WO2000049149A1 - Novel tnf receptor-like proteins - Google Patents

Novel tnf receptor-like proteins Download PDF

Info

Publication number
WO2000049149A1
WO2000049149A1 PCT/JP2000/000940 JP0000940W WO0049149A1 WO 2000049149 A1 WO2000049149 A1 WO 2000049149A1 JP 0000940 W JP0000940 W JP 0000940W WO 0049149 A1 WO0049149 A1 WO 0049149A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
seq
present
amino acid
Prior art date
Application number
PCT/JP2000/000940
Other languages
French (fr)
Japanese (ja)
Other versions
WO2000049149A9 (en
Inventor
Toshio Kitamura
Tetsuo Kojima
Original Assignee
Toshio Kitamura
Tetsuo Kojima
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 Toshio Kitamura, Tetsuo Kojima filed Critical Toshio Kitamura
Priority to AU25742/00A priority Critical patent/AU2574200A/en
Publication of WO2000049149A1 publication Critical patent/WO2000049149A1/en
Publication of WO2000049149A9 publication Critical patent/WO2000049149A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]

Definitions

  • the present invention relates to a novel receptor protein, an MA encoding the protein, a vector containing the MA, a host cell carrying the vector, an antibody against the protein, a method for screening a compound using the protein, It relates to a compound that can be isolated by a screening method. North
  • Cytokines and their receptors that act in the brain are important targets for the development of new brain-related drugs.
  • cytokines that act on the brain include nerve growth factor? (P 01138), brain-derived neurotrophic factor (P23560), neurotrophin-3 (P207 83), neurotrophin-4 (P34130), and neurotrophin-6 (P34132).
  • Nutritional factors such as neurotrophin-6 ⁇ (P34133) and neurotrophin-6r (P34134) and hormone-like cytokines such as leptin (P41160) have been reported.
  • An object of the present invention is to provide a novel TNF receptor-like protein and its gene. Another object of the present invention is to provide a vector into which the gene has been inserted, a host cell having the vector, and an antibody that binds to the protein. Sa Furthermore, an object of the present invention is to provide a method of screening for a compound that binds to the protein, such as a ligand, using the protein.
  • the present invention relates to novel receptor proteins and their genes and their uses, more specifically,
  • a method for producing the protein according to (5) comprising culturing the host cell according to (4), and collecting a protein expressed from the host cell or a culture supernatant thereof.
  • nucleotide having a length of at least 15 bases which hybridizes with a DNA consisting of the nucleotide sequence of SEQ ID NO: 1, 8, or 10 or a complementary strand thereof,
  • ligand refers to a protein that activates its function by binding to the protein of the present invention expressed on a cell membrane.
  • agonist refers to a protein that can cause a phenomenon similar to the binding of the protein of the present invention to a ligand (activation of the protein of the present invention).
  • ligand activation of the protein of the present invention.
  • molecules that can bind refers to molecules that can bind.
  • angigonist refers to a molecule that specifically binds to the protein of the present invention to suppress its function.
  • the present invention provides a novel membrane secreted protein.
  • the nucleotide sequence of the mouse-derived cDNA (named mouse Troy) isolated by the present inventors is shown in SEQ ID NO: 1, and the amino acid sequence of the protein encoded by the mouse Troy cDNA is shown in SEQ ID NO: 2.
  • the nucleotide sequence of human cDNA (named human Troy) corresponding to mouse Troy cDNA is shown in SEQ ID NO: 10, and the amino acid sequence of the protein encoded by human Troy cDNA is shown in SEQ ID NO: 11.
  • These proteins are compatible with known proteins belonging to the TNF receptor superfamily, have a signal sequence at the N-terminus, and have a transmembrane region in the middle that is expected to be a highly hydrophobic amino acid region.
  • these proteins are novel proteins belonging to the TNF receptor superfamily (Beulter.B, and Huffel.C.V (1994) SCIENCE 264, 667-668 Unraveling function in the TNF ligand and receptor knew s). It is believed that there is.
  • mouse dTroy a cDNA clone encoding a protein lacking most of the intracellular domain of the mouse Troy protein (designated mouse dTroy).
  • the nucleotide sequence of mouse dTroy cDNA is shown in SEQ ID NO: 8
  • the amino acid sequence of the protein encoded by mouse dTroy cDNA is shown in SEQ ID NO: 9. Since the protein lacks most of the intracellular domain and does not show NF-KB activation ability, it is expressed on the cell membrane and binds to the ligand to convert the complete ligand to Troy. It is considered that binding is inhibited, and thereby signal transmission is inhibited.
  • TNFR members form homotrimers and transmit signals, but may inhibit signal transmission by forming heterotrimers containing dTroy.
  • proteins having such functions for example, TRAIL-R3 (TRAIL decoy-type receptor) and DcR3 (FasL decoy-type receptor) are known.
  • the invention also includes proteins that are functionally equivalent to the mouse and human Troy proteins.
  • “functionally equivalent” means that the target protein is extracellular.
  • the function as such a receptor protein includes, for example, the ability to induce NF-kB activity.
  • the ability to induce NF-kB activity can be detected by the method described in Example 6.
  • the invention also includes proteins functionally equivalent to the mouse dTroy protein.
  • “functionally equivalent” means that the target protein functions as a decoy receptor protein.
  • the function as a decoy-type receptor protein includes, for example, a function of inhibiting the ability of a full-type receptor protein (for example, Troy protein) to induce NF-kB activity.
  • proteins having one or more mutated amino acids in the amino acid sequence of the mouse and human Troy protein or Pomatus dTroy protein, and functionally equivalent to these proteins are also present in the present invention. Included in the proteins of the invention.
  • the number of amino acids to be mutated in such a mutant is generally considered to be within 30 amino acids, preferably within 15 amino acids, more preferably within 5 amino acids, and further preferably within 3 amino acids.
  • the amino acid residue to be mutated is mutated to another amino acid in which the properties of the amino acid side chain are conserved.
