EP1240183A2 - Genes de mammifere, reactifs associes et procedes - Google Patents

Genes de mammifere, reactifs associes et procedes

Info

Publication number
EP1240183A2
EP1240183A2 EP00978596A EP00978596A EP1240183A2 EP 1240183 A2 EP1240183 A2 EP 1240183A2 EP 00978596 A EP00978596 A EP 00978596A EP 00978596 A EP00978596 A EP 00978596A EP 1240183 A2 EP1240183 A2 EP 1240183A2
Authority
EP
European Patent Office
Prior art keywords
stamp
dsp
protein
cells
cell
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.)
Withdrawn
Application number
EP00978596A
Other languages
German (de)
English (en)
Inventor
Constance H. Zlot
Gosse Jan Adema
Carl Figdor
Joseph H. Phillips
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.)
Stichting Katholieke Universiteit
Merck Sharp and Dohme LLC
Original Assignee
Schering 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 Schering Corp filed Critical Schering Corp
Publication of EP1240183A2 publication Critical patent/EP1240183A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention pertains to compositions related to proteins which function in controlling biology and physiology of mammalian cells, e.g., cells of a mammalian immune system.
  • mammalian cells e.g., cells of a mammalian immune system.
  • it provides purified genes, proteins, antibodies, and related reagents useful, e.g., to regulate activation, development, differentiation, and function of various cell types, including hematopoietic cells.
  • BACKGROUND OF THE INVENTION refers generally to the technique of integrating genetic information from a donor source into vectors for subsequent processing, such as through introduction into a host, whereby the transferred genetic information is copied and/or expressed in the new environment.
  • the genetic information exists in the form of complementary DNA (cDNA) derived from messenger RNA (mRNA) coding for a desired protein product.
  • cDNA complementary DNA
  • mRNA messenger RNA
  • the carrier is frequently a plasmid having the capacity to incorporate cDNA for later replication in a host and, in some cases, actually to control expression of the cDNA and thereby direct synthesis of the encoded product in the host .
  • B-cells which can produce and secrete immunoglobulins (proteins with the capability of recognizing and binding to foreign matter to effect its removal)
  • T-cells of various subsets that secrete lymphokines and induce or suppress the B-cells and various other cells (including other T-cells) making up the immune network.
  • lymphocytes interact with many other cell types .
  • DC Dendritic cells
  • APC professional antigen presenting cells
  • DC are bone marrow-derived cells, that in their immature stage are scattered throughout the body and are particularly efficient in antigen uptake through a variety of cell surface receptors specialized in capturing antigens. See Sallusto, et al. (1995) J. EXP. Med. 182:389-400. Upon inflammation, DC migrate via lymph or blood to the secondary lymphoid organs . This migration process seems to be regulated by a coordinated up- and downregulation of chemokine receptors. See Sallusto, et al . (1998) Eur . J . Immunol . 28:2760-2769; and Sozzani, et al. (1998) J . Immunol . 161:1083-1086.
  • DC Upon arrival in the T cells areas, DC are mature and fully stimulatory, well-equipped to attract and interact with naive T cells. These DC express high levels of MHC class I, MHC class II, adhesion molecules and co-stimulatory molecules, that supports the induction of primary T cell responses. Subsequently, CD40 ligation on the DC after interaction with CD4+ T helper cells leads to maximal activation of the mature DC, further increasing their capacity to stimulate naive cytotoxic T lymphocytes. See Caux, et al . (1994) J. Exp . Med . 180:1263-1272; Cella, et al . (1996) J. EXP. Med. 184:747-752: Schoenberger, et al .
  • mast cell which has not been positively identified in all mammalian species
  • NK cells NK cells
  • macrophages a granule-containing connective tissue cell located proximal to capillaries throughout the body. These cells are found in especially high concentrations in the lungs, skin, and gastrointestinal and genitourinary tracts.
  • Mast cells play a central role in allergy-related disorders, particularly anaphylaxis as follows: when selected antigens crosslink one class of immunoglobulins bound to receptors on the mast cell surface, the mast cell degranulates and releases mediators, e.g., histamine, serotonin, heparin, and prostaglandins, which cause allergic reactions, e.g., anaphylaxis.
  • mediators e.g., histamine, serotonin, heparin, and prostaglandins
  • the present invention provides new compositions and related compounds, and methods for their use.
  • the present invention is directed to mammalian, e.g., rodent, canine, feline, primate, proteins designated Dendritic Cell Specific Transmembrane Protein (DC-STAMP) and DNAX Surface Protein (DSP-1) and their biological activities.
  • DC-STAMP Dendritic Cell Specific Transmembrane Protein
  • DSP-1 DNAX Surface Protein
  • nucleic acids coding for polypeptides themselves and methods for their production and use include nucleic acids coding for polypeptides themselves and methods for their production and use.
  • the nucleic acids of the invention are characterized, in part, by their homology to complementary DNA (cDNA) sequences disclosed herein, and/or by functional assays applied to the polypeptides, which are typically encoded by these nucleic acids. Methods for modulating or intervening in the control of surface protein dependent physiology or an immune response are provided.
  • the present invention is based, in part, upon the discovery of novel surface proteins from dendritic cells or mast cells. In particular, it provides primate, e.g., human, sequences. Functional equivalents exhibiting significant sequence homology will be available from other mammalian, e.g., cow, horse, rat, mouse, and non-mammalian species, e.g., warm blooded animals, including birds.
  • the invention provides: a substantially pure or recombinant DC-STAMP or DSP-1 polypeptide exhibiting identity over a length of at least about 12 amino acids to SEQ ID NO: 2 or 5 or 7; a natural sequence DC-STAMP of SEQ ID NO: 2; a natural sequence DSP-1 of SEQ ID NO : 5 or 7; and a fusion protein comprising DC-STAMP or DSP-1 sequence.
  • the segment of identity is at least about 14, 17, or 19 amino acids.
  • the DC-STAMP or DSP-1 comprises a mature sequence comprising the sequence from Tables 1 or 2; or exhibits a post-translational modification pattern distinct from natural DC-STAMP or DSP-1; or the polypeptide: is from a warm blooded animal selected from a mammal, including a primate; comprises at least one polypeptide segment of SEQ ID NO : 2 or 5 or 7; exhibits a plurality of fragments; is a natural allelic variant of DC- STAMP or DSP-1; has a length at least about 30 amino acids; exhibits at least two non-overlapping epitopes which are specific for a primate DC-STAMP or DSP-1; exhibits sequence identity over a length of at least about 20 amino acids to primate DC-STAMP or DSP-1; is glycosylated; has a molecular weight of at least 30 kD with natural glycosylation; is a synthetic polypeptide; is attached to a solid substrate; is conjugated to another chemical moiety; is a
  • Preferred embodiments include a composition comprising: a sterile DC- STAMP or DSP-1 polypeptide; or the DC-STAMP or DSP-1 polypeptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
  • the protein can have: mature polypeptide sequence from Tables 1 or 2 ; a detection or purification tag, including a FLAG, His6, or Ig sequence; and/or sequence of another cytokine or chemokine, including Flt3 ligand.
  • Kit embodiments include those with a DC-STAMP or DSP-1 polypeptide, and: a compartment comprising the polypeptide; and/or instructions for use or disposal of reagents in the kit.
  • the compound may have an antigen binding site from an antibody, which specifically binds to a natural DC-STAMP or DSP-1 polypeptide, wherein: the DC- STAMP or DSP-1 is a primate protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a peptide sequence of a mature polypeptide portion from Tables 1 or 2; is raised against a mature DC-STAMP or DSP-1; is raised to a purified primate DC-STAMP or DSP-1; is immunoselected; is a polyclonal antibody; binds to a denatured DC-STAMP or DSP-1; exhibits a Kd of at least 30 ⁇ M; is attached to a solid substrate, including a
  • Kits containing binding compounds include those with: a compartment comprising the binding compound; and/or instructions for use or disposal of reagents in the kit. Often the kit is capable of making a qualitative or quantitative analysis.
  • Preferred compositions will comprise: a sterile binding compound; or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
  • Nucleic acid embodiments include an isolated or recombinant nucleic acid encoding a DC-STAMP or DSP-1 polypeptide or fusion protein, wherein: the DC-STAMP or DSP-1 is from a primate; and/or the nucleic acid: encodes an antigenic peptide sequence of Tables 1 or 2; encodes a plurality of antigenic peptide sequences of Tables 1 or 2; exhibits identity to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a primate, including a human; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding the DC-STAMP or DSP-1; or is a PCR primer, PCR product, or mutagenesis primer.
  • the invention also provides a cell, tissue, or organ comprising such a recombinant nucleic acid, and preferably the cell will be: a prokaryotic cell; a eukaryotic cell; a bacterial cell; a yeast cell; an insect cell; a mammalian cell; a mouse cell; a primate cell; or a human cell.
  • Kit embodiments include those with such nucleic acids, and: a compartment comprising the nucleic acid; a compartment further comprising the DC-STAMP or DSP-1 protein or polypeptide; and/or instructions for use or disposal of reagents in the kit.
  • the kit is capable of making a qualitative or quantitative analysis.
  • the nucleic acid hybridizes under wash conditions of 30° C and less than 2M salt, or of 45° C and/or 500 mM salt, or 55° C and/or 150 mM salt, to SEQ ID NO: 1 or 4 or 6; or exhibits identity over a stretch of at least about 30, 55, or 75 nucleotides, to a primate DC-STAMP or DSP- 1.
  • the invention embraces a method of modulating physiology or development of a cell or tissue culture cells comprising contacting the cell with an agonist or antagonist of a primate DC-STAMP or DSP-1.
  • the method may be where: the contacting is in combination with an agonist or antagonist of Flt3 ligand; or the contacting is with an antagonist, including a binding composition comprising an antibody binding site which specifically binds a DC-STAMP or DSP-1.
  • the present invention provides amino acid sequences and DNA sequences encoding various mammalian proteins which are membrane proteins, e.g., which are surface molecules which may mediate a signal between immune or other cells. See, e.g., Paul (1997) Fundamental Immunology (3d ed.) Raven Press, N.Y.
  • the proteins, and fragments, or antagonists will be useful in physiological modulation of cells expressing a receptor or binding partner.
  • DC-STAMP or DSP-1 has either stimulatory or inhibitory effects on hematopoietic cells, including, e.g., lymphoid cells, such as T-cells, B- cells, natural killer (NK) cells, macrophages, dendritic cells, hematopoietic progenitors, mast cells, etc.
  • lymphoid cells such as T-cells, B- cells, natural killer (NK) cells, macrophages, dendritic cells, hematopoietic progenitors, mast cells, etc.
  • NK natural killer
  • the proteins will also be useful as antigens, e.g., immunogens, for raising antibodies to various epitopes on the protein, both linear and conformational epitopes.
  • DC-STAMP Various cDNAs encoding DC-STAMP or DSP-1 were identified.
  • the DC-STAMP was identified from cDNA libraries prepared form human monocyte-derived dendritic cells.
  • the DSP-1 was identified from a cDNA library derived from a human HEL cell line .
  • DC dendritic cells
  • APC professional antigen presenting cells
  • DC are unique in their ability to present antigen to naive T cells, and play therefore a central role in the initiation of immune responses. Characterization of DC specific genes may help to unravel the mechanism underlying their potent antigen presenting capacity.
  • a 2.3 kb messenger RNA is specifically expressed by DC, and not in a panel of other leukocytes or non- hematopoietic cells. In addition, no expression was detected in tissue of several human organs.
  • the transcript encodes an approximately 470 amino acid protein, which is comprised of 7 putative transmembrane domains.
  • This novel protein has been designated Dendritic Cell Specific Transmembrane Protein (DC- STAMP) .
  • DC-STAMP Dendritic Cell Specific Transmembrane Protein
  • Expression of a DC-STAMP-GFP fusion protein in 293 cells indicates that DC-STAMP is expressed at the cell surface. No sequence homology was found with another protein or multimembrane spanning receptor.
  • DC-STAMP appears to be a novel DC-specific multimembrane spanning protein, representing a new group of transmembrane proteins .
  • cDNA libraries were prepared from human monocyte-derived dendritic cells (DC) and over 250 cDNA clones were characterized by nucleotide sequence analysis. See Marland, et al . (1997) in Ricciardi- Castognoli (ed.) Dendritic Cells in Fundamental and Clinical Immunology. Vol 3, Plenum Publ . Corp.
  • One of these cDNA clones was analyzed in further detail as it contained a unique sequence not present in the GenBank databases and its partial open reading frame (ORF) appeared to encode a putative transmembrane (TM) region.
  • the human DC-STAMP gene will encode a membrane protein, of about 470 amino acids. See Table 1 and SEQ. ID. NO: 1 and 2.
  • DC-STAMP exhibits structural motifs characteristic of a member of multiple membrane spanning proteins, e.g., 7 transmembrane receptors. Other notable motifs or features include asnl68- thrl70, asnl87-serl88, and asn357-ser359 (three predicted N- linked glycosylation sites) ; and thr286-lys288 (potential site for phosphorylation by PKC) ; and Iys426-ser429 and arg438- ser441 (potential sites for cAMP-dependent protein kinases) .
  • the human DSP-1 appears to exist in two forms.
  • One form designated the long form, encodes a membrane protein of about 313 amino acids
  • the other designated the short form, encodes a membrane protein of about 200 amino acids.
