Ile
TCA
Ser 150
GCT
Ala
GGA
Gly
CAC
His
CTG
Leu 55
AGA
Arg
ATT
Ile
ACT
Thr
ATT
Ile
GTT
Val1 135
TGG
Trp
CAT
His
GCC
Ala
CAG
Gin 40
GGG
Gly
GCG
Ala
TAT
Tyr
CTG
Leu
AAA
Lys 120
GGA
Gly
TTA
Leu
CTC
Leu
CCG
Pro 25
CCC
Pro
CTG
Leu
CAG
Gin
AGA
Arg
GAG
Giu 105
CAG
Gin
TCA
Ser
GAT
Asp
ACT
Thr 10
CAC
His
CCC
Pro
GGC
Gly
ATG
Met
ATT
Ile 90
AGT
Ser
GCC
Ala
CAG
Gin
CTG
Leu
ATT
Ile 170
GAG
Giu
GCC
Ala
CAG
Gin
GAT
Asp 75
TTG
Leu
CAA
Gin
TTT
Phe
CAC
His
GCC
Al a 155
AAT
Asn
GGC
Gly
GCC
Ala
GTT
Val'
CCT
Pro
AGA
Arg
GAT
Asp
CAA
Gin
ATC
Ile 140
AAG
Lys
GCC
Ala
CC
Pro rCC Ser
GTC
Val1
AAT
Asn
CTC
Leu
ACA
Thr
GGA
Gly 125
AGA
Arg
AGG
Arg
ACC
CTG
Leu
CGC
Arg
TGC
Cys
AGA
Arg
CAT
His
AAA
Lys 110
OCT
Ala
GCA
Ala
AGC
Ser
GAC
CAC4 His
TCC
Ser
AGC
Ser
ATA
Ile
GAA
Giu
TTA
Leu
GTG
Val1
GAG
Giu
AAG
Lys
ATC
GCC
Ala
ATG
Met
GTC
Val1
TCA
Ser
AAT
Asn
ATA
Ile
CAA
Gin
AAA
Lys
CTT
Leu 160
CCA
96 144 192 240 288 336 384 432 480 528 Thr Asp Ile Pro 175 TCT GGT TCC CAT AAA GTG AGT CTG TCC TCT TGG TAC CAT GAT CGG GGT Ser Oly Ser His Lys Val Ser Leu Ser Ser Trp Tyr His Asp Arg Gly 180 185 190 576 TGG GCC AAG ATC TCC AAC ATG ACT TTT AGC AAT GGA AAA CTA ATA GTT 624 Trp Ala Lys Ile Ser Asn Met Thr Phe Ser Asn Gly Lys Leu Ile Val 195 200 205 AAT CAG GAT GGC TTT TAT TAC CTG TAT GC AAC ATT TGC TTT CGA CAT 672 Asn Gin Asp Gly Phe Tyr Tyr Leu Tyr Ala Asn Ile Cys Phe Arg His 210 215 220 CAT GAA ACT TCA GGA GAC CTA GCT ACA GAG TAT CTT CCAA CTA ATG GTG 720 His Glu Thr Ser Gly Asp Leu Ala Thr Glu Tyr Leu.Gln Leu Met Val 225 230 235 240 TAC GTC ACT AAA ACC AGC ATC AAA ATC CCA AGT TCT CAT ACC CTG ATG 768 Tyr Val Thr Lys Thr Ser Ile Lys Ile Pro Ser Ser His Thr Leu Met 245 250 255 AAA GGA GGA AGC ACC AAG TAT TGG TCA GGG AAT TCT GAA TTC CAT TTT 816 Lys Gly Gly Ser Thr Lys.Tyr Trp Ser Gly Asn Ser Glu Phe His Phe 260 265 270 TAT TCC ATA AAC GTT GGT GGA TTT TTT AAG TTA CGG TCT GGA GAG GAA 864 Tyr Ser Ile Asn Val Gly Gly Phe Phe Lys Leu Arg Ser Gly Glu Glu 275 280 285 ATC AGC ATC GAG GTC TCC AAC CCC TCC TTA CTG GAT CCG GAT CAG GAT 912 Ile Ser Ile Glu Val Ser Asn Pro Ser Leu Leu Asp Pro Asp Gin Asp 290 295 300 GCA ACA TAC TTT GGG GCT TTT AAA GTT CGA GAT ATA GAT TGA 954 Ala Thr Tyr Phe Gly Ala Phe Lys Val Arg Asp Ile Asp 305 310 315 INFORMATION FOR SEQ ID NO:13: SEQUENCE CHARACTERISTICS: LENGTH: 317 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: Met Arg Arg Ala Ser Arg Asp Tyr Thr Lys Tyr Leu Arg Gly Ser Glu 1 5 10 Glu Met Gly Gly Gly Pro Gly Ala Pro His Glu Gly Pro Leu His Ala 25 Pro Pro Pro Pro Ala Pro His Gin Pro Pro Ala Ala Ser Arg Ser Met 40 Phe Val Ala Leu Leu Gly Leu Gly Leu Gly Gin Val Val Cys Ser Val 55 Ala Leu Phe Phe Tyr Phe Arg Ala Gin Met Asp Pro Asn Arg Ile Ser 70 75 Glu Asp Gly Thr His Cys Ile Tyr