  • the properties of the amino acid side chain include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V) and hydrophilic amino acids (R, D, N, C, E, Q, G , H, K, S, T), amino acids having aliphatic side chains (G, A, V, L, I, P), amino acids having hydroxyl-containing side chains (S, ⁇ , ⁇ ), sulfur atom-containing side Amino acids (D, N, E, Q) with carboxylic acids and amide-containing side chains, amino acids with side chains containing bases (R, K, ⁇ ), aromatic-containing Amino acids having a chain ( ⁇ , F, Y, W) can be mentioned (all brackets indicate one letter of amino acids).
  • Examples of the protein to which one or more amino acid residues are added include a fusion protein containing Troy protein or dTroy protein of the present invention.
  • the fusion protein is a fusion of these proteins with other peptides or proteins, and is included in the present invention.
  • the method for producing a fusion protein is as follows: a DNA encoding the protein of the present invention and another peptide or a DNA encoding the protein are ligated so that their frames coincide with each other, and this is introduced into an expression vector; And a known method can be used.
  • Other peptides or proteins to be fused with the protein of the present invention are not particularly limited.
  • Hybridization to isolate DNA encoding a functionally equivalent protein can be performed, for example, under conditions where the stringency is 10% formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm MA. . More preferred conditions (more stringent conditions) are 25% formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm DNA, and more preferred conditions (more stringent conditions) are 50% The conditions are formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm DNA.
  • the DNA encoded by the DNA isolated by the hybridization technique or the gene amplification technique usually has a high homology in amino acid sequence with the protein of the present invention described in SEQ ID NO: 2, 9, or 11.
  • High homology usually means a homology of 70% or more, preferably 85% or more, more preferably 95% or more.
  • the literature Wang, WJ an d Lipman, DJ Proc. Natl. Acad. Sci. USA (1983) 80, 726-730).
  • the protein of the present invention can be prepared as a recombinant protein or as a natural protein by methods known to those skilled in the art.
  • a recombinant protein for example, a portion other than the region required for membrane binding of the protein of the present invention is extracellularly secreted as a soluble protein.
  • the culture supernatant of the cells is collected and concentrated, and then subjected to chromatography such as ion exchange, reverse phase, gel filtration, or affinity chromatography in which the antibody against the protein of the present invention is immobilized on a column. Purification can be carried out by combining them or by combining a plurality of these columns.
  • the protein of the present invention is expressed in a host cell (for example, an animal cell or Escherichia coli) as a fusion protein with a glutathione S-transferase protein or as a recombinant protein to which a plurality of histidines are added.
  • the recombinant protein is purified using a glutathione column or a nickel column. Thereafter, if necessary, a region other than the target protein in the fusion protein can be prepared by a method of cleaving with thrombin or Factor-Xa and removing it.
  • the protein is a natural protein, it is purified by, for example, allowing an extract of cells expressing the protein of the present invention to act on an affinity column to which the antibody of the present invention described later is bound. Can be isolated.
  • the present invention also includes partial peptides of the protein of the present invention.
  • the partial peptide of the present invention has an amino acid sequence of at least 8 amino acids or more, preferably 15 amino acids or more, more preferably 50 amino acids or more (for example, 100 amino acids or more).
  • the partial peptide is used, for example, for preparing an antibody against the protein of the present invention, screening a compound such as a ligand that binds to the protein of the present invention, and preparing a competitive inhibitor of the protein of the present invention. obtain.
  • the partial peptide of the present invention includes, for example, the protein of the present invention in which the signal peptide is removed from the protein and the protein of the present invention.
  • soluble proteins from which the region required for membrane binding has been removed and proteins in the form of corn by removing the intracellular region.
  • a partial peptide (which functions as a competitive inhibitor of the receptor of the present invention) that has the ability to bind to a ligand but does not have the ability to transmit signals into cells is included.
  • the partial peptide of the present invention can be produced by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
  • the present invention also provides a DNA encoding the protein of the present invention.
  • the DNA of the present invention may be in any form as long as it can encode the protein of the present invention. That is, whether it is cDNA synthesized from mRNA, genomic DNA, or chemical synthesis! It does not matter whether it is NA or not. Also, as long as it can encode the protein of the present invention, MA having an arbitrary base sequence based on the degeneracy of the genetic code is included.
  • the DNA of the present invention can be prepared by a method known to those skilled in the art.
  • a cDNA library is prepared from cells expressing the protein of the present invention, and a part of the DNA sequence of the present invention (for example, the DNA sequence of SEQ ID NO: 1, 8, or 10) is probed. And by performing hybridization.
  • RNA is prepared from cells expressing the protein of the present invention, and oligo DNA is synthesized based on the sequence of the DNA of the present invention (for example, the DNA sequence described in SEQ ID NO: 1, 8, or 10).
  • it can also be prepared by performing a PCR reaction using this as a primer and amplifying a cDNA encoding the protein of the present invention.
  • the DNA of the present invention can be used to produce the protein of the present invention as a recombinant protein. If the DNA encoding the protein of the present invention is defective, application to antisense function inhibition, gene therapy for replacement with a normal gene, and the like can be considered.
  • the present invention also provides a vector into which the DNA of the present invention has been inserted.
  • the vector of the present invention is not particularly limited as long as it can express the DNA of the present invention in a host cell.
  • the vector is For example, JM109, DH5, and HB10U XLlBlue
  • JM109, DH5, and HB10U XLlBlue have an “ori” to be amplified in Escherichia coli for large-scale amplification and preparation in Escherichia coli
  • a selection gene for transformed Escherichia coli for example, any drug (A drug resistance gene that can be identified by ampicillin, tetracycline, kanamycin, chloramphenicol) is not particularly limited.
  • M13 system vector For example, M13 system vector, pUC system vector, pBR322, pBluescript, pCR-Script, and the like.
  • pGEM-T For the purpose of cDNA subcloning and excision, pGEM-T, pDIRECT, pT7, etc. may also be used in addition to the above vectors.
  • an expression vector is particularly useful.
  • an expression vector for example, when the purpose is expression in E. coli, in addition to having the above characteristics such that the vector is amplified in E. coli, when the host is E. coli such as JM109, DH5, HB10K XLlBlue, etc.
  • a promoter eg, lac, T7, etc.
  • a vector examples include pGEX, pEGFP, or pET (in this case, the host is preferably BL21 expressing T7 MA polymerase) in addition to the above vectors.