  • the short form seems to result from deletion of nucleotides 94-433 of the long form, and the corresponding amino acids of the protein.
  • Both forms seem to encode type I membrane proteins, with the transmembrane segment corresponding to long form residues about Ieul72-glyl88.
  • Other notable motifs or features include three ITIM motifs, corresponding to long form residues Ieu222-leu227 , val244-val251, and Ieu258-val263. See, e.g., Thomas (1995) J ⁇ _ Ex . Med.
  • the Immunoreceptor Tyrosine-based Inhibitory Motifs recruit intracellular tyrosine phosphatases, and the receptors provide an inhibitory signal to the cell.
  • the DSP-1 antigen is involved in a negative regulatory signaling pathway in the expressing cells, e.g., monocytes, T, NK, and/or mast cells.
  • the binding partner probably a surface receptor or soluble ligand, might inhibit monocyte, T, NK, and/or mast cell degranulation, chemotaxis, or signaling.
  • Table 1 Primate, e.g., human, DC-STAMP (SEQ ID NO: 1 and 2): gggggg ggc atttctgcat tcgaagaaga atctgagaga aacctgacgc agggagc 57 atg ggt ate tgg ace tea ggc act gat ate ttc eta agt ctt tgg gag 105 Met Gly He Trp Thr Ser Gly Thr Asp He Phe Leu Ser Leu Trp Glu 1 5 10 15 att tac gtg tct cca aga age ccc gga tgg atg gac ttt ate cag cat 153 He Tyr Val Ser Pro Arg Ser Pro Gly Trp Met Asp Phe He Gin His
  • 385 390 395 400 aaa ate ctg gtg tea gca tct ttc tac ccc age gtg gag agg aag cgc 1305
  • nucleic acids encoding polypeptides can be A, C, G, or T; SEQ ID NO: 3) : ATGGGNATHTGGACNWSNGGNACNGAYATHTTYYTNWSNYTNTGGGARATHTAYGTNWSNCCNMGNWSNCCNGG NTGGATGGAYTTYATHCARCAYYTNGGNGTNTGYTGYYTNGTNGCNYTNATHWSNGTNGGNYTNYTN SNGTNG CNGCNTGYTGGTTYYTNCCNWSNATHATHGCNGCNGCNGCNWSNTGGATHATHACNTGYGTNYTNYTNTGYTGY WSNAARCAYGCNMGNTGYTTYATHYTNYTNGTNTTYYTNWSNTGYGGNYTNMGNGARGGNMGNAAYGCNYTNAT HGCNGCNGGNACNGGNATHGTNATHYTNGGNCAYGTNGARAAYATHTTYCAYAAYTTYAARGGNYTNYTNGAYG GNATGACNTGYAAYYTNMGNGCNAAR
  • DSP-1L (SEQ ID NO: 4 and 5) : atg gcc tta cca gtg ace gcc ttg etc ctg ccg eta gcc ttg ctg etc 48 Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu Leu -20 -15 -10 cac gcc gcc agg ccg gat tac aag gac gat gac gac aag ate gat ctg 96 His Ala Ala Arg Pro Asp Tyr Lys Asp Asp Asp Asp Lys He Asp Leu -5 -1 1 5 10 age aaa tgc agg ace gtg gcg ggc ccc gtg ggg gga tec ctg agt gtg 144 Ser Lys Cys Arg Thr Val Ala G
  • DSP-IS form (SEQ ID NO: 6 and 7) : atg gcc tta cca gtg ace gcc ttg etc ctg ccg eta gcc ttg ctg etc 48 Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu Leu -20 -15 -10 cac gcc gcc agg ccg gat tac aag gac gat gac gac aag ate gat atg 96 His Ala Ala Arg Pro Asp Tyr Lys Asp Asp Asp Asp Lys He Asp Met -5 -1 1 5 10 aca cct gca agt ate act gcg gcc aag ace tea aca ate aca act gca 144 Thr Pro Ala Ser He Thr Ala Ala Lys Thr Ser Thr He Thr
  • DSP-1L 1 MALPVTALLLPLALLLHAARPDYKDDDDKIDLSKCRTVAGPVGGSLSVQC 50 DSP-IS 1 MALPVTALLLPLALLLHAARPDYKDDDDKID 31
  • DSP-1L 51 PYEKEHRTLNKYWCRPPQIFLCDKIVETKGSAGKRNGRVSIRDSPANLSF 100 DSP-IS 32 3i
  • DSP-1L 101 TVTLENLTEEDAGTY CGVDTPWLRDFHDPWEVEVSVFPASTSMTPASI 150
  • DSP-IS 38 TAAKTSTITTAFPPVSSTTLFAVGATHSASIQEETEEWNSQLPLLLSLL 87**************************************
  • N can be A, C, G, or T
  • 5 long SEQ ID NO: 8
  • N may be A, C, G, or T:
  • N may be A, C, G, or T:
  • GPCRs are found in a wide range of organisms and are typically involved in the transmission of signals to the interior of the cell, e.g., through interaction, e.g., with heterotrimeric G-proteins. They respond to a wide and diverse range of agents including lipid analogs, amino acid derivatives, small peptides, and other molecules.
  • a predicted model of the structure of the DC-STAMP protein has an extracellular N-terminus, a cytoplasmic C-terminus, 3 cytoplasmic loops, and 3 extracellular loops, containing 2 consensus sequences for N-linked glycosylation on the second and one on the third extracellular loop.
  • the serine residues in the C-terminus of DC-STAMP are putative targets for phosphorylation.
  • TM proteins it has been shown that phosphorylation of serine and threonine residues in the C-tail of the receptor by G protein coupled receptor kinases results in uncoupling of the activated receptor from its G proteins, thereby desensitizing the receptor. See B ⁇ hm, et al . (1997) J. Biol. Chem. 332:1-18. Further experiments can be performed to determine whether the DC-STAMP protein can be phosphorylated at these serine residues.
  • TM proteins include a signature of cysteine residues in the first two extracellular loops, which might form disulphide bridges and stabilize the protein structure. See Savarese and Fraser (1992) J. Biol. Chem. 283:1-19. Also, cysteine residues in the carboxyl tails are potential sites for palmitoylation and may serve to form a fourth intracellular loop. See O'Dowd et al . (1989) J. Biol. Chem. 264:7564-7569; and Strader, et al . (1994) Ann. Re . Biochem. 63:101-132.
  • the DC-STAMP protein contains an alternative signature of cysteines in TM1 and TM2 , and has no cysteine residues in its C-terminus. Combined with the absence of any sequence homology to 7 TM receptors, the described characteristics of the DC-STAMP protein suggest that this novel protein does not belong to any of the existing 7 TM subclasses. DC-STAMP could either form a novel 7 TM protein subclass or be the first member of a new family of multi-membrane spanning proteins .
  • Characteristic of the DC-STAMP terminus is its very basic amino acid composition. There is some indication that juxtamembrane clusters of positively charged residues in cytoplasmic receptor tails can associate with proteins of the ERM (ezrin, radaxin, moesin) family. See Bretscher (1999) Curr. Op. Cell Biol. 11:109-116. Since these ERM proteins have been implicated as membrane cytoskeletal linkers, this might suggest association of DC-STAMP with the cytoskeleton. Ligation of DC-STAMP might affect adhesive or migratory capacities, essential for proper DC function. These can be tested.
  • the position of potential glycosylation sites, putative phosphorylation recognition sites and the intracellular C-terminus favor this model.
  • the model supports a type IIlb integral membrane protein, with the N-terminus of DC-STAMP outside and the C-terminus on the luminal side of the membrane.
  • the DC-STAMP protein contains a proline residue between TMl and TM2. Prolines are known to disrupt helices and the proline residue at position 56 in DC-STAMP could help to establish a loop and redirect the protein into the membrane. This could possibly compensate for the rather short hydrophobic stretches of TMl (transmembrane region 1) and TM2, 17 and 18 amino acids in length, respectively. Also, both TMl and TM2 contain a pair of cysteine residues, which could further stabilize this intramembrane loop by a disulphide bridge near the outer membrane side.
  • TM2 , TM3 , and TM4 are rather weak TM regions, alternative models cannot be excluded comprising 5 or 4 TM regions, in which TMl and TM2 form a single transmembrane domain and either TM3 or TM4 or both are not present .
  • TMl and TM2 form a single transmembrane domain and either TM3 or TM4 or both are not present .
  • Only two 5 TM spanning proteins have been described so far, the 865 amino acid AC133 orphan receptor, expressed by hematopoietic stem cells (Miraglia, et al . (1997) Blood 90:5013-5021), and the CD47 molecule (Lindberg, et al . (1993) J. Cell. Biol.
  • the TM4 superfamily consists of nearly 20 genes, encoding proteins which are thought to be involved in the grouping and stabilization of cell-surface proteins.
  • the DC- STAMP protein does not show significant homology to either these TM4 or TM5 proteins, indicating that DC-STAMP represents a novel protein family.
  • Transmembrane segments are typically 20-25 amino acids in length. Based upon models and data on bacteriorhodopsin, these regions are predicted to be ⁇ -helices and be oriented to form a ligand binding pocket. See, e.g., Findley, et al . (1990) Trends Pharmacol. Sci. 11:492-499.
  • 7 TM receptors comprise a family of very heterogeneous proteins that signal through heterotrimeric G proteins (Strader, et al . (1994) Ann. Rev. Biochem. 63:101-132), including chemokine, hormone and photoreceptors .
  • the vast majority of 7 TM proteins are G-protein coupled.
  • the presence of an aspartate in the second transmembrane region and a so- called "DRY or ERY motif", closely following the third transmembrane region, are both thought to be involved in the signal transduction via G-proteins. See Savarese and Fraser (1992) J. Biol. Chem. 283:1-19; and Bourne (1997) Curr. Op . Cell Biol. 9:134-142.
  • DC-STAMP contains neither of these sequences, but such motifs may possibly not be recognized as such.
  • other 7 TM proteins such as the Duffy antigen receptor (DARC) on erythrocytes and the EGF-7TM receptors, also lack consensus sequences for G protein coupling and no signal transduction via these receptors has been proven as yet. See Horuk, et al . (1996) J. Leukoc . Biol. 59:29-38; and McKnight and Gordon (1996) Immunol . Today 17:283-287. Although the full spectrum of biological activities mediated by these 7 transmembrane receptors has not been fully determined, chemoattractant effects are recognized. Chemokine receptors are notable members of the GPCR family.
  • chemokine molecules relate to chemoattraction of leukocytes.
  • new chemokines and receptors are being discovered, and their biological effects on the various cells responsible for immunological responses are topics of continued study.
  • DC-STAMP agonists, or antagonists may also act as functional or receptor antagonists, e.g., which block DC interactions or physiology, or mediating the opposite actions.
  • DC are implicated in T cell mediated immunity, which is important in various diseases. T cell immunity is deficient in various contexts, e.g., in tumor immunotherapy and allergic responses. Conversely, it is overactive in autoimmune diseases and transplantation rejection contexts.
  • DC-STAMP, or its antagonists may be useful in the treatment of abnormal medical conditions, including immune disorders, e.g., immune deficiencies, chronic inflammation, or tissue rejection, or other physiological conditions.
  • the implication of antigen presentation in initiation of an immune response is a likely condition to be affected by the use of a DC-STAMP related reagent.
  • compositions combining the DC-STAMP and other DC affecting reagents will often be used. See below.
  • the DSP-1 forms are highly expressed in mast cells, which are implicated in allergic responses, particularly in release of histamine. See, e.g., Kaliner and Metcalfe (eds. 1992) The Mast Cell in Health and Disease. Reagents related to activation or deactivation of DSP-1 signaling may be important in medical conditions mediated by cells expressing the antigen.
  • Both the long (L) and short (s) forms are type I membrane proteins, and possess cytoplasmic domains with multiple ITIM motifs, suggesting an inhibitory receptor signaling role. See, e.g., Kung, et al . (1999) J . Immunol .
  • transmembrane segments correspond approximately to residues 172-188 (166-198) for the L form, and 59-75 (53-85) of the S form. However the actual boundaries of transmembrane segments may vary or depend upon kinetic and other factors.
  • the preferred embodiments characterized herein are from primate, e.g., human, but other species counterparts will exist in nature. Additional sequences for proteins in other mammalian species, e.g., primates, canines, felines, and rodents, should also be available, particularly the domestic animal species. See below. The descriptions below are directed, for exemplary purposes, to a human DC-STAMP or DSP-1, but are likewise applicable to related embodiments from other species.
  • Primate e.g., human, DC-STAMP or DSP-1 amino acid sequences
  • Tables 1 or 2 Other naturally occurring nucleic acids which encode the proteins can be isolated by standard procedures using the provided sequences, e.g., PCR techniques, or by hybridization.
  • Primer extension or RACE methods can extend to adjacent sequence, either on message or genomic. These amino acid sequences, provided amino to carboxy, are important in providing sequence information for the proteins allowing for distinguishing the protein antigen from other proteins and exemplifying numerous variants.
  • peptide sequences allow preparation of peptides to generate antibodies to recognize such segments
  • nucleotide sequences allow preparation of oligonucleotide probes, both of which are strategies for detection or isolation, e.g., cloning, of genes encoding such sequences.
  • human DC-STAMP shall encompass, when used in a protein context, a protein having amino acid sequence corresponding to a polypeptide shown in SEQ ID NO : 2, or significant fragments thereof.
  • Preferred embodiments comprise a plurality of distinct, e.g., nonoverlapping, segments of the specified length. Typically, the plurality will be at least two, more usually at least three, and preferably 5, 7, or even more. While the length minima are provided, longer lengths, of various sizes, may be appropriate, e.g., one of length 7, and two of length 12. Similarly with the term DSP-1 and SEQ ID NO: 4 and 6.
  • Binding components typically bind to an antigen with high affinity, e.g., at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably at better than about 3 nM.
  • polypeptide as used herein includes a significant fragment or segment, and encompasses a stretch of amino acid residues of at least about 8 amino acids, generally at least about 12 amino acids, typically at least about 16 amino acids, preferably at least about 20 amino acids, and, in particularly preferred embodiments, at least about 30 or more amino acids, e.g., 35, 40, 45, 50, etc.
  • Such fragments may have ends which begin and/or end at virtually all positions, e.g., beginning at residues 1, 2, 3, etc., and ending at, e.g., 150, 149, 148, etc., in all practical combinations.
  • Particularly interesting peptides have ends corresponding to structural domain boundaries, e.g., transmembrane segments or identified motifs. See Tables 1 and 2.
  • binding composition refers to molecules that bind with specificity to DC-STAMP or DSP-1, e.g., in an antibody-antigen interaction.
  • the specificity may be more or less inclusive, e.g., specific to a particular embodiment, or to groups of related embodiments, e.g., primate, rodent, etc. It also includes compounds, e.g., proteins, which specifically associate with DC-STAMP, including in a natural physiologically relevant protein-protein interaction, either covalent or non- covalent.