Arg Ile Leu Arg Leu His Glu Asn 90 Ala Asp Phe Gin Asp Thr Thr Leu Giu Ser Gin Asp.Thr Lys Leu Ile Pro Lys Ala 145 Giu Ser Trp Asn His 225 Tyr Lys Tyr Ile Ala 305 A~sp G1u 130 Met Ala Gly Ala Gin 210 Giu Val1 Giy Ser Ser 290 Thr Ser Leu Val1 Gin Ser Lys 195 Asp Thr Thr Gly Ile 275 Ile Tyr 100 Cys Gin Asp Pro His 180 Ile Gly Ser Lys Ser 260 Asn Glu Phe A\rg His Gly Phe 165 Lys Ser Phe Gly Thr 245 Thr Val1 Val1 Giy Arg Ile Ser 150 Ala Val1 Asn Tyr Asp 230 Ser Lys Gly Ser Al a 310 Ilie Val1 135 Trp His Ser Met TPyr 215 Leu Ile Tyr Gly Asn 295 Phe Lys 120 G ly Leu Leu Leu Thr 200 Leu Ala Lys Trp Phe 280 Pro Lys 105 Gln Ser Asp Thr Ser 185 Phe Tyr Thr Ile Ser 265 Phe Ser Val Ala Gin Leu Ile 170 Ser Ser Ala Giu Pro 250 Giy Lys Leu Arg Phe His Ala 155 Asn Trp Asn Asn Tyr 235 Ser Asn Leu Leu Asp 315 Gin Ilie 140 Lys Ala Tyr Gly Ilie 220 Leu Ser Ser Arg Asp 300 Ile 110 Gly 125 Arg Arg Thr His Lys 205 Cys Gin His Giu Ser 285 Pro Asp Ala Ala Ser Asp Asp 190 Leu Phe Leu Thr Phe 270 Gly Asp Val1 Glu Lys Ile 175 Arg Ile Arg Met Leu 255 His Giu Gin Gin Lys Leu 160 Pro Gly Val1 His Val 240 Met Phe Giu Asp
S
S S
S
*5 S S
S
INFORMATION FOR SEQ ID NO:14: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 1878 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO (vi) ORIGINAL SOURCE: ORGANISM: Murine (vii) IMMEDIATE SOURCE: LIBRARY: Murine Fetal Liver Epithelium CLONE: muRANK (ix) FEATURE: NAME/KEY: CDS LOCATION: l..1875 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
ATG
Met 1 GCC CCG CGC Ala Pro Arg
GCC
Ala 5 CGG CGG CGC CGC Arg Arg Arg Arg
CAG
Gln 10 CTG CCC GCG CCG Leu Pro Ala Pro CTG CTG Leu Leu GCG CTC TGC Ala Leu. Cys CCT CCA TGC Pro Pro Cys
GTG
Val CTG CTC GTT CCA Leu. Leu Val Pro CAG GTG ACT CTC Gln Val Thr Leu CAG GTC ACT Gln Val Thr CGG TGT TGC Arg Cys Cys ACC CAG GAG AGG Thr Gln Glu. Arg
CAT
His 40 TAT GAG CAT CTC Tyr Glu. His Leu.
GGA
Gly AGC AGA Ser Arg TGC GAA CCA GGA Cys Glu. Pro Gly
AAG
Lys TAC CTG TCC TCT Tyr Leu, Ser Ser TGC ACT CCT ACC Cys Thr Pro Thr
TCC
Ser GAC AGT GTG TGT Asp Ser Val Cys CCC TGT GGC CCC Pro Cys Gly Pro
GAT
Asp GAG TAC TTG GAC Glu Tyr Leu Asp ***too 0*6.00 TGG AAT GAA GAA Trp Asn Glu. Glu AAA TGC TTG CTG Lys Cys Leu Leu
CAT
His AAA GTC TGT GAT Lys Val Cys Asp GCA GGC Ala Gly AAG GCC CTG Lys Ala Leu TGT GCT TGC Cys Ala Cys lis
GTG
Val 100 GCG GTG GAT CCT Ala Val Asp Pro
GGC
Gly 105 AAC CAC ACG GCC Asn His Thr Ala CCG CGT CGC Pro Arg Arg 110 GAG TGC TGC Glu Cys Cys 192 240 288 336 384 432 480 528 ACG GCT GGC TAC Thr Ala Gly Tyr
CAC
His 120 TGG AAC TCA GAC Trp Asn Ser Asp CGC AGG Arg Arg 130 AAC ACG GAG TGT Asn Thr Glu Cys
GCA
Ala 135 CCT GGC TTC GGA Pro Gly Phe Gly CAG CAT CCC TTG Gln His Pro Leu
CAG
Gln 145 CTC AAC AAG GAT Leu Asn Lys Asp