  • the promoters required for expression in cells (SV40, MMLV-LT1, EF1, CMV It is essential to have a promoter, etc., and it is more preferable to have a gene for selecting transformation into cells (for example, a drug resistance gene that can be distinguished by a drug (neomycin, G418, etc.)).
  • Vectors having such characteristics include, for example, bandits, pDR2, pBK-RSV, pBK-CMV ⁇ pOPRSV, and pOP13.
  • a vector having a DHFR gene that complements a nucleic acid synthesis pathway-deficient CH0 cell eg, , PCHOI, etc.
  • amplifying with methotrexate (MTX) MTX
  • COS cells having the SV40 T antigen on the chromosome are used to transform the cells with the replication mechanism of SV40 (such as pcD ").
  • the DNA of the present invention is incorporated into an appropriate vector, and the retroviral method, the ribosome method, the cationic liposome method, the adenovirus method, and the like are used to express the DNA in vivo.
  • the vector to be used is not particularly limited, and examples include pAdexlcw and pZIPneo.
  • General genetic manipulation such as insertion of the DNA of the present invention into a vector can be performed according to a conventional method (Molecular Cloning, 5.6, 5.63).
  • the present invention also provides a host cell into which the vector of the present invention has been introduced.
  • the host cell into which the vector of the present invention is introduced is not particularly limited, and for example, Escherichia coli and various animal cells can be used. Examples of Escherichia coli include JM109, DH5 and HB101, and examples of animal cells include CH0 cells, COS cells, 3T3 cells, and HeLa cells. In animal cells, when large-scale expression is intended, CH0 cells are particularly preferred.
  • the introduction of the vector into a host cell can be carried out by, for example, a calcium phosphate method, a DEAE dextran method, electo-portation, lipofection, and the like.
  • the present invention also provides an antibody that binds to the protein of the present invention.
  • the form of the antibody of the present invention is not particularly limited, and includes a monoclonal antibody as well as a polyclonal antibody. Also included are antisera obtained by immunizing egrets and the like with the protein of the present invention, all classes of polyclonal and monoclonal antibodies, and human antibodies and humanized antibodies obtained by genetic recombination.
  • the antibody of the present invention can be prepared by the following method.
  • a polyclonal antibody for example, a small animal such as a heron is immunized with the protein of the present invention to obtain a serum, which is then subjected to an affinity column in which the protein of the present invention is coupled. Of immunoglobulin G or M from this fraction was applied to a protein A or protein G column. It can be prepared by further purification.
  • a monoclonal antibody a small animal such as a mouse is immunized with the protein of the present invention, the spleen is excised from the mouse, and the spleen is crushed into cells, and mouse myeloma cells and reagents such as polyethylene glycol are used.
  • a clone that produces an antibody against the protein of the present invention is selected from the resulting fused cells (hybridomas).
  • the obtained hybridoma was transplanted into a mouse intraperitoneal cavity, ascites was recovered from the mouse, and the obtained monoclonal antibody was subjected to, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, It can be prepared by purifying the protein of the present invention using a coupled affinity column or the like.
  • the antibody of the present invention is used for purification and detection of the protein of the present invention, and is also a candidate for an agonist and an agonist of the protein of the present invention. It is also conceivable to apply the antibody of the present invention to antibody therapy for neurological diseases. When used for antibody therapy, human or humanized antibodies are preferred to reduce immunogenicity.
  • a transgenic animal having a repertoire of human antibody genes is immunized with a protein serving as an antigen, a protein-expressing cell or a lysate thereof to obtain an antibody-producing cell, and a hybridoma obtained by fusing the antibody-producing cell with myeloma cells is used.
  • a human antibody against the protein see International Publication Nos. W092-03918, W093-2227, W094-02602, W094-25585, W096-33735 and W096-34096).
  • the antibody of the present invention may be an antibody fragment or modified antibody thereof as long as it binds to the protein of the present invention.
  • an antibody fragment Fab, F (ab r ) 2, Fv or a single chain Fv (scFv) in which an Fv of an H chain and an L chain are linked by an appropriate linker
  • the antibody is treated with an enzyme, for example, papain or pepsin, to generate an antibody fragment, or a gene encoding these antibody fragment is constructed and introduced into an expression vector.
  • Expression in host cells eg, Co, M. S.
  • modified antibody an antibody bound to various molecules such as polyethylene glycol (PEG) can be used.
  • PEG polyethylene glycol
  • the “antibody” of the present invention also includes these modified antibodies.
  • Such a modified antibody can be obtained by subjecting the obtained antibody to chemical modification. These methods are already established in this field.
  • the present invention also provides a nucleotide that hybridizes with a DNA consisting of the nucleotide sequence shown in SEQ ID NO: 1, 8, or 10, or a complementary strand thereof, and has a length of at least 15 bases.
  • a nucleotide is preferably a nucleotide that specifically hybridizes with a DNA consisting of the nucleotide sequence shown in SEQ ID NO: 1, 8, or 10, or a complementary strand thereof.
  • the term "specifically hybridize” as used herein refers to a state in which hybridization with DNA encoding another protein is carried out under ordinary hybridization conditions, preferably under stringent hybridization conditions. It means that no hybridization occurs significantly.
  • nucleotides include probes and primers capable of specifically hybridizing with the DNA encoding the protein of the present invention or its complementary strand, nucleotides or nucleotide derivatives (for example, antisense oligonucleotides and ribozymes). .
  • the present invention also provides a method for screening for a compound that binds to the protein of the present invention, using the protein of the present invention.
  • the protein of the present invention used for screening may be any of a recombinant type, a natural type and a partial peptide.
  • purified proteins and their partial peptides Or a form expressed on the cell surface or a form as a membrane fraction.
  • This screening method comprises: (a) a step of bringing a test sample into contact with the protein of the present invention or a partial peptide thereof; (b) a step of selecting a compound having an activity of binding to the protein of the present invention or a partial peptide thereof; including.
  • the test sample used in this screening method is not particularly limited, and examples include a cell extract, a cell culture supernatant, a protein, a peptide, and a synthetic low-molecular compound.
  • the protein of the present invention that makes a test sample come into contact with the test sample can be, for example, a purified protein, a form expressed on a cell membrane, or a cell membrane fraction.
  • a method for isolating a ligand to the protein using the protein of the present invention many methods known to those skilled in the art can be used.