  • the molecule may be a polymer, or chemical reagent.
  • a functional analog may be a protein with structural modifications, or it may be a molecule which has a molecular shape which interacts with the appropriate binding determinants.
  • the compounds may serve as agonists or antagonists of a receptor binding interaction, see, e.g., Goodman, et al . (eds.) Goodman & Gilman's: The Pharmacological Bases of Therapeutics (current ed.) Pergamon Press.
  • Substantially pure typically means that the protein is free from other contaminating proteins, nucleic acids, or other biologicals derived from the original source organism. Purity may be assayed by standard methods, typically by weight, and will ordinarily be at least about 40% pure, generally at least about 50% pure, often at least about 60% pure, typically at least about 80% pure, preferably at least about 90% pure, and in most preferred embodiments, at least about 95% pure. Carriers or excipients will often be added.
  • Solubility of a polypeptide or fragment depends upon the environment and the polypeptide. Many parameters affect polypeptide solubility, including temperature, electrolyte environment, size and molecular characteristics of the polypeptide, and nature of the solvent. Typically, the temperature at which the polypeptide is used ranges from about 4° C to about 65° C. Usually the temperature at use is greater than about 18° C. For diagnostic purposes, the temperature will usually be about room temperature or warmer, but less than the denaturation temperature of components in the assay. For therapeutic purposes, the temperature will usually be body temperature, typically about 37° C for humans and mice, though under certain situations the temperature may be raised or lowered in situ or in vitro.
  • the size and structure of the polypeptide should generally be in a substantially stable state, and usually not in a denatured state.
  • the polypeptide may be associated with other polypeptides in a quaternary structure, e.g., to confer solubility, or associated with lipids or detergents.
  • the solvent and electrolytes will usually be a biologically compatible buffer, of a type used for preservation of biological activities, and will usually approximate a physiological aqueous solvent. Usually the solvent will have a neutral pH, typically between about 5 and 10, and preferably about 7.5.
  • one or more detergents will be added, typically a mild non-denaturing one, e.g., CHS (cholesteryl hemisuccinate) or CHAPS (3- [3- cholamidopropyl) dimethylammonio] -1-propane sulfonate), or a low enough concentration as to avoid significant disruption of structural or physiological properties of the protein.
  • CHS cholesteryl hemisuccinate
  • CHAPS 3- [3- cholamidopropyl) dimethylammonio] -1-propane sulfonate
  • a harsh detergent may be used to effect significant denaturation.
  • This invention also encompasses proteins or peptides having substantial amino acid sequence identity with the amino acid sequences of the DC-STAMP or DSP-1 antigens.
  • the variants include species, polymorphic, or allelic variants.
  • Amino acid sequence homology, or sequence identity is determined by optimizing residue matches, if necessary, by introducing gaps as required. See also Needleham, et al .
  • Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • the conservation may apply to biological features, functional features, or structural features.
  • Homologous amino acid sequences are typically intended to include natural polymorphic or allelic and interspecies variations of a protein sequence. Typical homologous proteins or peptides will have from 25-100% identity (if gaps can be introduced) , to 50-100% identity (if conservative substitutions are included) with the amino acid sequences of the antigens.
  • Identity measures will be at least about 35%, generally at least about 40%, often at least about 50%, typically at least about 60%, usually at least about 70%, preferably at least about 80%, and more preferably at least about 90%.
  • the isolated DC-STAMP or DSP-1 DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of short nucleotide stretches. These modifications may result in novel DNA sequences which encode these antigens, their derivatives, or proteins having similar physiological, immunogenic, antigenic, or other functional activity. These modified sequences can be used to produce mutant antigens or to enhance expression. Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms.
  • “Mutant DC-STAMP” encompasses a polypeptide otherwise falling within the sequence identity definition of the DC-STAMP as set forth above, but having an amino acid sequence which differs from that of the antigen as normally found in nature, whether by way of deletion, substitution, or insertion. This generally includes proteins having significant identity with a protein having sequence of SEQ ID NO: 2, and as sharing various biological activities, e.g., antigenic or immunogenic, with those sequences, and in preferred embodiments contain most of the natural full length disclosed sequences. Full length sequences will typically be preferred, though truncated versions will also be useful, likewise, genes or proteins found from natural sources are typically most desired.
  • DC-STAMP or DSP-1 mutagenesis can also be conducted by making amino acid insertions or deletions. Substitutions, deletions, insertions, or any combinations may be generated to arrive at a final construct. Insertions include amino- or carboxy- terminal fusions. Random mutagenesis can be conducted at a target codon and the expressed mutants can then be screened for the desired activity.
  • substitution mutations at predetermined sites in DNA having a known sequence are well known in the art, e.g., by M13 primer mutagenesis or polymerase chain reaction (PCR) techniques. See, e.g., Sambrook, et al . (1989); Ausubel, et al . (1987 and Supplements); and Kunkel, et al . (1987) Methods in Enzymol . 154:367-382.
  • Preferred embodiments include, e.g., 1-fold, 2- fold, 3-fold, 5-fold, 7-fold, etc., preferably conservative substitutions at the nucleotide or amino acid levels.
  • the substitutions will be away from the conserved cysteines, and often will be in the regions away from the extramembrane domains. Such variants may be useful to produce specific antibodies, and often will share many or all biological properties.
  • the present invention also provides recombinant proteins, e.g., heterologous fusion proteins using segments from these proteins.
  • a heterologous fusion protein is a fusion of proteins or segments which are naturally not normally fused in the same manner.
  • a similar concept applies to heterologous nucleic acid sequences.
  • new constructs may be made from combining similar functional domains from other proteins.
  • target-binding or other segments may be "swapped" between different new fusion polypeptides or fragments. See, e.g., Cunningham, et al . (1989) Science 243:1330-1336; and O'Dowd, et al. (1988) J. Biol. Chem. 263:15985-15992.
  • a double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence, e.g., PCR techniques .
  • Structural analysis can be applied to this gene, in comparison to members of related gene families, e.g., GPCRs .
  • ⁇ -sheet and ⁇ -helix residues can be determined using, e.g., RASMOL program, see Bazan, et al . (1996) Nature 379:591; Lodi, et al .
  • residues for substitutions include the surface exposed extramembrane residues which would be predicted to interact with a counterstructure or ligand. Other residues which should conserve function will be conservative substitutions, particularly at a position far from the surface exposed residues, e.g., an intramembrane residue.
  • the blocking of physiological response to DC-STAMP or DSP- 1 may result from the competitive inhibition of binding of a ligand or counterstructure to the antigen.
  • DC-STAMP or DSP-1 antigens include amino acid sequence mutants from naturally occurring forms, glycosylation variants, and covalent or aggregate conjugates with other chemical moieties.
  • Covalent derivatives can be prepared by linkage of functionalities to groups which are found in amino acid side chains or at the N- or C- termini, e.g., by standard means. See, e.g., Lundblad and Noyes (1988) Chemical Reagents for Protein Modification, vols. 1-2, CRC Press, Inc., Boca Raton, FL; Hugli (ed.
  • glycosylation alterations are included, e.g., made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps. See, e.g., Elbein (1987) Ann. Rev. Biochem. 56:497-534. Also embraced are versions of the peptides with the same primary amino acid sequence which have other minor modifications, including phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine .
  • Fusion polypeptides between DC-STAMP or DSP-1 and other homologous or heterologous proteins are also provided.
  • Many 7TM receptors or other surface proteins are multimeric, e.g., homodimeric entities, and a repeat construct may have various advantages, including lessened susceptibility to proteolytic cleavage.
  • Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a protein, e.g., a receptor-binding segment, so that the presence or location of the fused ligand may be easily determined. See, e.g., Dull, et al . , U.S. Patent No. 4,859,609.
  • gene fusion partners include bacterial ⁇ -galactosidase, trpE, Protein A, ⁇ -lactamase, alpha amylase, alcohol dehydrogenase, yeast alpha mating factor, and detection or purification tags such as a FLAG sequence of His6 sequence. See, e.g., Godowski , et al. (1988) Science 241:812-816.
  • Fusion peptides will typically be made by either recombinant nucleic acid methods or by synthetic polypeptide methods. Techniques for nucleic acid manipulation and expression are described generally, e.g., in Sambrook, et al . (1989) Molecular Cloning: A Laboratory Manual (2d ed.), vols. 1-3, Cold Spring Harbor Laboratory; and Ausubel, et al . (eds. 1993) Current Protocols in Molecular Biology. Greene and Wiley, NY. Techniques for synthesis of polypeptides are described, e.g., in Merrifield (1963) J. Amer. Chem. Soc. 85:2149-2156; Merrifield (1986) Science 232: 341-347; Atherton, et al .
  • a DC-STAMP or DSP-1 can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated SEPHAROSE, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in the assay or purification of antibodies or an alternative binding composition.
  • a solid support such as cyanogen bromide-activated SEPHAROSE
  • These proteins can also be labeled with a detectable group, e.g., for use in diagnostic assays.
  • Purification of antigen may be effected by an immobilized antibody or complementary binding partner, e.g., binding portion of a receptor.
  • a solubilized fragment of this invention can be used as an immunogen for the production of antisera or antibodies specific for binding.
  • Purified antigen can be used to screen monoclonal antibodies or antigen-binding fragments, encompassing antigen binding fragments of natural antibodies, e.g., Fab, Fab', F(ab)2, etc.
  • Purified DC-STAMP or DSP-1 antigens can also be used as a reagent to detect antibodies generated in response to the presence of elevated levels of the antigen, which may be diagnostic of an abnormal or specific physiological or disease condition.
  • This invention contemplates antibodies raised against amino acid sequences encoded by nucleotide sequence shown in SEQ ID NO: 1 or 4 or 6 , or fragments of proteins containing it.
  • this invention contemplates antibodies having binding affinity to or being raised against specific domains, e.g., extracellular segments.
  • the present invention contemplates the isolation of additional closely related species variants. Southern and Northern blot analysis will establish that similar genetic entities exist in other mammals. It is likely that antigens are widespread in species variants, e.g., rodents, lagomorphs, carnivores, artiodactyla, perissodactyla, and primates.
  • the invention also provides means to isolate a group of related antigens displaying both distinctness and similarities in structure, expression, and function. Elucidation of many of the physiological effects of the molecules will be greatly accelerated by the isolation and characterization of additional distinct species or polymorphic variants of them.
  • the present invention provides useful probes for identifying additional homologous genetic entities in different species.
  • the isolated genes will allow transformation of cells lacking expression of DC-STAMP or DSP-1, e.g., either species types or cells which lack corresponding proteins and exhibit negative background activity. This should allow analysis of the function of antigen in comparison to untransformed control cells .
  • Intracellular functions would probably involve receptor signaling. However, protein internalization may occur under certain circumstances, and interaction between intracellular components and ligand or receptor may occur. Specific segments of interaction of membrane antigen with interacting components may be identified by mutagenesis or direct biochemical means, e.g., cross-linking or affinity methods. Structural analysis by crystallographic or other physical methods will also be applicable. Further investigation of the mechanism of signal transduction will include study of associated components which may be isolatable by affinity methods or by genetic means, e.g., complementation analysis of mutants.
  • controlling elements associated with the antigens should exhibit differential physiological, developmental, tissue specific, or other expression patterns. Upstream or downstream genetic regions, e.g., control elements, are of interest .
  • Antibodies can be raised to various epitopes of the membrane proteins, including species, polymorphic, or allelic variants, and fragments thereof, both in their naturally occurring forms and in their recombinant forms. Additionally, antibodies can be raised to the proteins in either their active forms or in their inactive forms, including native or denatured versions. Anti-idiotypic antibodies are also contemplated.