ACG
Thr 150 GTG TGC ACA CCC Val Cys Thr Pro
TGC
Cys IS5 CTC CTG GGC TTC! Leu. Leu Gly Phe
TTC
Phe 160 TCA GAT GTC TTT Ser Asp Val Phe
TCG
Ser 165 TCC ACA GAC AAA Ser Thr Asp Lys
TGC
Cys 170 AAA CCT TGG ACC Lys Pro Trp Thr AAC TGC Asn Cys 175 ACC CTC CTT Thr Leu. Leu
GGA
Gly 180 AAG CTA GAA GCA Lys Leu. Glu Ala CAG GGG ACA ACG Gln Gly Thr Thr GAA TCA GAT Glu. Ser Asp 190 GTG GTC TGC Val Val Cys 195 AGC TCT TCC Ser Ser Ser ATG ACA CTG AGG AGA Met Thr 200 CTC ATC Leu Ile 215 Leu Arg Arg CCA CCC ALAG GAG GCC Pro Pro Lys Giu Ala 205 CAG GCT Gin Ala 210 TAG CTG CCC AGT Tyr Leu Pro Ser GTT CTG CTC CTC Val Leu Leu Leu TTC ATC TCT GTG Phe Ile Ser Val
GTA
Val1 225 GTA GTG GCT GCC Val Val Ala Ala ATC TTC GGC GTT Ile Phe Gly Val
TAC
Tyr 235 TAC AGG AAG GGA Tyr Arg Lys Gly
GGG
Gly 240 AAA GCG CTG ACA Lys Ala Leu Thr
GCT
Ala 245 AAT TTG TGG AAT Asn Leu Trp Asn
TGG
Trp 250 GTC MAT GAT GCT Val Asn Asp Ala TG AGT Cys Ser 255 AGT CTA AGT Ser Leu Ser CAC TCG GCA His Ser Ala 275
GGA
Gly 260 AAT AAG GAG TCC Asn Lys Glu Ser
TCA
Ser 265 GGG GAC CGT TOT Gly Asp Arg Cys OCT GGT TCC Ala Gly Ser 270 ATC TTA CTA Ile Leu Leu 816 864 ACC TCC AGT CAG Thr Ser Ser Gin GAA GTG TGT GMA Giu Val Cys Glu
GT
Gly 285 ATG ACT Met Thr 290 COG GAG GAG MAG Arg Glu Glu Lys
ATG
Met 295 GTT CCA GAA GAC Val Pro Glu Asp GCT GOA GTC TOT Ala Oly Val Cys
GG
Gly 305 CCT GTG TGT GCG Pro Val Cys Ala
GCA
Ala 310 GOT GGG CCC TG Oly Gly Pro Trp
GCA
Ala 315 GMA OTC AGA GAT Glu Val Arg Asp
TCT
Ser 320 0O S. S 5 09 S S
S
Oe S
S.
AGG ACO TTC ACA Arg Thr Phe Thr GTC AGC GAG GTT Val Ser Olu Val
GAG
Oiu 330 ACG CMA GGA GAC Thr Gin Oly Asp CTC TCG Leu Ser 335 912 960 1008 1056 1104 AGO MAG ATT Arg Lys Ile TCG ACT GGT Ser Thr Gly 355 ACA GAG GAT GAG Thr Oiu Asp Oiu
TAC
Tyr 345 ACG GAG COO CCC Thr Asp Arg Pro TCG CAG CCT Ser Gin Pro 350 TCT ATA CCC Ser Ile Pro TCA CTG CTC CTA Ser Leu Leu Leu
ATC
Ile 360 CAG CAG GGA AGC Gin Gin Gly Ser CCA TTC Pro Phe 370 CAG GAG CCC CTG Gin Oiu Pro Leu
GMA
Giu 375 GTG GGG GAG AAC Vai Giy Oiu Asn
GAC
Asp 380 AGT TTA AGC CAG Ser Leu Ser Gin S. S S
S
*SSS
0 eS..
TGT
Cys 385 TTC ACC GGG ACT Phe Thr Gly Thr
GMA
Glu 390 AGC ACO GTG GAT Ser Thr Vai Asp
TCT
Ser 395 GAG GOC TGT GAC Oiu Giy Cys Asp 1152 1200 1248 1296 ACT GAG CCT CCG Thr Giu Pro Pro
AGC
Ser 405 AGA ACT GAC TCT ATO CCC GTG TCC CCT Arg Thr Asp Ser Met Pro Val Ser Pro 410 GMA MG Giu Lys 415 CAC CTG ACA His Leu Thr
AMA
Lys 420 GMA ATA GMA GOT GAC AGT TGC CTC CCC Gu Ile Oiu Gly Asp Ser Cys Leu Pro 425 TOO GTG GTC Trp Val Val 430 AGC TCC AAC Ser Ser Asn 435 TCA ACA GAT GGC Ser Thr Asp Gly TAC ACA Tyr Thr 440 GGC AGT GGG AAC ACT CCT GGG 1344 Gly Ser Gly Asn 445 Thr Pro Gly GAG GAC Glu Asp 450 CAT GAA CCC TTT His Glu Pro Phe
CCA