  • a cDNA library using a phage vector eg, gtll, ZAP, etc.
  • the expressed protein is fixed, the protein of the present invention is purified as a fusion protein with a biotin label or a GST protein, and the purified protein is reacted with the above-mentioned filter, and a plaque expressing the protein to be bound is obtained.
  • a cDNA library is prepared that can be expressed in the form of a cell, and is introduced into the yeast cells described above.
  • the cDNA derived from the library is isolated from the positive clones detected and introduced into E. coli for expression.
  • “Two hybrid system” (“ MATCHMARKER Two-Hybrid Systemj, "Ma thigh alian MATC HMAKER Two-Hybrid Assay Kitj,” MATC thigh KER One-Hybrid Systemj (all manufactured by clontech), "HybriZAP Two-Hybrid Vector Systemj (stratagene), literature”
  • screening for a ligand that specifically binds to the protein of the present invention comprises screening the extracellular domain of the protein of the present invention and the intracellular domain containing the transmembrane domain of a receptor protein such as a hemopoietin receptor having a known signal transduction ability. And expressed on the cell surface of an appropriate cell line, preferably a cell line that can survive and grow only in the presence of an appropriate growth factor (growth factor-dependent cell line). Thereafter, the cell line can be cultured by adding a material expected to contain various growth factors, cytodynamic factors, hematopoietic factors, and the like.
  • This method utilizes the fact that the above-mentioned growth factor-dependent cell line can survive and proliferate only when the test material contains a ligand that specifically binds to the extracellular domain of the protein of the present invention.
  • Known hemopoietin receptors include, for example, thrombopoietin receptor, erythropoietin receptor, G-CSF receptor, gpl30, and the like.Partners of the chimeric receptor used in the screening system of the present invention include those known The receptor is not limited to the hemopoietin receptor, and any receptor having a structure necessary for signal transduction activity in the cytoplasmic domain may be used.
  • the 0X40 receptor belonging to the same TNF receptor superfamily as the protein of the present invention can be selected as a partner of the chimeric receptor (Mallet, S. et al., EMB 0 J., 19, 1063). -1068 (1990)). Since the 0X40 receptor can transmit a positive signal into cells, screening for ligands using proliferative response etc. as an index (Baum, PR et al., EMBO J. 5 13, 3992-4001 (1994)). Proliferation factors As resident cell lines, for example, IL3-dependent cell lines such as BaF3 and FDC-P1 can be used.
  • the protein of the present invention is expressed in a cell that does not express the ligand, and then an expressed cDNA library constructed from cells that are expected to express the ligand is expressed in the cell.
  • “Direct expression cloning method” in which the culture supernatant obtained by introduction into cells is added, and ligands are searched based on the functional or morphological changes of the cells (Yokota T, Otsuka T, Mosmann II, Banchereau J, DeFrance T, Blanchard D, De Vries JE, Lee F, and Arai K.
  • the culture supernatant of cells expected to express the ligand of the present invention is placed on an affinity column on which the protein of the present invention is immobilized, and the protein that specifically binds to the column is purified.
  • the DNA encoding the ligand can be obtained by analyzing the amino acid sequence of the obtained protein (ligand), synthesizing oligo DNA based on the amino acid sequence, and screening a cDNA library using the DNA as a probe. It is possible.
  • an antibody-like chimeric protein can be used as an agonist.
  • Methods for isolating an agonist and an engonist against the protein using the protein of the present invention include, for example, a compound, a natural product bank, or a random phage in the immobilized protein of the present invention.
  • Screening for binding molecules using peptide display libraries and high-throughput screening methods using combinatorial chemistry technology (Wrighton NC; Farrell FX; Chang R; Kashyap AK; Barbone FP; Mu lcahy LS; Johnson DL; Barrett RW; Jolliffe LK; Dower WJ., Small peptides as potent mimetics of the protein hormone erythropoietin, Science (UNITE D STATES) Jul 26 1996, 273 p458-64, Verdine GL., The Nature (ENGLAND) Nov 7 1996, 384 pi 13, Hogan JC Jr., Di rected combinatorial chemistry.Nature (ENGLAND) Nov 7 1996, 384 pl 7-9) are known to those skilled in the art.
  • the present invention also provides a method for screening for a compound that regulates the activity of the protein of the present invention.
  • This screening method comprises the steps of: (a) covering cells containing a vector that expresses a DNA encoding the protein of the present invention and a vector that expresses a repo allele in response to binding of NF-KB; Contacting a test sample, (b) detecting the repo overnight activity in the cells, and (c) comparing the repo activity to the test sample without contacting the cells. Decrease or increase the activity overnight Selecting a compound.
  • pCOSl or pME18 can be used as a vector for expressing DNA encoding the protein of the present month.
  • a vector that expresses a reporter gene in response to activation of NF-kB for example, kB-luc described in Examples can be used.
  • kB-luc expresses a reporter gene according to the following principle. That is, when a signal is transmitted to the protein of the present invention, kinases downstream thereof are activated via TRAF (TNFR associated factor). This kinase activates the IkB kinase complex and phosphorylates the inhibitory protein IkB bound to NF-kB. Then, IkB is degraded, and NF-kB is released and translocated into the nucleus. When this NF-kB binds to the binding site on kB-luc, the repo overnight gene is expressed accordingly.
  • TRAF TNFR associated factor
  • the HEK293T cell line is preferable.
  • Introduction of a vector into a cell can be performed by a method known to those skilled in the art.
  • a test sample is brought into contact with cells into which a vector has been introduced, and the repo overnight activity is detected.
  • the test sample is not particularly limited, and includes, for example, a cell extract, a cell culture supernatant, a protein, a peptide, and a synthetic low-molecular compound. It is also conceivable to use a compound isolated by screening for a compound that binds to the protein of the present invention.
  • the test sample used inhibits the activity of the protein of the present invention if the repo overnight activity is lower than when the test sample is not brought into contact with the cells.
  • the reporter activity is increased, it is determined that the test sample used induces or increases the activity of the protein of the present invention.
  • the ligand, agonist or aminogonist isolated by the screening of the present invention can be applied to the treatment of a disease associated with the protein of the present invention (for example, a nerve-related disease).