  • Antibodies including binding fragments and single chain versions, against predetermined fragments of the antigens can be raised by immunization of animals with conjugates of the fragments with immunogenic proteins. Monoclonal antibodies are prepared from cells secreting the desired antibody. These antibodies can be screened for binding to normal or defective proteins, or screened for agonistic or antagonistic activity, e.g., mediated through a receptor. Antibodies may be agonistic or antagonistic, e.g., by sterically blocking binding to a receptor.
  • These monoclonal antibodies will usually bind with at least a KJ-J of about 1 mM, more usually at least about 300 ⁇ M, typically at least about 100 ⁇ M, more typically at least about 30 ⁇ M, preferably at least about 10 ⁇ M, and more preferably at least about 3 ⁇ M or better.
  • a DC-STAMP or DSP-1 protein that specifically binds to or that is specifically immunoreactive with an antibody generated against a defined immunogen, such as an immunogen consisting of the amino acid sequence of SEQ ID NO: 2, is typically determined in an immunoassay.
  • the immunoassay typically uses a polyclonal antiserum which was raised, e.g., to a protein of SEQ ID NO: 2. This antiserum is selected to have low crossreactivity against other related proteins, e.g., human or rodent DC-STAMP, preferably from the same species, and any such crossreactivity is removed by immunoabsorption prior to use in the immunoassay.
  • the protein of SEQ ID NO: 2, or a combination thereof is isolated as described herein.
  • recombinant protein may be produced in a mammalian cell line.
  • An appropriate host e.g., an inbred strain of mice such as Balb/c, is immunized with the selected protein, typically using a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane, supra) .
  • a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier protein can be used an immunogen.
  • Polyclonal sera are collected and titered against the immunogen protein in an immunoassay, e.g., a solid phase immunoassay with the immunogen immobilized on a solid support.
  • Polyclonal antisera with a titer of 10 4 or greater are selected and tested for their cross reactivity against other related family members, e.g., rodent DC-STAMP, using a competitive binding immunoassay such as the one described in Harlow and Lane, supra, at pages 570-573.
  • at least one other related family member is used in this determination in conjunction with, e.g., the primate embodiment.
  • the desired target family members can be produced as recombinant proteins and isolated using standard molecular biology and protein chemistry techniques as described herein.
  • Immunoassays in the competitive binding format can be used for the crossreactivity determinations.
  • the protein of SEQ ID NO: 2 can be immobilized to a solid support. Proteins added to the assay compete with the binding of the antisera to the immobilized antigen. The ability of the above proteins to compete with the binding of the antisera to the immobilized protein is compared to the protein of SEQ ID NO: 2. The percent crossreactivity for the above proteins is calculated, using standard calculations. Those antisera with less than 10% crossreactivity with each of the proteins listed above are selected and pooled. The cross-reacting antibodies are then removed from the pooled antisera by immunoabsorption with the above-listed proteins.
  • the immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second protein to the immunogen protein (e.g., the protein of SEQ ID NO: 2) .
  • the two proteins are each assayed at a wide range of concentrations and the amount of each protein required to inhibit 50% of the binding of the antisera to the immobilized protein is determined. If the amount of the second protein required is less than twice the amount of the protein of the selected protein or proteins that is required, then the second protein is said to specifically bind to an antibody generated to the immunogen.
  • the antibodies of this invention can also be useful in diagnostic applications. As capture or non-neutralizing antibodies, they can be screened for ability to bind to the antigens without inhibiting binding to a receptor. As neutralizing antibodies, they can be useful in competitive binding assays. They will also be useful in detecting or quantifying DC-STAMP or DSP-1 protein or their receptors. See, e.g., Chan (ed. 1987) Immunology: A Practical Guide, Academic Press, Orlando, FL; Price and Newman (eds. 1991) Principles and Practice of Immunoassay, Stockton Press, N.Y.; and Ngo (ed. 1988) Nonisotopic Immunoassay, Plenum Press, N.Y. Cross absorptions, depletions, or other means will provide preparations of defined selectivity, e.g., unique or shared species specificities. These may be the basis for tests which will identify various groups of antigens.
  • the antibodies, including antigen binding fragments, of this invention can be potent antagonists that bind to the antigen and inhibit functional binding, e.g., to a receptor which may elicit a biological response. They also can be useful as non-neutralizing antibodies and can be coupled to toxins or radionuclides so that when the antibody binds to antigen, a cell expressing it, e.g., on its surface, is killed. Further, these antibodies can be conjugated to drugs or other therapeutic agents, either directly or indirectly by means of a linker, and may effect drug targeting.
  • Antigen fragments may be joined to other materials, particularly polypeptides, as fused or covalently joined polypeptides to be used as immunogens .
  • An antigen and its fragments may be fused or covalently linked to a variety of immunogens, such as keyhole limpet hemocyanin, bovine serum albumin, tetanus toxoid, etc. See Microbiology, Hoeber Medical Division, Harper and Row, 1969; Landsteiner (1962) Specificity of Serological Reactions, Dover Publications, New York; Williams, et al . (1967) Methods in Immunology and Immunochemistry, vol. 1, Academic Press, New York; and Harlow and Lane (1988) Antibodies: A Laboratory Manual, CSH Press, NY, for descriptions of methods of preparing polyclonal antisera.
  • monoclonal antibodies from various mammalian hosts, such as mice, rodents, primates, humans, etc. Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites, et al .
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents, teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. Also, recombinant immunoglobulins may be produced, see Cabilly, U.S. Patent No. 4,816,567; Moore, et al . , U.S. Patent No.
  • the antibodies of this invention can also be used for affinity chromatography in isolating the protein. Columns can be prepared where the antibodies are linked to a solid support. See, e.g., Wilchek et al . (1984) Meth. Enzymol . 104:3-55. The converse may be used to purify antibodies. Antibodies raised against DC-STAMP or DSP-1 will also be useful to raise anti-idiotypic antibodies. These will be useful in detecting or diagnosing various immunological conditions related to expression of the respective antigens.
  • the described peptide sequences and the related reagents are useful in detecting, isolating, or identifying a DNA clone encoding DC-STAMP or DSP-1, e.g., from a natural source.
  • it will be useful in isolating a gene from mammal, and similar procedures will be applied to isolate genes from other species, e.g., warm blooded animals, such as birds and mammals.
  • Cross hybridization will allow isolation of DC-STAMP from the same, e.g., polymorphic variants, or other species.
  • a number of different approaches will be available to successfully isolate a suitable nucleic acid clone.
  • the purified protein or defined peptides are useful for generating antibodies by standard methods, as described above. Synthetic peptides or purified protein can be presented to an immune system to generate monoclonal or polyclonal antibodies. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene; and Harlow and Lane (1989) Antibodies: A Laboratory Manual , Cold Spring Harbor Press.
  • the specific binding composition could be used for screening of an expression library made from a cell line which expresses a DC-STAMP. Screening of intracellular expression can be performed by various staining or immunofluorescence procedures. Binding compositions could be used to affinity purify or sort out cells expressing a surface fusion protein.
  • the peptide segments can also be used to predict appropriate oligonucleotides to screen a library.
  • the genetic code can be used to select appropriate oligonucleotides useful as probes for screening. See, e.g., SEQ ID NO: 1 or 4 or 6.
  • synthetic oligonucleotides will be useful in selecting correct clones from a library.
  • Complementary sequences will also be used as probes, primers, or antisense strands.
  • Various fragments should be particularly useful, e.g., coupled with anchored vector or poly-A complementary PCR techniques or with complementary DNA of other peptides.
  • This invention contemplates use of isolated DNA or fragments to encode an antigenic or biologically active corresponding polypeptide, particularly lacking the portion coding an untranslated 5' portion of the described sequence.
  • this invention covers isolated or recombinant DNA which encodes a biologically active protein or polypeptide and which is capable of hybridizing under appropriate conditions with the DNA sequences described herein.
  • Said biologically active protein or polypeptide can be an intact antigen, or fragment, and have an amino acid sequence disclosed in, e.g., SEQ ID NO: 2 or 5 or 7 , particularly a mature, secreted polypeptide.
  • this invention covers the use of isolated or recombinant DNA, or fragments thereof, which encode proteins which exhibit high identity to membrane DC-STAMP or DSP-1.
  • the isolated DNA can have the respective regulatory sequences in the 5' and 3' flanks, e.g., promoters, enhancers, poly-A addition signals, and others.
  • expression may be effected by operably linking a coding segment to a heterologous promoter, e.g., by inserting a promoter upstream from an endogenous gene .
  • nucleic acid is a nucleic acid, e.g., an RNA, DNA, or a mixed polymer, which is substantially separated from other components which naturally accompany a native sequence, e.g., ribosomes, polymerases, and/or flanking genomic sequences from the originating species.
  • the term embraces a nucleic acid sequence which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates and chemically synthesized analogs or analogs biologically synthesized by heterologous systems.
  • a substantially pure molecule includes isolated forms of the molecule.
  • the nucleic acid will be in a vector or fragment less than about 50 kb, usually less than about 30 kb, typically less than about 10 kb, and preferably less than about 6 kb.
  • An isolated nucleic acid will generally be a homogeneous composition of molecules, but will, in some embodiments, contain minor heterogeneity. This heterogeneity is typically found at the polymer ends or portions not critical to a desired biological function or activity.
  • a "recombinant" nucleic acid is defined either by its method of production or its structure. In reference to its method of production, e.g., a product made by a process, the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence, typically selection or production. Alternatively, it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants.
  • nucleic acids comprising sequence derived using any synthetic oligonucleotide process. Such is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a sequence recognition site. Alternatively, it is performed to join together nucleic acid segments of desired functions to generate a single genetic entity comprising a desired combination of functions not found in the commonly available natural forms. Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site specific targets, e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design.
  • a similar concept is intended for a recombinant, e.g., fusion, polypeptide.
  • synthetic nucleic acids which, by genetic code redundancy, encode polypeptides similar to fragments of these antigens, and fusions of sequences from various different species or polymorphic variants.
  • a significant "fragment" in a nucleic acid context is a contiguous segment of at least about 17 nucleotides, generally at least about 22 nucleotides, ordinarily at least about 29 nucleotides, more often at least about 35 nucleotides, typically at least about 41 nucleotides, usually at least about 47 nucleotides, preferably at least about 55 nucleotides, and in particularly preferred embodiments will be at least about 60 or more nucleotides, e.g., 67, 73, 81, 89, 95, etc.
  • a DNA which codes for a DC-STAMP or DSP-1 protein will be particularly useful to identify genes, mRNA, and cDNA species which code for related or similar proteins, as well as DNAs which code for homologous proteins from different species. There will be homologs in other species, including primates, rodents, canines, felines, birds, and fish. Various DC-STAMP or DSP-1 proteins should be homologous and are encompassed herein. However, even proteins that have a more distant evolutionary relationship to the antigen can readily be isolated under appropriate conditions using these sequences if they are sufficiently homologous. Primate membrane proteins are of particular interest.
  • Recombinant clones derived from the genomic sequences, e.g., containing introns, will be useful for transgenic studies, including, e.g., transgenic cells and organisms, and for gene therapy. See, e.g., Goodnow (1992) "Transgenic Animals” in Roitt (ed.) Encyclopedia of Immunology. Academic Press, San Diego, pp. 1502-1504; Travis (1992) Science 256:1392-1394; Kuhn, et al . (1991) Science 254:707-710; Capecchi (1989) Science 244:1288; Robertson (ed. 1987) Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, IRL Press, Oxford; Rosenberg (1992) J.
  • expression may be effected by operably linking a coding segment to a heterologous promoter, e.g., by inserting a promoter upstream from an endogenous gene. See, e.g., Treco, et al . W096/29411 or USSN 08/406,030.
  • Substantial homology in the nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 50% of the nucleotides, generally at least about 58%, ordinarily at least about 65%, often at least about 71%, typically at least about 77%, usually at least about 85%, preferably at least about 95 to 98% or more, and in particular embodiments, as high as about 99% or more of the nucleotides.
  • substantial homology exists when the segments will hybridize under selective hybridization conditions, to a strand, or its complement, typically using a sequence of DC-STAMP or DSP-1, e.g., in SEQ ID NO: 1 or 4 or 6.