Pro 455 GGG TCC CTG AAA Gly Ser Leu Lys
TGT
Cys 460 GGA CCA TTG CCC Gly Pro Leu Pro
CAG
Gin 465 TGT GCC TAC AGC Cys Ala Tyr Ser
ATG
Met 470 GGC TTT CCC AGT Gly Phe Pro Ser
GAA
Glu 475 GCA GCA GCC AGC Ala Ala Ala Ser
ATG
Met 480 GCA GAG GCG GGA Ala Glu Ala Gly
GTA
Val 485 CGG CCC CAG GAC Arg Pro Gin Asp
AGG
Arg 490 GCT GAT GAG AGG Ala Asp Glu Arg GGA GCC Gly Ala 495 TCA GGG TCC Ser Gly Ser GTG ACT GGA Val Thr Gly 515 AGC TCC CCC AGT Ser Ser Pro Ser
GAC
Asp 505 CAG CCA CCT GCC Gin Pro Pro Ala TCT GGG AAC Ser Gly Asn 510 AAC AGT AAC TCC Asn Ser Asn Ser
ACG
Thr 520 TTC ATC TCT AGC GGG CAG GTG ATG Phe Ile Ser Ser Gly Gin Val Met 525 AAC TTC Asn Phe 530 AAG GGT GAC ATC Lys Gly Asp Ile
ATC
Ile 535 GTG GTG TAT GTC Val Val Tyr Val
AGC
Ser 540 CAG ACC TCG, CAG Gin Thr Ser Gin 1392 1440 1488 1536 1584 1632 1680 1728 1776 1824 1872 1878
GAG
Glu 545 GGC CCG GGT TCC Gly Pro Gly Ser GAG CCC GAG TCG Glu Pro Glu Ser
GAG
Glu 555 CCC GTG GGC CGC Pro Val Gly Arg GTG CAG GAG GAG Val Gin Glu Glu
ACG
Thr 565 CTG GCA CAC AGA Leu Ala His Arg TCC TTT GCG GGC Ser Phe Ala Gly ACC GCG Thr Ala 575 CCG CGC TTC Pro Arg Phe GGG GCA CCC Gly Ala Pro 595
CCC
Pro 580 GAC GTC TGT GCC Asp Val Cys Ala
ACC
Thr 585 GGG GCT GGG CTG Gly Ala Gly Leu CAG GAG CAG Gin Glu Gin 590 CAG GAG CAG Gin Glu Gin CGG CAG AAG GAC Arg Gin Lys Asp
GGG
Gly 600 ACA TCG CGG CCG Thr Ser Arg Pro GGT GGG Gly Gly 610 GAA TGA Glu 625 GCG CAG ACT TCA Ala Gin Thr Ser
CTC
Leu 615 CAT ACC CAG GGG His Thr Gin Gly
TCC
Ser 620 GGA CAA TGT GCA Gly Gin Cys Ala INFORMATION FOR SEQ ID SEQUENCE
CHARACTERISTICS:
LENGTH: 625 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID Met Ala Pro Arg Ala Arg Arg Arg Arg Gin Leu Pro Ala Pro Leu Leu a. a a a. a.
Ala Pro Ser Ser Trp Lys Cys Arg Gin 145 Ser Thr Val1 Gin Val1 225 Lys Ser His Leu Pro Arg Asp Asn Ala Ala Arg 130 Leu Asp Leu Val1 Ala 210 Val1 Ala Leu Ser Cys Cys Cys Ser Glu Leu Cys 115 As n Asn Val Leu Cys 195 Tyr Val Leu Ser Ala 275 Val1 Thr Glu Val Glu Val 100 Thr Thr Lys Phe Gly 180 Ser Leu Ala Thr Gly 260 Thr Leu Gin Pro Cys Asp Ala Ala Glu Asp Ser 165 Lys Ser Pro Ala Ala 245 Asn Sex Leu Giu Gly Leu 70 Lys Val1 Gly Cys Thr 150 Ser Leu Ser Ser Ile 230 *Asn Lays *Ser Vai A~rg Lys 55 Pro Cys Asp Tyr Ala 135 Val Thr Glu Met Leu 215 Ile Leu Glu Gin Pro His 40 Tyr Cys Leu Pro His 120 Pro Cys Asp Ala Thr 200 Ile Phe Trp Ser Gin 280 Leu 25 Tyr Leu Gly Leu Gly 105 Trp Gly Thr Lys His 185 Leu Val1 Gly Asn Ser 265 Glu Gin Giu Ser Pro His 90 Asn Asn Phe Pro Cys 170 Gin Arg Leu Val1 *Trp 250 *Gly Val Val1 His Ser Asp 75 Lys His Ser Gly Cys 155 Lys Giy Arg Leu Tyr 235 Va 1 Asp Cys Thr Leu Lys Glu Val1 Thr Asp Ala 140 Leu Pro Thr Pro Leu 220 Tyr Asn Arg Glu Leu Gly Cys Tyr Cys Ala Cys 125 Gin Leu Trp