  • Compound isolated by the screening method of the present invention When used as a drug, it can be formulated into a drug by a known pharmaceutical manufacturing method. For example, it is administered to a patient together with a pharmacologically acceptable carrier or vehicle (eg, saline, vegetable oils, suspensions, surfactants, stabilizers, etc.). Administration will be transdermal, intranasal, transbronchial, intramuscular, intravenous, or oral, depending on the nature of the compound. The dose varies depending on the patient's age, body weight, symptoms, administration method, and the like, but those skilled in the art can appropriately select an appropriate dose.
  • a pharmacologically acceptable carrier or vehicle eg, saline, vegetable oils, suspensions, surfactants, stabilizers, etc.
  • Administration will be transdermal
  • the dosage of the compound having the binding activity to the protein of the present invention may vary depending on the condition. It is believed to be 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
  • the single dose varies depending on the subject of administration, target organ, symptoms, and administration method.
  • parenteral injections usually for adults (with a body weight of 60 kg)
  • the dose can be administered in terms of the amount converted per 60 kg body weight or the amount converted per body surface area.
  • FIG. 1 shows the results of analyzing the ability of mouse Troy and dTroy to induce NF-kB activity.
  • Example 1 Mouse brain cDNA library by signal sequence trap (SST) method Bally Screening
  • BstXI adapter-1 Invitrogen was added, and a cDNA of 400 bp or more was fractionated using a SizeSep 400 Spun Column (Pharmacia). Separately, the mixture was mixed with BstXI (Takara Shuzo) and pMXGM (-) v-mpl M2 (see Japanese Patent Application No.
  • Plasmids extracted from the recombinant Escherichia coli were purified using a JETstar column (GEN0M ED). Acad. Sci. USA, vol. 90: 8392-8396, 1993) was used to transfect a plasmid cDNA library using LipofectMINE (LIFE TECHNOLOGIES).
  • B0SC23 is seeded on a 6 cm dish (Corning) with Dulbecco's Modified Eagel Medium (DM EM, LIFE TECHNOLOGIES) containing 10% fetal calf serum (FCS, JRH BIOSCIENCES), and after 16 hours, washed with DMEM and washed first.
  • DM EM Dulbecco's Modified Eagel Medium
  • FCS fetal calf serum
  • JRH BIOSCIENCES fetal calf serum
  • IL-3 Mouse interleukin-3
  • 10 g / ml hexadimethrine bromide were added to the culture supernatant containing the recombinant virus, and Ba / F3 cells were suspended and infected with the mixture. Twenty-four hours after the infection, the cells were washed three times with a phosphate buffer, and cultured with RPMI1640 containing 10% FCS.
  • a chromosomal DNA was extracted from a clone grown in the absence of IL-3, and a primer (5, -gggggtggaccatcctcta-3 '/ SEQ ID NO: 3, which was set so as to sandwich the cDNA insertion site, And 5, -cgcgcagctgtaaacggtag-3, / SEQ ID NO: 4), and a cDNA fragment was recovered by PCR.
  • PCR 500 ng chromosomal DNA, 500 pM each primer, TaKaRa LA Taq (Takara Shuzo) 2.5 units, 2.5 mM MgCl 2 , 0.3 mM dNTPs and enzyme attached buffer went.
  • a mouse brain cDNA library was synthesized using oligo dT primers, and the above cDNA fragment was screened as a probe.
  • 0.1 ⁇ g of the DNA fragment previously obtained by the SST method was converted into type III and its internal primer (50 pmoles) (55S; 5, one caaggtcctacctctacaca—3 // Rooster column numbers: 6, and 511A; 5′-aaggtt caccttgctggtac-3, / SEQ ID NO: 7), 2.5 mM MgCl 2 , 0.2 mM dATP, dGTP, dCTP ⁇ 0.
  • a cDNA fragment obtained using a mouse Multiple Tissue Northern (MTN) Blot was hybridized to a probe, A strong signal was observed in the brain, a weak signal was observed in the heart 'lung' and liver, and an extremely weak signal was also observed in the skeletal muscle and kidney 'testes', suggesting that the receptor was expressed specifically in the brain.
  • MTN Multiple Tissue Northern
  • Example 4 A clone containing almost no intracellular domain “Isolation of mouse dTroyj” A skin cDNA library of a mouse embryo on day 17 was constructed using a random hexamer as a primer, and the extracellular domain of mouse cDNA was constructed. Using the portion as a probe, clones were isolated by the RecA method, and as a result, a clone dTroy containing almost no intracellular domain was isolated, and the nucleotide sequence of the dTroy cDNA was represented by SEQ ID NO: 8, and the cDNA was coded. The amino acid sequence of the protein is shown in SEQ ID NO: 9.
  • a human gliosarcoma cell line (gliosarcoma) GI-1 expression of a human homologous molecule of the mouse Troy gene was observed in a Northern plot. Therefore, a cDNA library derived from the human glial sarcoma cell line GI-1 was constructed using random hexamers as primers, and the RecA method was performed using the extracellular domain portion of mouse cDNA as a probe. As a result, a human clone corresponding to the mouse Troy gene was isolated.
  • the nucleotide sequence of human Troy cDNA is shown in SEQ ID NO: 10
  • the sequence of the protein encoded by the cDNA is shown in FIG.
  • the amino acid sequence is shown in SEQ ID NO: 11.
  • TNFR-associated factor 2
  • TNFR-associated factor 2
  • FuGene6 FuGene6
  • the repo overnight plasmid kB-luc contains five NF-kB binding sites upstream of the basic HTLV-1 promoter dN55 (cancer, 79, 800-804 (1988)) and a luciferase gene downstream. Having. After 24 hours, the cells were washed with PBS, suspended in 250 mM Tris buffer (PH7.8), and frozen and thawed three times to break the cells. Using 0-nitrophenyl -.- D-galactobyranoside as a substrate using the centrifuged supernatant as a sample, the galactosidase activity was measured as described in the literature (Molecular Cloning).