  • selective hybridization will occur when there is at least about 55% identity over a stretch of at least about 30 nucleotides, preferably at least about 75% over a stretch of about 25 nucleotides, and most preferably at least about 90% over about 20 nucleotides. See, Kanehisa (1984) Nuc . Acids Res . 12:203-213.
  • the length of identity comparison may be over longer stretches, and in certain embodiments will be over a stretch of at least about 17 nucleotides, usually at least about 28 nucleotides, typically at least about 40 nucleotides, and preferably at least about 75 to 100 or more nucleotides.
  • Stringent conditions in referring to homology in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters, typically those controlled in hybridization reactions.
  • Stringent temperature conditions will usually include temperatures in excess of about 30° C, usually in excess of about 37° C, typically in excess of about 55° C, 60° C, or 65° C, and preferably in excess of about 70° C.
  • Stringent salt conditions will ordinarily be less than about 1000 mM, usually less than about 400 mM, typically less than about 250 mM, preferably less than about 150 mM, including about 100, 50, or even 20 mM. However, the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wetmur and Davidson (1968) J. Mol. Biol. 31:349-370.
  • Hybridization under stringent conditions should give a background of at least 2-fold over background, preferably at least 3-5 or more.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence (s) relative to the reference sequence, based on the designated program parameters.
  • Optical alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. Appl . Math. 2:482, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol . 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc.
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendrogram showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol . Evol . 35:351-360.
  • the method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5:151-153.
  • the program can align up to 300 sequences, each of a maximum length of 5,000 nucleotides or amino acids.
  • the multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences. This cluster is then aligned to the next most related sequence or cluster of aligned sequences. Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments.
  • the program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison and by designating the program parameters. For example, a reference sequence can be compared to other test sequences to determine the percent sequence identity relationship using the following parameters: default gap weight (3.00), default gap length weight (0.10), and weighted end gaps .
  • HSPs high scoring sequence pairs
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat ' 1 Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test 5 nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • L0 polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by
  • DC-STAMP or DSP-1 from other mammalian species can be 0 cloned and isolated by cross-species hybridization of closely related species. Homology may be relatively low between distantly related species, and thus hybridization of relatively closely related species is advisable. Alternatively, preparation of an antibody preparation which exhibits less 5 species specificity may be useful in expression cloning approaches .
  • DNA which encodes the DC-STAMP or DSP-1 or fragments 0 thereof can be obtained by chemical synthesis, screening cDNA libraries, or screening genomic libraries prepared from a wide variety of cell lines or tissue samples. See, e.g., Okayama and Berg (1982) Mol. Cell. Biol. 2:161-170; Gubler and Hoffman (1983) Gene 25:263-269; and Glover (ed. 1984) DNA Cloning: A 5 Practical Approach, IRL Press, Oxford.
  • the sequences provided herein provide useful PCR primers or allow synthetic or other preparation of suitable genes encoding a DC- STAMP or DSP-1; including naturally occurring embodiments.
  • This DNA can be expressed in a wide variety of host cells for the synthesis of a full-length DC-STAMP or DSP-1 or 5 fragments which can in turn, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified molecules; and for structure/function studies.
  • Vectors as used herein, comprise plasmids, viruses,
  • DNA sequences are operably linked when they are functionally related to each other.
  • DNA for a presequence or secretory leader is operably linked to a polypeptide if it is expressed as a preprotein or
  • a promoter is operably linked to a coding sequence if it controls the transcription of the polypeptide; a ribosome binding site is operably linked to a coding sequence if it is positioned to permit translation.
  • operably linked means contiguous and in reading frame, however, certain genetic elements such as repressor genes are not contiguously linked but still bind to operator sequences that in turn control expression. See, e.g., Rodriguez, et al . , Chapter 10, pp. 205-236; Balbas and Bolivar (1990) Methods in 0 Enzymology 185:14-37; and Ausubel, et al . (1993) Current Protocols in Molecular Biology, Greene and Wiley, NY.
  • Suitable expression vectors include pCDNAl ; pCD, see Okayama, et al . (1985) Mol. Cell Biol. 5:1136-1142; pMClneo Poly-A, see Thomas, et al . (1987) Cell 5 51:503-512; and a baculovirus vector such as pAC 373 or pAC
  • the DC-STAMP or DSP-1, or a fragment thereof may be engineered to be phosphatidyl inositol (PI) linked to a cell membrane, but can be removed from membranes by treatment with a phosphatidyl inositol cleaving enzyme, e.g., phosphatidyl inositol phospholipase-C.
  • PI phosphatidyl inositol
  • a phosphatidyl inositol cleaving enzyme e.g., phosphatidyl inositol phospholipase-C.
  • DC-STAMP or DSP-1 has been characterized, fragments or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis, Pierce Chemical Co.,
  • the present invention provides reagents which will find use in diagnostic applications as described elsewhere herein, e.g., in DC, T, NK, monocyte, or mast cell mediated conditions, or below in the description of kits for diagnosis.
  • the gene may be useful in forensic sciences, e.g., to distinguish rodent from human, or as a marker to distinguish between different cells exhibiting differential expression or modification patterns .
  • This invention also provides reagents with significant commercial and/or therapeutic potential.
  • the DC-STAMP or DSP-1 naturally occurring or recombinant
  • fragments thereof, and antibodies thereto, along with compounds identified as having binding affinity to DC-STAMP or DSP-1 should be useful as reagents for teaching techniques of molecular biology, immunology, or physiology.
  • kits may be prepared with the reagents, e.g., in practical laboratory exercises in production or use of proteins, antibodies, cloning methods, histology, etc.
  • the reagents will also be useful in the treatment of conditions associated with abnormal physiology or development, including immunological conditions. They may be useful in vitro tests for presence or absence of interacting components, which may correlate with success of particular treatment strategies.
  • modulation of physiology of various, e.g., hematopoietic or lymphoid, cells will be achieved by appropriate methods for treatment using the compositions provided herein. See, e.g., Thomson (ed.
  • a disease or disorder associated with abnormal expression or abnormal signaling by a DC-STAMP should be a likely target for an agonist or antagonist.
  • the new membrane proteins should play a role in regulation or development of hematopoietic cells, e.g., lymphoid cells, which affect immunological responses, e.g., inflammation and/or autoimmune disorders. Alternatively, it may affect vascular physiology or development, or neuronal effects.
  • DC-STAMP is likely to be important in mediating DC function.
  • DC are the professional antigen presenting cells to T and B cells, and should be important in T cell mediated immune responses. Increases in T cell immunity should be important in, e.g., tumor immunotherapy, allergic conditions, and vaccine adjuvants.
  • Important tumors include, e.g., carcinomas, including lung, colon, prostate, and breast, and melanomas. See, e.g., Bertino, et al . (eds. 1996) Encyclopedia of Cancer Academic Press; Devita, et al . (eds. 1997) Cancer: Principles & Practice of Oncology Lippincott, Williams and Wilkins; Devita (1997) Principles and Practice of Oncology Lippincott Williams and Wilkins; Cavalli, et al .
  • Surgical Oncology Current Concepts and Practice Butterworth- Heinemann.
  • allergic conditions e.g., where a shift from Th2 humoral responses to Thl cellular responses may be indicated, include asthma, pollen rhinitis, medicament allergies, food allergies, house dust mite allergies, etc.
  • Combination treatments might be used, combining a therapeutic related to the DC-STAMP or DSP-1 signaling with another therapeutic used to treat symptoms of the conditions, e.g., with Flt3 ligand, G, CSF, radiation or chemotherapy, antihistamines, IL-10, Tregl cells, cyclosporin, or interferons .
  • the DSP-1 therapeutic reagents may be useful to modulate function of monocyte, T, NK, or mast cell mediated conditions. It should be useful as a mast cell marker, being present on those cells, and likely modulates signaling with or by those cells.
  • DC-STAMP or DSP-1, antagonists, antibodies, etc. can be purified and then administered to a patient, veterinary or human.
  • These reagents can be combined for therapeutic use with additional active or inert ingredients, e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, along with physiologically innocuous stabilizers, excipients, or preservatives.
  • additional active or inert ingredients e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, along with physiologically innocuous stabilizers, excipients, or preservatives.
  • These combinations can be sterile filtered and placed into dosage forms as by lyophilization in dosage vials or storage in stabilized aqueous preparations.
  • This invention also contemplates use of antibodies or binding fragments thereof, including forms which are not complement binding .
  • Drug screening using DC-STAMP or DSP-1 or fragments thereof can be performed to identify compounds having binding affinity to or other relevant biological effects on DC-STAMP or DSP-1 functions, including isolation of associated components. Subsequent biological assays can then be utilized to determine if the compound has intrinsic stimulating activity and is therefore a blocker or antagonist in that it blocks the activity of DC-STAMP or DSP-1 signaling. Likewise, a compound having intrinsic stimulating activity can activate the signal pathway and is thus an agonist in that it simulates the activity of DC-STAMP or DSP-1 signaling.
  • Antibodies may be used to mediate, e.g., antigen dependent cell-mediated cytotoxicity, complement fixation, to localize enzymes or other means of activating inert pro-toxins, as diagnostic labels, or conjugated to compounds which will absorb energy to eliminate cells in proximity to where the antibody binds.
  • This invention further contemplates the therapeutic use of blocking antibodies to these antigens as antagonists and of stimulatory antibodies as agonists. This approach should be particularly useful with other DC-STAMP or DSP-1 species variants .
  • reagents necessary for effective therapy will depend upon many different factors, including means of administration, target site, physiological state of the patient, and other medicants administered. Thus, treatment dosages should be titrated to optimize safety and efficacy. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents. Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage. Various considerations are described, e.g., in Gilman, et al . (eds.) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, latest Ed. , Pergamon Press; and Remington's Pharmaceutical Sciences, latest ed. , Mack Publishing Co., Easton, Penn.
  • Pharmaceutically acceptable carriers will include water, saline, buffers, and other compounds described, e.g., in the Merck Index, Merck & Co., Rahway, New Jersey. Dosage ranges would ordinarily be expected to be in amounts lower than 1 mM concentrations, typically less than about 10 ⁇ M concentrations, usually less than about 100 nM, preferably less than about 10 pM (picomolar) , and most preferably less than about 1 fM (femtomolar) , with an appropriate carrier.
  • DC-STAMP or DSP-1, fragments thereof, and antibodies to it or its fragments, antagonists, and agonists may be administered directly to the host to be treated or, depending on the size of the compounds, it may be desirable to conjugate them to carrier proteins such as ovalbumin or serum albumin prior to their administration.
  • Therapeutic formulations may be administered in many conventional dosage formulations. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation.
  • Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof .
  • Formulations include those suitable for oral, rectal, nasal, topical, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. See, e.g., Gilman, et al . (eds. 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed. , Pergamon Press; and Remington's Pharmaceutical Sciences, 17th ed. (1990), Mack Publishing Co., Easton, Penn. ; Avis, et al . (eds.
  • compositions Parenteral Medications. Dekker, New York; Lieberman, et al . (eds. 1990) Pharmaceutical Dosage Forms: Tablets, Dekker, New York; and Lieberman, et al . (eds. 1990) Pharmaceutical Dosage Forms: Disperse Systems, Dekker, New York.
  • the therapy of this invention may be combined with or used in association with other agents, e.g., other therapeutics for treatment of symptoms of the indications described.
  • Both naturally occurring and recombinant forms of the DC- STAMPS or DSP- Is of this invention are particularly useful in kits and assay methods which are capable of screening compounds for binding activity to the proteins.
  • Several methods of automating assays have been developed in recent years so as to permit screening of tens of thousands of compounds in a short period. See, e.g., Fodor, et al . (1991) Science 251:767-773, which describes means for testing of binding affinity by a plurality of defined polymers synthesized on a solid substrate.