Thr Pro 205 Phe Arg Asp Cys Gly 285 Gin Arg Thr Leu Asp Pro 110 Glu His Gly Thr Glu 190 Lys Ile Lys Ala Ala 270 Ile Val1 Cys Pro Asp Ala Arg Cys Pro Phe Asn 175 Ser Glu Ser Gly Cys 255 Gly Leu Thr Cys Thr Thr Gly Arg Cys Leu Phe 160 Cys Asp Ala Val1 Gly 240 Ser Ser Leu Met Thr 290 Arg Giu Glu Lys Met 295 Val Pro Giu Asp Gly 300 Ala Gly Val Cys WO 98/28424 PCT/US97/23866 Cys Ala Ala Gly Gly Pro Trp Ala Glu Val Arg Asp Ser 310 315 320 dly 305 Arg Arg Ser Pro Cys 385 Thr His Ser Giu Gin 465 Ala Ser Val1 As n Glu 545 Val Pro Gly Pro Th r Lys Thr Phe 370 Phe Glu Leu Ser Asp 450 Cys Glu Gly Thr Phe 530 Gly Gin Arg Ala *Val Phe Ile Gly 355 Gin Thr Pro Thr Asn 435 His Ala Ala Ser dly 515 Lys Pro Glu Phe Pro 595 Thr Pro 340 Ser Giu Gly Pro Lys 420 Ser Giu Tyr Gly Gly 500 Asn Gly Gly Glu Pro 580 Arg Leu 325 Thr Leu Pro Thr Ser 405 Giu Thr Pro Ser Val1 485 Ser Ser Asp Ser Thr 565 Asp Gin Val1 Glu Leu Leu Giu 390 Arg Ile Asp Phe Met 470 Arg Ser Asn Ile Ala 550 Leu Val1 Lys 9cr Asp Leu Glu 375 Ser Thr Glu Giy Pro 455 Gly Pro Pro Ser Ile 535 Giu Ala Cys Asp Glu Glu Ile 360 Val1 Thr Asp Gly Tyr 440 Giy Phe Gin Ser Thr 520 Val1 Pro His Ala Gly 600 Val1 Tyr 345 Gin Giy Val1 Ser Asp 425 Thr Ser Pro Asp Asp 505 Phe Val Giu Arg Thr 585 Thr Giu 330 Thr Gin Giu Asp Met 410 Ser Giy Leu Ser Arg 490 Gin Ile Tyr Scr Asp 570 Gly Ser Thr Asp Gly Asn Ser 395 Pro Cys Ser Lys Giu 475 Ala Pro Ser Val1 Giu 555 Ser Ala Arg Gin Arg Ser Asp 380 Giu Vai Leu Gly Cys 460 Ala Asp Pro Ser Ser 540 Pro Phe Gly Pro Gly Pro Lys 365 Ser Giy Ser Pro Asn 445 Gly Ala Giu Aia Gly 525 Gin Val1 Ala Leu Val1 605 Asp 9cr 350 Ser Leu Cys Pro Trp 430 Thr Pro Ala Arg Ser 510 Gin Thr Gly Gly Gin 590 Gin Leu 335 Gin Ile Ser Asp Giu 415 Val1 Pro Leu Ser Gly 495 Gly Val1 Ser Arg Thr 575 Glu Giu Ser Pro Pro Gin Phe 400 Lys Val1 Gly Pro Met 480 Ala Asn Met Gin Pro 560 Ala Gin Gin Gly Gly 610 Ala Gin Thr Ser Leu 615 His Thr Gin Gly Ser Gly Gin Cys Ala 620 Glu 625 INFORMATION FOR SEQ ID NO:16: SEQUENCE CHARACTERISTICS: LENGTH: 20 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16: Met Glu Thr Asp Thr Leu Leu Leu Trp Val Leu Leu Leu Trp Val Pro 1 5 10 Gly Ser Thr Gly INFORMATION FOR SEQ ID NO:17: SEQUENCE CHARACTERISTICS: LENGTH: 5 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17: Asp Tyr Lys Asp Glu o INFORMATION FOR SEQ ID NO:18: SEQUENCE CHARACTERISTICS: LENGTH: 6 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18: His His His His His His INFORMATION FOR SEQ ID NO:19: SEQUENCE CHARACTERISTICS: LENGTH: 33 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: Arg Met Lys Gin Ile Giu Asp Lys Ile Giu Giu Ile Leu Ser Lys Ile 1 5 10 Tyr His Ile Glu Asn Glu Ile Ala Arg Ile Lys Lys Leu Ile Gly Glu 25 Arg 68