  • luciferase activity was measured using Pitka Gene (manufactured by Toyo Ink). As a result, when mouse Troy was expressed compared to Mock, luciferase activity was induced 3.5 times or more, suggesting that NF-kB was activated by the receptor. On the other hand, expression of the short molecule dTroy hardly induced luciferase activity (Fig. 1). Industrial applicability
  • TNF receptor-like protein and its gene have been provided. These proteins and genes can be used as useful tools for purifying and cloning new factors involved in brain function and skin development, and for screening drug candidate compounds.

Abstract

A gene (mouse Troy) encoding a novel membrane secretory protein belonging to the TNF receptor super family is isolated from a mouse brain cDNA library by using an originally developed method for specifically cloning a gene encoding a membrane secretory protein. This mouse Troy gene is expressed strongly in the brain and skin. Further, another gene (mouse dTroy) encoding a protein derived from the mouse Troy protein by deleting the intracellular domain is isolated. Furthermore, a human gene (human Troy) corresponding to the mouse Troy gene is isolated. Mouse Troy has a function of activating NF-kB, while mouse dTroy has a function of inhibiting this function of mouse Troy.

Description

新規 T N F受容体様 技術分野  New TNF receptor-like technology
本発明は、 新規な受容体タンパク質、 該タンパク質をコードする MA、 該 MA を含むベクタ一、 該ベクタ一を保持する宿主細胞、 該タンパク質に対する抗体、 該タンパク質を利用した化合物のスクリーニング方法、 並びに該スクリーニング 方法により単離しうる化合物に関する。 北  The present invention relates to a novel receptor protein, an MA encoding the protein, a vector containing the MA, a host cell carrying the vector, an antibody against the protein, a method for screening a compound using the protein, It relates to a compound that can be isolated by a screening method. North
-景技  -Science
脳において作用するサイ トカインやその受容体は、 脳に関連した新規な医薬品 開発の上で重要な標的となる。  Cytokines and their receptors that act in the brain are important targets for the development of new brain-related drugs.
従来、 脳において作用するサイ トカインとしては、 nerve growth factor ? (P 01138)、 brain-derived neurotrophic factor (P23560)、 neurotrophin-3 (P207 83)、 neurotrophin-4 (P34130)、 neurotrophin-6 (P34132)、 neurotrophin-6 β (P34133 ), neurotrophin-6 r (P34134)等の栄養因子や leptin (P41160) のよ うなホルモン様サイ トカインなどが報告されている。  Conventionally, cytokines that act on the brain include nerve growth factor? (P 01138), brain-derived neurotrophic factor (P23560), neurotrophin-3 (P207 83), neurotrophin-4 (P34130), and neurotrophin-6 (P34132). Nutritional factors such as neurotrophin-6β (P34133) and neurotrophin-6r (P34134) and hormone-like cytokines such as leptin (P41160) have been reported.
生体内においては、 これら以外にも脳において機能するサイ トカインやその受 容体が多数存在すると考えられ、 その同定およびこれを標的とした医薬品の開発 が望まれる。 発明の開示  It is thought that there are many other cytokines and their receptors that function in the brain in vivo, and it is desired to identify them and develop drugs targeting them. Disclosure of the invention
本発明は、 新規な TNF受容体様タンパク質およびその遺伝子を提供することを 課題とする。 また、 本発明は、 該遺伝子が挿入されたべクタ一、 該ベクタ一を保 持する宿主細胞、 該タンパク質に結合する抗体を提供することを課題とする。 さ らに、 本発明は、 該タンパク質を利用した、 リガンドなどの該タンパク質に結合 するィ t合物をスクリーニングする方法を提供することを課題とする。 An object of the present invention is to provide a novel TNF receptor-like protein and its gene. Another object of the present invention is to provide a vector into which the gene has been inserted, a host cell having the vector, and an antibody that binds to the protein. Sa Furthermore, an object of the present invention is to provide a method of screening for a compound that binds to the protein, such as a ligand, using the protein.
本発明者らは、 脳に発現する新たな因子若しくはその受容体のクローン化を目 指し、 独自に開発したシグナルシーケンストラップ (signal sequence trap; SS T)法 (特願平 9-324912号) を利用して、 脳由来のポリ A RNAを材料に、 分泌 '膜 タンパク質をコードする cDNAのスクリーニングを行った結果、 TNF受容体ス一パ 一ファミリ一に属すると考えられる新規な膜分泌夕ンパク質をコードする遺伝子 を単離することに成功した。 この遺伝子は、 マウス組織の中で、 脳や皮膚におい て強く発現していた。 遺伝子発現の特異性やその構造上の特徴から、 該遺伝子が コードするタンパク質が神経細胞などにおける細胞外から細胞内へのシグナル伝 達分子として機能することが示唆された。 該タンパク質は、 脳機能や皮膚の発達 などに関与する因子の精製やクロ一ニング、 さらには神経関連疾患の医薬品候補 化合物のスクリーニングのためのツールとして有用である。  The present inventors aimed at cloning a new factor expressed in the brain or its receptor, and developed a signal sequence trap (SST) method (Japanese Patent Application No. 9-324912) developed uniquely. Screening of cDNA encoding secreted 'membrane protein using brain-derived poly A RNA as a material, a novel membrane-secreted protein considered to belong to the TNF receptor spa family 1 Was successfully isolated. This gene was highly expressed in brain and skin in mouse tissues. The specificity of gene expression and its structural characteristics suggested that the protein encoded by the gene functions as a signal transduction molecule from extracellular to intracellular in neurons and the like. The protein is useful as a tool for purifying and cloning factors involved in brain function and skin development, and for screening drug candidate compounds for neurological diseases.