  • the development of suitable assays can be greatly facilitated by the availability of large amounts of purified, soluble antigens as provided by this invention.
  • antagonists can normally be found once the antigen has been structurally defined, e.g., by tertiary structure data. Testing of potential interacting analogs is now possible upon the development of highly automated assay methods using a purified DC-STAMP or DSP-1. In particular, new agonists and antagonists will be discovered by using screening techniques described herein. Of particular importance are compounds found to have a combined binding affinity for a spectrum of DC-STAMP molecules, e.g., compounds which can serve as antagonists for species variants of DC-STAMP.
  • One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant DNA molecules expressing a DC-STAMP or DSP-1.
  • Cells may be isolated which express an antigen in isolation from other molecules.
  • Such cells either in viable or fixed form, can be used for standard binding partner binding assays . See also, Parce, et al . (1989) Science 246:243-247; and Owicki, et al. (1990) Proc. Nat ' 1 Acad. Sci. USA 87:4007-4011, which describe sensitive methods to detect cellular responses.
  • Another technique for drug screening involves an approach which provides high throughput screening for compounds having suitable binding affinity to an antigen and is described in detail in Geysen, European Patent Application 84/03564, published on September 13, 1984.
  • a solid substrate e.g., plastic pins or some other appropriate surface, see Fodor, et al . (1991).
  • all the pins are reacted with solubilized, unpurified or solubilized, purified DC-STAMP, and washed.
  • the next step involves detecting bound DC-STAMP.
  • Rational drug design may also be based upon structural studies of the molecular shapes of the DC-STAMP or DSP-1 and other effectors or analogs. Effectors may be other proteins which mediate other functions in response to binding, or other proteins which normally interact with the membrane proteins, e.g., a receptor.
  • One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x-ray crystallography or 2 dimensional NMR techniques. These will provide guidance as to which amino acid residues form molecular contact regions, as modeled, e.g., against other cytokine-receptor models.
  • protein structural determination see, e.g.,
  • kits This invention also contemplates use of DC-STAMP or DSP-1 proteins, fragments thereof, peptides, and their fusion products in a variety of diagnostic kits and methods for detecting the presence of another DC-STAMP or DSP-1 or binding partner.
  • the kit will have a compartment containing either a defined DC-STAMP or DSP-1 peptide or gene segment or a reagent which recognizes one or the other, e.g., DC-STAMP or DSP-1 fragments or antibodies.
  • a kit for determining the binding affinity of a test compound to a DC-STAMP would typically comprise a test compound; a labeled compound, for example a binding partner or antibody having known binding affinity for DC-STAMP; a source of DC-STAMP (naturally occurring or recombinant) ; and a means for separating bound from free labeled compound, such as a solid phase for immobilizing the molecule. Compartments containing reagents, and instructions, will normally be provided. Once test compounds are screened, those having suitable binding affinity to the antigen can be evaluated in suitable biological assays, as are well known in the art, to determine whether they act as agonists or antagonists to DC- STAMP signaling pathway. The availability of recombinant DC- STAMP polypeptides also provides well defined standards for calibrating such assays.
  • Antibodies including antigen binding fragments, specific for the DC-STAMP or DSP-1 or fragments are useful in diagnostic applications to detect the presence of elevated levels of antigen and/or its fragments.
  • diagnostic assays can employ lysates, live cells, fixed cells, immunofluorescence, cell cultures, body fluids, and further can involve the detection of antigens related to the antigen in serum, or the like. Diagnostic assays may be homogeneous (without a separation step between free reagent and antigen-binding partner complex) or heterogeneous (with a separation step) .
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • EMIT enzyme-multiplied immunoassay technique
  • SFIA substrate-labeled fluorescent immunoassay
  • Anti-idiotypic antibodies may have similar use to diagnose presence of antibodies against a DC-STAMP or DSP-1, as such may be diagnostic of various abnormal states.
  • overproduction of DSP-1 may result in production of various immunological reactions which may be diagnostic of abnormal physiological states, particularly in allergic conditions.
  • the reagents for diagnostic assays are supplied in kits, so as to optimize the sensitivity of the assay.
  • the protocol, and the label either labeled or unlabeled antibody or binding partner, or labeled antigen is provided. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like.
  • the kit will also contain instructions for proper use and disposal of the contents after use.
  • the kit has compartments for each useful reagent.
  • the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concentrations of reagents for performing the assay.
  • Many of the aforementioned constituents of the drug screening and the diagnostic assays may be used without modification or may be modified in a variety of ways. For MH ⁇ » . ⁇
  • RNA and DNA nucleotide sequences from patients suspected of having an abnormal condition, e.g., inflammatory or autoimmune.
  • an abnormal condition e.g., inflammatory or autoimmune.
  • the preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature. See, e.g., Langer-Safer, et al . (1982) Proc. Nat ' 1. Acad. Sci. 79:4381- 4385; Caskey (1987) Science 236:962-967; and Wilchek et al . (1988) Anal. Biochem. 171:1-32.
  • kits which also test for the qualitative or quantitative expression of other molecules are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers. See, e.g., Viallet, et al . (1989) Progress in Growth Factor Res . 1:89-97. Kits may contain additional reagents to evaluate other cell subsets.
  • Both DC-STAMP and DSP-1 are cell surface antigens, which may be receptor for a ligand or another surface antigen. Having isolated one component of such an interaction, methods exist for isolating a ligand or binding receptor partner. See, Gearing, et al . (1989) EMBO J. 8:3667-3676.
  • means to label the antigen without interfering with the binding to its partner can be determined.
  • an affinity label can be fused to either the amino- or carboxyl-terminus of the ligand.
  • Such label may be a FLAG epitope tag, or, e.g., an Ig or Fc domain.
  • An expression library can be screened for specific binding of the antigen, e.g., by cell sorting, or other screening to detect subpopulations which express such a binding component. See, e.g., Ho, et al . (1993) Proc . Nat ' 1 Acad. Sci. USA 90:11267-11271; and Liu, et al . (1994) J ⁇ Immunol . 152:1821-29. Alternatively, a panning method may be used. See, e.g., Seed and Aruffo (1987) Proc. Nat ' 1 Acad. Sci. USA 84:3365-3369. Protein cross-linking techniques with label can be applied to isolate binding partners of the DC-STAMP or DSP-1. This would allow identification of proteins which specifically interact with the antigen, e.g., in a ligand-receptor or receptor-receptor manner.
  • Methods for protein purification include such methods as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization, and others. See, e.g., Ausubel, et al . (1987 and periodic supplements) ; Deutscher (1990) "Guide to Protein Purification,” Methods in Enzymology vol. 182, and other volumes in this series; Coligan, et al . (1995 and supplements) Current Protocols in Protein Science John Wiley and Sons, New York, NY; P. Matsudaira (ed. 1993) A Practical Guide to
  • Protein and Peptide Purification for Microsequencing Academic Press, San Diego, CA; and manufacturer's literature on use of protein purification products, e.g., Pharmacia, Piscataway, NJ, or Bio-Rad, Richmond, CA.
  • Combination with recombinant techniques allow fusion to appropriate segments (epitope tags) , e.g., to a FLAG sequence or an equivalent which can be fused, e.g., via a protease-removable sequence.
  • Assays for vascular biological activities are well known in the art. They will cover angiogenic and angiostatic activities in tumor, or other tissues, e.g., arterial smooth muscle proliferation (see, e.g., Koyoma, et al . (1996) Cell 87:1069-1078), monocyte adhesion to vascular epithelium (see McEvoy, et al . (1997) J. Exp. Med. 185:2069-2077), etc. See also Ross (1993) Nature 362:801-809; Rekhter and Gordon (1995) Am. J. Pathol. 147:668-677; Thyberg, et al . (1990) Atherosclerosis 10:966-990; and Gumbiner (1996) Cell 84:345- 357.
  • arterial smooth muscle proliferation see, e.g., Koyoma, et al . (1996) Cell 87:1069-1078
  • monocyte adhesion to vascular epithelium see McEvoy,
  • the sequence of the primate e.g., human, DC-STAMP gene is provided in Table 1.
  • the sequence is derived from sequence of a cDNA clone isolated from dendritic cells. This sequence allows preparation of PCR primers, or probes, to determine cellular distribution of the gene. The sequence allows isolation of genomic DNA which encode the message.
  • the ORF of DC-STAMP predicts a protein of 470 amino acids, with a predicted molecular weight of around 53 kD and an isoelectric point of 9.41.
  • the amino terminus of the protein starts with a short stretch of hydrophobic amino acids, which predicts an uncleavable signal sequence (pSORT, Osaka University, Japan) .
  • Hydrophobicity analysis of the sequence revealed 5 strong and 2 weak hydrophobic stretches of 18-20 amino acids, suggesting that the DC-STAMP molecule is spanning the membrane multiple times.
  • the TM Predict program from BCM Search Launcher suggests a topology model in which the DC-STAMP protein contains 7 transmembrane spanning regions, with the N-terminus located outside and the C-terminus on the luminal side of the membrane.
  • the DC-STAMP protein contains two pairs of cysteine residues, one at the start of the first transmembrane domain (TMl) , the other at the end of the second transmembrane domain (TM2) .
  • These cysteines might form a disulphide bridge near the outer side of the membrane, and stabilize the protein structure.
  • Prosite analysis of the protein revealed 3 potential glycosylation sites, two on the second and one on the third putative extracellular loop.
  • there is a consensus sequence for phosphorylation by protein kinase C between the fifth and sixth transmembrane region which is the second intracellular loop according to the proposed topology.
  • the 72 amino acid cytoplasmic tail of DC-STAMP contains several serine residues, two of which might serve as a target for phosphorylation .
  • the C-terminus of the DC- STAMP protein is surprisingly rich in positively charged residues, comprising 25% of the tail and conferring an overall positive charge (+14) .
  • the DSP-1 gene was isolated from a cDNA library made from human HEL cells.
  • PCR products are cloned using, e.g., a TA cloning kit (Invitrogen) .
  • the resulting cDNA plasmids are sequenced from both termini on an automated sequencer (Applied Biosystems) .
  • PBMC Leukocyte preparations PBMC were obtained by leukophoresis of blood from healthy donors, and adherence for 2 hours resulted in a non-adherent PBL fraction. Monocytes were elutriated from PBMC by counterflow centrifugation, resulting in a population of cells that were greater than 85% CD14 . Fractions were cultured in Iscove's medium supplemented with 5% FCS and 1% antibiotics/antimyotics (Life Technologies Inc., Grand Island, NY) .
  • Both the non-adherent PBL and total PBMC were stimulated with phytohemagglutinin (PHA; 1 ⁇ g/ml; Murex Diagnostics Ltd, Dartford, England) and rIL-2 (200 U/ml; Cetus Corp., Emeryville, CA) for 16 h.
  • PHA phytohemagglutinin
  • rIL-2 200 U/ml; Cetus Corp., Emeryville, CA
  • Elutriated monocytes were stimulated with 2 ⁇ g/ml LPS for 16 h.
  • DC were generated in vitro from monocytes using a modification of described methods. See Ridge, et al . (1998) Nature 393:474-478; and Bennett, et al . (1998) Nature 393:478- 480. Monocytes were cultured in AIM-V medium (Life
  • Resulting DC were collected directly or after activation with either LPS for 16 h (2 ⁇ g/ml) , or after the sequential addition of TNF ⁇ (10 ng/ml, 24 h) and the activating anti-CD40 antibody MAB89, generously provided by DNAX, Palo Alto, CA (1.5 ⁇ g/ml, 24 h) .
  • Purified tonsil B lymphocytes were isolated according to the method described by Falkoff, et al . (1982) . J . Immunol .
  • cDNA library preparation Complementary DNA libraries were prepared. See Marland, et al . (1997) in Ricciardi-Castognoli (ed.) Dendritic Cells in Fundamental and Clinical Immunology, Vol 3 , Plenum Publ . Corporation; and Adema, et al . (1997) Nature 387:713-717. Nucleotide sequences were analyzed against the non-redundant GenBank and EMBL databases using the BLAST program. See Altschul, et al . (1990) J. Mol. Biol. 215:403-410.
  • the cDNA was purified with a QIAQuick PCR purification kit (Qiagen) and subsequently tailed using 50 U of Terminal Transferase (Boehringer Mannheim) in the presence of dCTP (5 ⁇ M) and 0.75 mM C0CI2 (15 minutes
  • the tailed cDNA was extracted once with phenol/chloroform and precipitated using glycogen (50 ⁇ g) .
  • 5% of the tailed cDNA was used in a hemi-nested PCR reaction, using nested DC-STAMP specific primer 5'RACE-2 and a 5' primer annealing to the C-tail of the cDNA.
  • 30 PCR cycles were performed using a standard program (1 min 94° C, 1 min 58° C, 1 min 72° C, 10 min extension at 72° C) .
  • the resulting PCR product was gel -purified and cloned into the TA-cloning vector pGEM-T (Promega) .
  • the overlapping cDNA fragments were sequenced by the dideoxy chain reaction (AutoRead Sequencing kit, Pharmacia Biotech) on the ALF Express automated sequencer (Pharmacia Biotech) .
  • the complete ORF of DC-STAMP was amplified from oligo dT transcribed cDNA (Superscript II, Gibco BRL) using the Expand Long Template PCR System (30 cycles, Boehringer Mannheim) and cloned into pGEM-T Easy. Sequence analysis of several clones confirmed the sequence obtained by 5 'RACE PCR.
  • DC-STAMP cDNA encodes a 470 amino acid multimembrane spanning molecule