Claims (19)

1. A method of screening a molecule for its capacity to antagonize or agonize RANK, said method comprising an assay selected from the group consisting of: a) i) contacting a RANK polypeptide with a RANKL polypeptide in the presence of the molecule under conditions that permit binding of the RANK and RANKL polypeptides; ii) detecting the binding of the RANK and RANKL polypeptides; iii) determining that the molecule is a RANK antagonist if the amount of RANK-RANKL binding detected is decreased when the molecule is present; and iv) determining that the molecule is a RANK agonist if the amount of RANK-RANKL binding detected is increased when the molecule is present; *e b) i) triggering RANK in a culture of cells that is transfected with a plasmid in which an NF-KB-dependent promoter is operably linked to a reporter 20 gene, wherein during said triggering the cultured cells are exposed to the molecule; ii) detecting a product of the reporter gene in said exposed cells; iii) determining that the molecule is a RANK antagonist if the amount of the reporter gene product detected in said exposed cells is decreased; 25 and iv) determining that the molecule is a RANK agonist if the amount of the reporter gene product detected in said exposed cells is increased; c) i) incubating the molecule and a RANKL polypeptide with isolated CDla+ dendritic cells; ii) monitoring the formation of dendritic cell clusters; iii) determining that the molecule is a RANK antagonist if dendritic cell clustering is decreased when the molecule is present; and IC W:\ilona\Sharon\SJJspec\SP713473.doc iv) determining that the molecule is a rank agonist if dendritic cell clustering is increased when the molecule is present; d) i) incubating the molecule and a RANKL polypeptide with isolated CD1a' dendritic cells in a medium comprising GM-CSF, IL-4, TNF-a and Flt3L; ii) irradiating the incubated dendritic cells; iii) monitoring the allostimulatory capacity of the irradiated dendritic cells in a mixed lymphocyte reaction; iv)determining that the molecule is a RANK antagonist if the allostimulatory capacity of the irradiated dendritic cells is decreased when the molecule is present; and v) determining that the molecule is a RANK agonist if the allostimulatory capacity of the irradiated dendritic cells is increased when the molecule is present; e) i) triggering RANK in the presence of the molecule in cells capable of expressing RANK; 20 ii) measuring TNF-a production or NF-KB activation in the cells; iii) determining that the molecule is a RANK antagonist if TNF-a production or NF-KB activation in said cells is decreased when the molecule is present; and iv) determining that the molecule is a RANK agonist if TNF- production or NF-KB activation in said cells is increased when the molecule is S. present; f) i) activating peripheral blood T cells in culture in the presence of the molecule and one or more cytokines capable of upregulating RANK, said cytokine(s) being selected from the group consisting of TGF-P and IL-4; IC W:lonaShron\SJJspeci\SP713473.doc 71 ii) incubating said T cells for at least 4 days; iii) detecting viable T cells in said culture; iv) determining that the molecule is a RANK antagonist if the number of viable T cells detected is reduced when the molecule is present; and v) determining that the molecule is a RANK agonist if the number of viable T cells detected is increased when the molecule is present; g) i) incubating the molecule with cells expressing RANK; ii) detecting TNF-a or activated NF-KB in the incubated cells; iii) determining that the molecule is a RANK antagonist if the amount of TNF-a or activated NF-KB detected is decreased when the molecule is present; and iv) determining that the molecule is a RANK antagonist if the amount of TNF-a or activated NF-KB detected is increased when the molecule is present; and h) I) incubating the molecule with cells expressing RANK, said cells being transfected with a plasmid in which an NF-KB-dependent promoter is 20 operably linked to a reporter gene; ii) detecting a product of the reporter gene in the incubated cells; iii) determining that the molecule is a RANK antagonist if the amount of the reporter gene product detected is decreased when the molecule is present; and iv) determining that the molecule is a RANK agonist if the amount of the reporter gene product detected in said incubated cells is increased when the molecule is present.
2. A method according to claim 1 wherein the cells capable of expressing RANK are selected from the group consisting of macrophages, epithelial cells, B- cells, activated T cells and cells transfected with recombinant RANK. IC W:flonalShamn\SJJspecdSP713473.doc 72
3. The method of claim 1 or 2, wherein the molecule is a soluble RANK polypeptide selected from the group consisting of: a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO: 6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and amino acids 3-232 of SEQ ID NO:8; b) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID wherein x is amino acid 1 or 31 SEQ ID NO: 15, and amino acids 3-232 of SEQ ID NO:8; and c) amino acids x to 213 of SEQ ID NO:6, Wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, or fragments thereof.
4. A method according to any one of claims 1 to 3, wherein the assay employs a RANKL polypeptide and the RANKL polypeptide is selected from the group consisting of membrane-associated RANKL and soluble RANKL. 0 5. A method according to any one of claims 1, 2 or 4, wherein the molecule is an 20 antibody against RANK or RANKL.
6. A method according to any one of claims 1 to 5, wherein the detecting step is selected from the group consisting of enzyme-linked immunosorbent assay (ELISA), dot blot employing an antibody that binds native RANK, fluorescence activated cell 25 sorter assay (FACS assay) and a solid phase binding assay.
7. A method according to claim 1 wherein said NF-KB-dependent promoter is IL-8 promoter.
8. A method according to claim 1, wherein the assay employs RANK triggering and the RANK triggering step is selected from the group consisting of RANK over- expression, expression of membrane-bound RANKL in a cell that is expressing RANK, addition of a soluble RANKL polypeptide to cells expressing RANK, incubation of IC W:\ilona\SharnSJJspeciSP713473.doc 73 cells expressing RANK with cells expressing membrane-bound RANKL and addition of an agonistic antibody against RANK to cells expressing RANK.