本発明は新規な受容体タンパク質およびその遺伝子並びにそれらの用途に関し、 より具体的には、  The present invention relates to novel receptor proteins and their genes and their uses, more specifically,
1 . 下記 (a ) から (d ) のいずれかに記載の DNA、  1. DNA described in any of (a) to (d) below,
( a ) 配列番号: 2または 1 1に記載のアミノ酸配列からなるタンパク質をコー ドする DNA  (a) DNA encoding a protein consisting of the amino acid sequence of SEQ ID NO: 2 or 11
( b ) 配列番号: 1または 1 0に記載の塩基配列のコード領域を含む MA  (b) MA containing the coding region of the nucleotide sequence set forth in SEQ ID NO: 1 or 10
( c ) 配列番号: 2または 1 1に記載のアミノ酸配列において 1若しくは複数の アミノ酸が置換、 欠失、 挿入、 および/または付加したアミノ酸配列からなり、 配列番号: 2または 1 1に記載のアミノ酸配列からなるタンパク質と機能的に同 等なタンパク質をコ一ドする DNA  (c) an amino acid sequence according to SEQ ID NO: 2 or 11, which comprises an amino acid sequence in which one or more amino acids have been substituted, deleted, inserted and / or added in the amino acid sequence according to SEQ ID NO: 2 or 11; DNA that encodes a protein that is functionally equivalent to a protein consisting of a sequence
( d ) 配列番号: 1または 1 0に記載の塩基配列からなる DNAとハイブリダィズ する DNAであって、 配列番号: 2または 1 1に記載のアミノ酸配列からなるタン パク質と機能的に同等なタンパク質をコードする DNA 2. 下記 (a) から (d) のいずれかに記載の DNA、 (d) DNA that hybridizes with the DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or 10, and is functionally equivalent to the protein consisting of the amino acid sequence of SEQ ID NO: 2 or 11 DNA encoding 2. DNA described in any of (a) to (d) below,
( a > "配列番号: 8に記載のァミノ酸配列からなるタンパク質をコードする DNA (a> "DNA encoding a protein consisting of the amino acid sequence of SEQ ID NO: 8
(b) 配列番号: 9に記載の塩基配列のコード領域を含む DNA (b) DNA containing the coding region of the nucleotide sequence set forth in SEQ ID NO: 9
(c) 配列番号: 8に記載のアミノ酸配列において 1若しくは複数のアミノ酸が 置換、 欠失、 挿入、 および/または付加したアミノ酸配列からなり、 配列番号: 9に記載のアミノ酸配列からなるタンパク質と機能的に同等なタンパク質をコー ドする DNA  (c) a protein consisting of the amino acid sequence of SEQ ID NO: 8 with one or more amino acids substituted, deleted, inserted, and / or added, and a protein comprising the amino acid sequence of SEQ ID NO: 9; DNA that encodes an equivalent protein
( d ) 配列番号: 8に記載の塩基配列からなる DNAとハイブリダイズする DNAで あって、 配列番号: 9に記載のアミノ酸配列からなるタンパク質と機能的に同等 な夕ンパク質をコ一ドする DNA  (d) DNA that hybridizes with the DNA consisting of the nucleotide sequence of SEQ ID NO: 8 and that is functionally equivalent to the protein consisting of the amino acid sequence of SEQ ID NO: 9 DNA
3. ( 1) または (2) に記載の DNAが挿入されたベクター、  3. a vector into which the DNA of (1) or (2) has been inserted,
4. (3) に記載のベクタ一が導入された宿主細胞、  4. a host cell into which the vector according to (3) has been introduced,
5. ( 1) または (2) に記載の DNAによりコードされるタンパク質、  5. a protein encoded by the DNA of (1) or (2),
6. (4) に記載の宿主細胞を培養し、 該宿主細胞またはその培養上清から発 現させたタンパク質を回収する工程を含む、 ( 5)に記載のタンパク質の製造方法、 6. A method for producing the protein according to (5), comprising culturing the host cell according to (4), and collecting a protein expressed from the host cell or a culture supernatant thereof.
7. (5) に記載のタンパク質に対する抗体、 7. an antibody against the protein of (5),
8. (5) に記載のタンパク質の部分ペプチド、  8. A partial peptide of the protein according to (5),
9. 配列番号: 1、 8、 または 10に記載の塩基配列からなる DNAまたはその 相補鎖とハイブリダィズし、 少なくとも 15塩基の鎖長を有するヌクレオチド、 9. a nucleotide having a length of at least 15 bases, which hybridizes with a DNA consisting of the nucleotide sequence of SEQ ID NO: 1, 8, or 10 or a complementary strand thereof,
10. (4) に記載のタンパク質に結合する活性を有する化合物のスクリー二 ング方法であって、 10. A method for screening a compound having an activity of binding to a protein according to (4),
(a) (4 )に記載の夕ンパク質またはその部分べプチドに被検試料を接触させる 工程、  (a) contacting the test sample with the protein or its partial peptide according to (4),
(b) (4)に記載のタンパク質またはその部分べプチドに結合する活性を有する 化合物を選択する工程、 を含む方法、  (b) selecting a compound having an activity of binding to the protein or the partial peptide thereof according to (4),
1 1. ( 10) に記載の方法により単離されうる、 (4) に記載のタンパク質に 結合する化合物、 1 1. The protein described in (4), which can be isolated by the method described in (10) A compound that binds,
12.— ( 1) に記載の DNAによりコードされるタンパク質の活性を調節する化 合物のスクリーニング方法であって、  12.—A method for screening a compound that regulates the activity of a protein encoded by the DNA according to (1),
(a) ( 1)に記載の DNAを発現するべクタ一および NF-kBの結合に応答してレポ 一夕一遺伝子を発現するベクターを含む細胞に被検試料を接触させる工程、 (a) contacting a test sample with a cell containing a vector that expresses a repo overnight gene in response to binding of a vector expressing the DNA according to (1) and NF-kB,
(b) 該細胞におけるレポーター活性を検出する工程、 および (b) detecting a reporter activity in the cell, and
( c) 被検試料を該細胞に接触させないで検出した場合と比較して、 該レポ一夕 一活性を低下または増加させる化合物を選択する工程、 を含む方法、  (c) selecting a compound that reduces or increases the repo overnight activity as compared to a case where the test sample is detected without contacting the cells.
13. ( 12) に記載の方法により単離されうる、 (1) に記載の DNAによりコ 一ドされるタンパク質の活性を調節する化合物、  13. A compound that modulates the activity of a protein encoded by the DNA of (1), which can be isolated by the method of (12),
14. 天然由来である、 ( 1 1) または (13) に記載の化合物、  14. The compound according to (11) or (13), which is derived from nature,
15. リガンド、ァゴニスト、またはアン夕ゴニストである、 ( 1 1)または( 1 3) に記載の化合物、 を提供するものである。  15. A compound according to (11) or (13), which is a ligand, an agonist, or an antagonist.