  • DC-STAMP Since expression of DC-STAMP was specifically detected in DC, a DC cDNA library was screened with a specific probe for DC-STAMP in order to obtain the full-length transcript.
  • DC-STAMP cDNA clones were isolated, of which the longest clone contained an insert of 1.4 kb, identical to the original clone at its 3' end. 5' RACE PCR resulted in the cloning of the most 5' region of the DC-STAMP messenger.
  • the sequence of the primate, e.g., human, DSP-1 gene is provided in Table 2.
  • the sequence is derived from sequence of a cDNA clone isolated human HEL cells.
  • probes specific for cDNA encoding primate antigen are prepared. Typically, the probe is labeled, e.g., by random priming .
  • Southern Analysis DNA (5 ⁇ g) from a primary amplified cDNA library was digested with appropriate restriction enzymes to release the inserts, run on a 1% agarose gel and transferred to a nylon membrane (Schleicher and Schuell, Keene, NH) .
  • Samples for human mRNA isolation may include: peripheral blood mononuclear cells (monocytes, T cells, NK cells, granulocytes, B cells) , resting (T100) ; peripheral blood mononuclear cells, activated with anti-CD3 for 2, 6, 12 h pooled (T101) ; T cell, THO clone Mot 72, resting (T102) ; T cell, THO clone Mot 72, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T103) ; T cell, THO clone Mot 72, anergic treated with specific peptide for 2, 7, 12 h pooled (T104) ; T cell, TH1 clone HY06, resting (T107) ; T cell, TH1 clone HY06, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T108) ; T cell, TH1 clone HY
  • Samples for mouse mRNA isolation can include: resting mouse fibroblastic L cell line (C200) ; Braf:ER (Braf fusion to estrogen receptor) transfected cells, control (C201) ; Mell4+ naive T cells from spleen, resting (T209) ; Mell4+ naive T cells from spleen, stimulated with IFN ⁇ , IL-12, and anti IL-4 to polarize to TH1 cells, exposed to IFN ⁇ and IL-4 for 6, 12, 24 h, pooled (T210) ; Mell4+ naive T cells from spleen, stimulated with IL-4 and anti IFN ⁇ to polarize to Th2 cells, exposed to IL-4 and anti IFN ⁇ for 6, 13, 24 h, pooled (T211) ; T cells, TH1
  • M202 macrophage cell line J774 + LPS + anti-IL-10 at 0.5, 1, 3, 6, 12 h pooled (M203); macrophage cell line J774 + LPS + IL-10 at 0.5, 1, 3, 5, 12 h pooled (M204); unstimulated mast cell lines MC-9 and MCP-12 (M208) ; immortalized endothelial cell line derived from brain microvascular endothelial cells, unstimulated (E200) ; immortalized endothelial cell line derived from brain microvascular endothelial cells, stimulated overnight with TNF ⁇ (E201) ; immortalized endothelial cell line derived from brain microvascular endothelial cells, stimulated overnight with TNF ⁇ (E202) ; immortalized endothelial cell line derived from brain microvascular endothelial cells, stimulated overnight with TNF ⁇ and IL-10 (E203) ; total aorta from wt C57 bl/6 mouse; total a
  • RT-PCR was performed on RNA from a panel of freshly isolated resting or activated leukocyte populations and several cell lines of haematopoietic as well as non-haematopoietic origin.
  • the PCR products were Southern blotted and hybridized with a specific DC-STAMP oligonucleotide.
  • a distinct band of the expected size was detected in immature as well as in mature DC, stimulated with either LPS or a combination of TNF ⁇ and an activating anti-CD40 antibody.
  • freshly isolated monocytes did not express the DC-STAMP RNA, even after overnight stimulation with LPS.
  • CCR1-EGFP into 293 cells resulted in a bright membrane fluorescence, often accompanied by an additional highly fluorescent spot in the cytoplasm, possibly representing the Golgi .
  • Analysis of transient as well as stable transfectants of the DC-STAMP-EGFP construct showed a similar fluorescence staining pattern as seen for CCR1-EGFP, indicating that DC- STAMP can also be expressed at the cell surface.
  • Transfectants expressing the EGFP protein alone showed a bright cytoplasmic fluorescence, not localized to a particular cell structure.
  • the localization of the C-terminus of the DC-STAMP-EGFP protein was determined by staining the DC-STAMP-EGFP transient transfectants with polyclonal anti-GFP serum either before or after permeabilization. Cytospin stainings showed that EGFP could only be detected after permeabilization, indicating that DC-STAMP has an intracellular C-terminus. The amount of positive cells was consistent with the percentage of GFP positive cells in the transient transfected bulk population as observed by FACS analysis (30%) . The few cells that stained positive after pre-incubation with the anti-GFP serum were due to leakage of the antibody into dead cells.
  • Chromosome mapping is a standard technique. See, e.g., BIOS Laboratories (New Haven, CT) and methods for using a mouse somatic cell hybrid panel with PCR.
  • transfected cell lines are screened for one which expresses the desired antigen at a high level compared with other cells.
  • Various cell lines are screened and selected for their favorable properties in handling.
  • Natural antigen can be isolated from natural sources, or by expression from a transformed cell using an appropriate expression vector. Purification of the expressed protein is achieved by standard procedures, or may be combined with engineered means for effective purification at high efficiency from cell lysates or supernatants. FLAG or His ⁇ segments can be used for such purification features. Alternatively, affinity chromatography may be used with specific antibodies, see below. Protein is produced in coli, insect cell, or mammalian expression systems, as desired.
  • HEK 293 cells were transfected with 3 ⁇ g DC-STAMP DNA using LipofectAMINE (Gibco BRL) . See Lanier, et al . (1994) J. Immunol . 153:2417-2428. 2 days after transfection, cells were harvested and used for Confocal Laser Scanning Microscopy (CLSM) . Expression was checked by FACScan analysis in the FITC channel (Becton Dickinson & Co . , Oxnard, CA) and usually 30-60% of the cells were positive for expression. In order to obtain a stable bulk population, G418 (1 mg/ml; Life Technologies Ltd, Paisley, Scotland) was added to the culture medium at day 2 after transfection.
  • Cytospins were fixed with acetone for 10 minutes, incubated with a horse anti-mouse biotinylated antibody and positive cells visualized by immunoperoxidase staining (Vectastain Elite ABC kit, Vector Laboratories, Burlingame, USA; AEC Substrate Kit, Zymed Laboratories, CA) .
  • CSLM Confocal Laser Scanning Microscopy
  • the CSLM settings were: lens, 60x; gain, 1100-1350; pinhole, 1.5 ⁇ m; and magnification, 60x.
  • the DC-STAMP or DSP-1 cDNA, or other species counterpart sequence can be used as a hybridization probe to screen a library from a desired source, e.g., a primate cell cDNA library. Many different species can be screened both for stringency necessary for easy hybridization, and for presence using a probe. Appropriate hybridization conditions will be used to select for clones exhibiting specificity of cross hybridization .
  • Species variants are isolated using cross-species hybridization techniques based upon isolation of a full length isolate or fragment from one species as a probe.
  • antibodies raised against human antigen will be used to screen for cells which express cross-reactive proteins from an appropriate, e.g., cDNA library.
  • the purified protein or defined peptides are useful for generating antibodies by standard methods, as described above.
  • Synthetic peptides or purified protein are presented to an immune system to generate monoclonal or polyclonal antibodies. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene; and Harlow and Lane (1989) Antibodies: A Laboratory Manual Cold Spring Harbor Press.
  • the resulting antibodies are used for screening, purification, or diagnosis, as described.
  • Synthetic peptides or purified protein are presented to an immune system to generate monoclonal or polyclonal antibodies. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene; and Harlow and Lane (1989) Antibodies: A
  • the effect on proliferation or differentiation of various cell types are evaluated with various concentrations of antibody.
  • a dose response analysis is performed.
  • antibodies will be tested on cord blood cells to see if they have effects on proliferation or differentiation of early progenitor cells derived therefrom.
  • the cells are early precursor cells, e.g., stem cells, originating from, e.g., cord blood, bone marrow, thymus, spleen, or CD34+ progenitor cells.
  • the antibodies will be tested for effects on myeloid and/or erythroid precursors, including B cell precursors.
  • PBMC Total PBMC are isolated from buffy coats of normal healthy donors by centrifugation through ficoll-hypaque as described (Boyum, et al . ) .
  • PBMC are cultured in 200 ⁇ l Yssel ' s medium (Gemini Bioproducts, Calabasas, CA) containing 1% human AB serum in 96 well plates (Falcon, Becton-Dickinson, NJ) in the absence or presence of antibodies.
  • Cells are cultured in medium alone or in combination with 100 U/ml IL-2 (R&D Systems) for 120 hours.
  • 3H-Thymidine (0.1 mCi) is added during the last six hours of culture and 3H-Thymidine incorporation determined by liquid scintillation counting.
  • the antibodies would be tested for blocking signaling activity in many other biological assay systems, e.g., on T- cells, B-cells, NK, macrophages, dendritic cells, mast cells, hematopoietic progenitors, etc.
  • Antibodies are evaluated for effects in macrophage/dendritic cell activation and antigen presentation assays, T cell cytokine production or proliferation in response to antigen or allogeneic stimulus. See, e.g., de Waal Malefyt et al. (1991) J. Exp. Med. 174:1209-1220; de Waal Malefyt et al. (1991) J. Exp. Med. 174:915-924; Fiorentino, et al . (1991) J . Immunol . 147, 3815-3822; Fiorentino, et al . (1991) J. Immunol . 146:3444-3451; and Groux, et al . (1996) J . Exp . Med. 184:19-29. Antibodies will be tested for ability to affect mast cell degranulation, chemotaxis, etc.
  • Antibodies will also be evaluated for effects on NK cell stimulation. Assays may be based, e.g., on Hsu, et al . (1992) Internat . Immunol . 4:563-569; and Schwarz, et al . (1994) J. Immunother . 16:95-104. Other assays are applied to evaluate effects on cytotoxic T cells and LAK cells. See, e.g., Namien and Mire-Sluis (1998) .
  • B cell growth and differentiation effects will be analyzed, e.g., by the methodology described, e.g., in Defrance, et al . (1992). J . Exp . Med. 175:671-682; Rousset, et al. (1992) Proc. Nat ' 1 Acad. Sci. USA 89:1890-1893; including IgG2 and IgA2 switch factor assays. Note that, unlike COS7 supernatants, NIH3T3 and COP supernatants apparently do not interfere with human B cell assays.
  • Monocytes are purified by negative selection from peripheral blood mononuclear cells of normal healthy donors.
  • 3 x 10 8 ficoll banded mononuclear cells are incubated on ice with a cocktail of monoclonal antibodies (Becton- Dickinson; Mountain View, CA) consisting, e.g., of 200 ⁇ l of ⁇ CD2 (Leu-5A) , 200 ⁇ l of ⁇ CD3 (Leu-4) , 100 ⁇ l of ⁇ CD8 (Leu 2a) , 100 ⁇ l of ⁇ CD19 (Leu-12 ) , 100 ⁇ l of ⁇ CD20 (Leu-16) , 100 ⁇ l of ⁇ CD56 (Leu-19), 100 ⁇ l of ⁇ CD67 (IOM 67; Immunotech, Westbrook, ME) , and anti-glycophorin antibody (10F7MN, ATCC, Rockville, MD) .
  • a cocktail of monoclonal antibodies consisting, e.g., of 200 ⁇ l of ⁇ CD2 (Leu-5A) , 200 ⁇ l of ⁇ CD3
  • Antibody bound cells are washed and then incubated with sheep anti-mouse IgG coupled magnetic beads (Dynal, Oslo, Norway) at a bead to cell ratio of 20:1. Antibody bound cells are separated from monocytes by application of a magnetic field. Subsequently, human monocytes are cultured in Yssel ' s medium (Gemini Bioproducts, Calabasas, CA) containing 1% human monocytes.
  • Analyses of the expression of cell surface molecules can be performed by direct immunofluorescence. For example, 2 x 10 5 purified human monocytes are incubated in phosphate buffered saline (PBS) containing 1% human serum on ice for 20 minutes. Cells are pelleted at 200 x g. Cells are resuspended in 20 ml PE or FITC labeled mAb. Following an additional 20 minute incubation on ice, cells are washed in PBS containing 1% human serum followed by two washes in PBS alone. Cells are fixed in PBS containing 1% paraformaldehyde and analyzed on PBS.
  • PBS phosphate buffered saline
  • FACScan flow cytometer (Becton Dickinson; Mountain View, CA) .
  • Exemplary mAbs are used, e.g.: CDllb (anti-macl) , CDllc (a gpl50/95) , CD14 (Leu-M3) , CD54 (Leu 54) , CD80 (anti-BBl/B7) , HLA-DR (L243) from Becton-Dickinson and CD86 (FUN 1; Pharmingen), CD64 (32.2; Medarex), CD40 (mAb89; Schering-Plough France) .
  • CDllb anti-macl
  • CDllc a gpl50/95
  • CD14 Leu-M3
  • CD54 Leu 54
  • CD80 anti-BBl/B7
  • HLA-DR L243
  • CD64 32.2; Medarex
  • CD40 mAb89; Schering-Plough France
  • Human monocytes are isolated as described and cultured in Yssel's medium (Gemini Bioproducts, Calabasas, CA) containing 1% human AB serum in the absence or presence of antibodies.
  • monocytes are stimulated with LPS (E. coli 0127 :B8 Difco) in the absence or presence of antibodies and the concentration of cytokines (IL-l ⁇ , IL-6, TNF ⁇ , GM-CSF, and IL- 10) in the cell culture supernatant determined by ELISA. Additional assays will be tested in the areas of bone remodeling, chondriocytes, neurons, adipocytes, gastrointestinal epithelium, or bronchial epithelium.
  • Such animals are useful to determine the effects of deletion of the gene, in specific tissues, or completely throughout the organism. Such may provide interesting insight into development of the animal or particular tissues in various stages. Moreover, the effect on various responses to biological stress can be evaluated. See, e.g., Hogan, et al . (1995) Manipulating the Mouse Embryo: A Laboratory Manual (2d ed.) Cold Spring Harbor Laboratory Press.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne des gènes purifiés codant pour des protéines de mammifère, des réactifs associés à ceux-ci et incluant les protéines purifiées, des anticorps spécifiques et des acides nucléiques codant pour ces molécules. L'invention concerne également des procédés d'utilisation de ces réactifs et des trousses diagnostiques.
EP00978596A 1999-11-15 2000-11-15 Genes de mammifere, reactifs associes et procedes Withdrawn EP1240183A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US43973599A 1999-11-15 1999-11-15
US439735 1999-11-15
PCT/US2000/031167 WO2001036463A2 (fr) 1999-11-15 2000-11-15 Genes de mammifere, reactifs associes et procedes