9. A method according to claim 4, wherein the RANK polypeptide is selected from the group consisting of: a) a polypeptide having the amino acid sequence of SEQ ID b) a polypeptide having the amino acid sequence of SEQ ID NO:12; c) a soluble poly-His RANKL comprising a kappa chain leader having the amino acid sequence of SEQ ID NO: 16, a FLAGTM tag having the amino acid sequence of SEQ ID NO: 17, Gly Ser, a poly-His tag having the amino acid sequence of SEQ ID NO:18, Gly Thr Ser and amino acids S* 138-317 of SEQ ID NO:13; d) a soluble poly-His RANKL comprising amino acids x to 31 of SEQ ID NO:9, wherein x is amino acid 21, 32 or 29 of SEQ ID NO:9, Arg Thr Ser, 20 a poly-His tag having the amino acid sequence of SEQ ID NO:18, and amino acids 119-294 of SEQ ID NO:11; and e) a soluble oligomeric form of RANKL comprising amino acids x to 31 of SEQ ID NO:9, wherein x is amino acid 21, 32 OR 29 OF SEQ ID NO:9, 25 Asp, a leucine zipper having the amino acid sequence of SEQ ID NO:19, Thr Arg Ser and amino acids 138-317 of SEQ ID NO:13. A method of screening a molecule for its capacity to antagonize or agonize the binding of a TRAF-binding RANK polypeptide to a TRAF polypeptide comprising the steps of: IC W:\iona\Shamn\SJJspedcSP713473.doc 74 a) incubating a mixture comprising the molecule, a TRAF polypeptide and a TRAF-binding RANK polyeptide, wherein said mixture is incubated under conditions that allow the RANK polypeptide and the TRAF polypeptide to form RANK/TRAF complexes; b) detecting the RANK/TRAF complexes; c) determining that the molecule antagonizes binding of the rank polypeptide to the TRAF polypeptide if the amount of the RANK/TRAF complexes detected is decreased when the molecule is present; and d) determining that the molecule agonizes binding of the RANK polyeptide to the TRAF polypeptide if the amount of the RANK/TRAF complexes detected is increased when the molecule is present.
11. A method according to claim 10, wherein the TRAF polypeptide is selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5, and TRAF6. 20 12. A method according to claim 10, wherein the TRAF-binding RANK polypeptide Sos is selected from the group consisting of: a) a RANK/GST fusion protein; and 9 o* 25 b) a RANK/Fc fusion protein c) a polypeptide comprising amino acids 234-616 of SEQ ID NO:6; d) a polypeptide having an amino terminus selected from the group consisting of an amino acid between amino acid 1 and amino acid 33, inclusive, of SEQ ID NO:6, and a carboxy terminus selected from the group consisting of an amino acid between amino acid 234 and amino acid 616, inclusive; IC W:.ionaSharo\SJJSpeaSP7i3473.doc e) a polypeptide having an amino terminus selected from the group consisting of an amino acid between amino acid 1 and amino acid inclusive, of SEQ ID NO:15, and a carboxy terminus selected from the group consisting of an amino acid between amino acid 246 and amino acid 625, inclusive; f) a polypeptide comprising amino acids 544-616 of SEQ ID NO:6; g) a polypeptide comprising amino acids 339-421 of SEQ ID NO:6; h) a polypeptide comprising amino acids 236-625 of SEQ ID i) a fragment of a polypeptide of or j) a polypeptide having an amino acid sequence e at least about identical to a polypeptide of or
13. A method for therapeutically inhibiting RANK activity in a subject in need thereof, said method comprising the step of administering to said subject a therapeutically sufficient amount of a composition comprising a RANK antagonist.
14. A method according to claim 13, wherein the RANK antagonist is selected from the group consisting of an antagonistic antibody against RANK, an antagonistic antibody against RANKL and a soluble RANK polypeptide. a
15. A method according to claim 48, wherein the RANK antagonist is a soluble RANK polypeptide that is selected from the group consisting of: a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO;6, and amino acids 3-232 of SEQ ID NO:8; IC W:MIonaShamnSJJspecSP713473.d b) a RANK;Fc polypeptide comprising amino acids x to 213 of SEQ ID wherein x is amino acid 1 or 31 of SEQ ID NO:15, and amino acids 3-232 of SEQ ID NO:8: c) amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1 or any one of amino acids 24-33, inclusive, or fragments thereof; and d) a RANK/GST fusion protein.
16. A method according to any one of claims 13 to 15, wherein said composition further comprises a physiologically acceptable carrier, excipient or diluent.
17. A method according to any one of claims 13 to 16, wherein said composition is administered by bolus injection, continuous infusion or sustained release from an implant. o.
18. A method according to claims 13 to 17, wherein the subject is selected from the group consisting of a subject having a tumor or neoplastic disease, a subject 20 having an inflammatory condition, a subject having septic shock and a subject having graft-versus-host reaction.
19. A method according to any one of claims 13 to 18, wherein the subject has a tumor or neoplastic condition and is undergoing radiation therapy. A method of therapeutically inhibiting RANK activity in a subject having a tumor or neoplastic disease, septic shock, an inflammatory condition or a graft- versus-host reaction comprising administering to said subject a RANK polypeptide selected from the group consisting of: a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and amino acids 3-232 of SEQ ID NO:8; IC W:\naShamnnSJJspec\SP713473.doC 77 b) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID wherein x is amino acid 1 or 31 of SEQ ID NO:15, and amino acids 3-232 of SEQ ID NO:8; c) amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, or fragments thereof; and d) a RANK/GST fusion protein.
21. A method for stimulating an immune response in a subject who is undergoing vaccination comprising administering to said subject an amount of a RANKL polypeptide sufficient to induce DC maturation.
22. The method of claim 21, wherein the RANKL polypeptide is selected from the group consisting of: a) a polypeptide having the amino acid sequence of SEQ ID NO:11; b) a polypeptide having the amino acid sequence of SEQ ID NO:13; c) a soluble poly-His RANKL comprising a kappa chain leader having the amino acid sequence of SEQ ID NO:16, a FLAGTM tag having the amino acid sequence of SEQ ID NO: 17, Gly Ser, a poly-His tag having the amino acid sequence of SEQ ID NO: 18, Gly Thr Ser and amino acids 138-317 of 25 SEQ ID NO:13; d) a soluble poly-His RANKL comprising a CMV leader having the amino acid sequence of SEQ ID NO:9, Arg Thr Ser, a poly-His tag having the amino acid sequence of SEQ ID NO:18, and amino acids 119-294 of SEQ ID NO:11; and IC W:\lonalShamnSJJspeci\SP713473.doc 78 e) a soluble oligomeric form of RANKL comprising a CMV leader having the amino acid sequence of SEQ ID NO:9, Asp, a leucine zipper having the amino acid sequence of SEQ ID NO:19, Thr Arg Ser and amino acids
138-317 of SEQ ID NO:13. 23. Use of a composition comprising a RANK antagonist for the manufacture of a medicament for therapeutically inhibiting RANK activity in a mammal. 24. A use according to claim 23, wherein the RANK antagonist is selected from the group consisting of an antagonistic antibody against RANK, an antagonistic antibody against RANKL and a soluble RANK polypeptide. A use according to claim 23, wherein the RANK antagonist is a soluble RANK polypeptide that is selected from the group consisting of: a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and amino acids 3-232 of SEQ ID NO:8; b) a RANK;Fc polypeptide comprising amino acids x to 213 of SEQ ID wherein x is amino acid 1 or 31 of SEQ ID NO:15, and amino acids 3-232 of SEQ ID NO:8; 25 c) amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1 or any one of amino acids 24-33, inclusive, or fragments thereof; and d) a RANK/GST fusion protein. 26. A use according to any one of claims 23 to 25, wherein said composition further comprises a physiologically acceptable carrier, excipient or diluent. IC W:llonaShamn\SJJspec\SP713473.doc 79 27. A use according to any one of claims 23 to 26, wherein said composition is administered by bolus injection, continuous infusion or sustained release from an implant. 28. A use according to any one of claims 23 to 27, wherein said mammal is selected from the group consisting of a mammal having a tumor or neoplastic disease, a mammal having an inflammatory condition, a mammal having septic shock and a mammal having graft-virus host reaction. 29. A use according to any one of claims 23 to 28, wherein the mammal has a tumor or neoplastic condition and is undergoing reaction therapy. A use of a RANK polypeptide for the manufacture of a medicament for therapeutically inhibiting RANK activity in a mammal having a tumor or neoplastic disease, septic shock, an inflammatory condition or a graft-versus-host reaction, wherein said RANK polypeptide is selected from the group consisting of: a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, 20 wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and amino acids 3-232 of SEQ ID NO:8; b) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:15, wherein x is amino acid 1 or 31 of SEQ ID NO:15, and amino S 25 acids 3-232 of SEQ ID NO:8; Sc) amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, or fragments thereof; and d) a RANK/GST fusion protein. 31. Use of a RANKL polypeptide in an amount sufficient to induce DC maturation, for the manufacture of a medicament for stimulating on immune response in a mammal. IC W:\ilona\SharonSJJspec\SP713473.doc 32. A use according to claim 31, wherein the RANKL polypeptide is selected from the group consisting of: a) a polypeptide having the amino acid sequence of SEQ ID NO:11; b) a polypeptide having the amino acid sequence of SEQ ID NO:13; c) a soluble poly-His RANKL comprising a kappa chain leader having the amino acid sequence of SEQ ID NO:16, a FLAGTM tag having the amino acid sequence of SEQ ID NO:17, Gly Ser, a poly-His tag having the amino acid sequence of SEQ ID NO:18, Gly Thr Ser and amino acids 138-317 of SEQ ID NO:13; d) a soluble poly-His RANKL comprising a CMV leader having the amino acid sequence of SEQ ID NO:9, Arg Thr Ser, a poly-His tag having the amino acid sequence of SEQ ID NO:18, and amino acids 119-294 of SEQ ID NO:11; and 20 e) a soluble oligomeric form of RANKL comprising a CMV leader having the amino acid sequence of SEQ ID NO:9, Asp, a leucine zipper having the amino acid sequence of SEQ ID NO:19, Thr Arg Ser and amino acids 138-317 of SEQ ID NO:13. 33. A method according to claim 1, substantially as hereinbefore described. 34. A use according to claim 23, substantially as hereinbefore described. DATED: 1 March, 2000 PHILLIPS ORMONDE FITZPATRICK Attorneys for: IMMUNEX CORPORATION IC W:UJionShamnSdJspecMP71347.doc
AU20619/00A 1996-12-23 2000-03-02 Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily Expired AU741504B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU20619/00A AU741504B2 (en) 1996-12-23 2000-03-02 Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60/059978 1996-12-23
US08/813509 1997-03-07
US60/064671 1997-10-14
AU20619/00A AU741504B2 (en) 1996-12-23 2000-03-02 Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU57184/98A Division AU713473B2 (en) 1996-12-23 1997-12-22 Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily

Publications (2)

Publication Number Publication Date
AU2061900A true AU2061900A (en) 2000-06-01
AU741504B2 AU741504B2 (en) 2001-12-06

Family

ID=3709910

Family Applications (1)

Application Number Title Priority Date Filing Date
AU20619/00A Expired AU741504B2 (en) 1996-12-23 2000-03-02 Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily

Country Status (1)

Country Link
AU (1) AU741504B2 (en)

Also Published As

Publication number Publication date
AU741504B2 (en) 2001-12-06

Similar Documents

Publication Publication Date Title
AU713473B2 (en) Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily
WO1998028426A9 (en) Ligand for receptor activator of nf-kappa b, ligand is member of tnf superfamily
AU741504B2 (en) Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily
CA2274803C (en) Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily

Legal Events

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