Publications (1)

Publication Number Publication Date
EP1240183A2 true EP1240183A2 (fr) 2002-09-18

Family

ID=23745922

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00978596A Withdrawn EP1240183A2 (fr) 1999-11-15 2000-11-15 Genes de mammifere, reactifs associes et procedes

Country Status (6)

Country Link
EP (1) EP1240183A2 (fr)
JP (1) JP2003514546A (fr)
AU (1) AU1604701A (fr)
CA (1) CA2391669A1 (fr)
MX (1) MXPA02004825A (fr)
WO (1) WO2001036463A2 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6933367B2 (en) 2000-10-18 2005-08-23 Maxygen Aps Protein C or activated protein C-like molecules
CA2462883A1 (fr) 2001-10-12 2003-04-17 Schering Corporation Utilisation d'anticorps bispecifiques pour reguler des reponses immunitaires
AU2002952993A0 (en) * 2002-11-29 2002-12-12 The Corporation Of The Trustees Of The Order Of The Sisters Of Mercy In Queensland Therapeutic and diagnostic agents
JP4633491B2 (ja) * 2004-02-12 2011-02-16 第一三共株式会社 破骨細胞関連タンパク質を標的とした抗体
TWI359026B (en) * 2004-02-12 2012-03-01 Sankyo Co Pharmaceutical composition for the osteoclast rela

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5356498A (en) * 1996-11-27 1998-06-22 Schering Corporation Isolated mammalian dendritic cell genes; related reagents
CA2229449A1 (fr) * 1997-04-25 1998-10-25 Takeda Chemical Industries, Ltd. Proteine receptrice nouvelle et son utilisation
US6617428B1 (en) * 1998-01-14 2003-09-09 The Corporation Of The Trustees Of The Order Of The Sisters Of Mercy In Queensland Human CMRF-35-H9 receptor which binds IgM

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0136463A3 *

Also Published As

Publication number Publication date
WO2001036463A2 (fr) 2001-05-25
AU1604701A (en) 2001-05-30
JP2003514546A (ja) 2003-04-22
MXPA02004825A (es) 2004-08-12
CA2391669A1 (fr) 2001-05-25
WO2001036463A3 (fr) 2001-11-22

Similar Documents

Publication Publication Date Title
US7608690B2 (en) IL-B30 antibodies
EP1002084B1 (fr) Cytokine de mammifère: interleukine-b30 et reactifs correspondants
US9422355B2 (en) Antibodies to IL-B50
JP2013255510A (ja) ヒトインターロイキン−b50、治療的使用
EP1159299B1 (fr) Cytokines de mammifere : reactifs et procedes associes
US20100261190A1 (en) Mammalian Genes; Related Reagents and Methods
US6800460B1 (en) Mammalian cytokine complexes
EP1240183A2 (fr) Genes de mammifere, reactifs associes et procedes
WO2002068596A2 (fr) Cytokines de mammifere, reactifs associes
EP2138510B1 (fr) Méthodes de génération d'anticorps dirigés contre des cytokines

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020521

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO PAYMENT 20020521;SI PAYMENT 20020521

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: STICHTING KATHOLIEKE UNIVERSITEIT

Owner name: SCHERING CORPORATION

17Q First examination report despatched

Effective date: 20041102

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20050313