なお、 本発明において、 「リガンド」 とは、 細胞膜に発現する本発明のタンパク 質に結合することにより、 その機能を活性化するタンパク質を指す。  In the present invention, the term “ligand” refers to a protein that activates its function by binding to the protein of the present invention expressed on a cell membrane.
また、 本発明において 「ァゴ二スト」 とは、 本発明のタンパク質とリガンドと の結合と同様の現象(本発明のタンパク質の活性化)を引き起こすことができる、 本発明のタンパク質に特異的に結合できる分子を指す。  In the present invention, “agonist” refers to a protein that can cause a phenomenon similar to the binding of the protein of the present invention to a ligand (activation of the protein of the present invention). Refers to molecules that can bind.
また、 「アン夕ゴニスト」とは、本発明のタンパク質に特異的に結合することに よってその機能を抑制する分子を指す。  In addition, the term “angigonist” refers to a molecule that specifically binds to the protein of the present invention to suppress its function.
本発明は、 新規な膜分泌タンパク質を提供する。 本発明者らにより単離された マウス由来の cDNA (マウス Troyと命名した) の塩基配列を配列番号: 1に、 マ ウス Troy cDNAによりコードされるタンパク質のアミノ酸配列を配列番号: 2に 示す。 また、 マウス Troy cDNAに対応するヒ卜 cDNA (ヒト Troyと命名した) の 塩基配列を配列番号: 10に、 ヒト Troy cDNAによりコードされるタンパク質の アミノ酸配列を配列番号: 1 1に示す。 これらタンパク質は、 TNF 受容体スーパーファミリ一に属する既知のタンパク質 と相 生を有しており、 その N末端にシグナル配列を持ち、 中間部に膜貫通領域 と予想される疎水性に富んだアミノ酸領域を有する。 従って、 これらタンパク質 は、 TNF受容体スーパ一フアミリー (Beulter.B, and Huffel . C. V ( 1994) SCIENCE 264, 667-668 Unravel ing function in the TNF ligand and receptor familie s) に属する新規なタンパク質であると考えられる。 The present invention provides a novel membrane secreted protein. The nucleotide sequence of the mouse-derived cDNA (named mouse Troy) isolated by the present inventors is shown in SEQ ID NO: 1, and the amino acid sequence of the protein encoded by the mouse Troy cDNA is shown in SEQ ID NO: 2. The nucleotide sequence of human cDNA (named human Troy) corresponding to mouse Troy cDNA is shown in SEQ ID NO: 10, and the amino acid sequence of the protein encoded by human Troy cDNA is shown in SEQ ID NO: 11. These proteins are compatible with known proteins belonging to the TNF receptor superfamily, have a signal sequence at the N-terminus, and have a transmembrane region in the middle that is expected to be a highly hydrophobic amino acid region. Having. Therefore, these proteins are novel proteins belonging to the TNF receptor superfamily (Beulter.B, and Huffel.C.V (1994) SCIENCE 264, 667-668 Unraveling function in the TNF ligand and receptor familie s). It is believed that there is.
ノーザンプロット解析の結果、マウス Troyタンパク質をコ一ドする遺伝子は脳 や皮膚に強い発現が見出された。 また、 皮膚の毛包 (hair follicle)や汗腺や歯 の発達に関与すると考えられている Edar( Nature Genetics 1999 vol .22. 370-3 74, 366- 369)と 0X40以上の高い相同性を示した。 従って、 troyタンパク質は脳 機能や皮膚の発達 ·恒常性の維持に関与していることが期待され、 脳機能や皮膚 の発達などに関連する因子の精製やクローニング、 さらには神経関連疾患の治療 薬候補化合物のスクリーニングなどのための有用なッールとなる。  Northern blot analysis showed that the gene encoding mouse Troy protein was strongly expressed in brain and skin. It has a high homology of 0X40 or more with Edar (Nature Genetics 1999 vol.22. 370-374, 366-369), which is considered to be involved in the development of hair follicles, sweat glands and teeth. Was. Therefore, the troy protein is expected to be involved in the maintenance of brain function and skin development and homeostasis, and purification and cloning of factors related to brain function and skin development, as well as therapeutic agents for neurological diseases. This is a useful tool for screening candidate compounds.
本発明者等は、 また、マウス Troyタンパク質の細胞内ドメインのほとんどを欠 失したタンパク質をコ一ドする cDNAクローンを単離した(マウス dTroyと命名し た)。マウス dTroy cDNAの塩基配列を配列番号: 8に、 マウス dTroy cDNAにより コ一ドされるタンパク質のアミノ酸配列を配列番号: 9に示す。該夕ンパク質は、 細胞内ドメインのほとんどを欠失し、 また、 NF- kB活性化能を示さないことから、 細胞膜上に発現し、 リガンドと結合することで、 リガンドの完全型 Troyへの結合 を阻害し、 これによりシグナル伝達を阻害していることが考えられる。 また、 TN FRの仲間はホモ 3量体を形成し、 シグナルを伝達するが、 dTroyを含むヘテロ 3 量体を形成することによりシグナル伝達を阻害することも考えられる。 このよう な機能を有するタンパク質としては、 例えば、 TRAIL- R3 (TRAIL のデコイ型受容 体) や DcR3 (FasLに対するデコイ型受容体) が知られている。  We have also isolated a cDNA clone encoding a protein lacking most of the intracellular domain of the mouse Troy protein (designated mouse dTroy). The nucleotide sequence of mouse dTroy cDNA is shown in SEQ ID NO: 8, and the amino acid sequence of the protein encoded by mouse dTroy cDNA is shown in SEQ ID NO: 9. Since the protein lacks most of the intracellular domain and does not show NF-KB activation ability, it is expressed on the cell membrane and binds to the ligand to convert the complete ligand to Troy. It is considered that binding is inhibited, and thereby signal transmission is inhibited. In addition, TNFR members form homotrimers and transmit signals, but may inhibit signal transmission by forming heterotrimers containing dTroy. As proteins having such functions, for example, TRAIL-R3 (TRAIL decoy-type receptor) and DcR3 (FasL decoy-type receptor) are known.
本発明は、 また、マウスおよびヒト Troyタンパク質と機能的に同等なタンパク 質を包含する。 ここで 「機能的に同等」 とは、 対象となるタンパク質が細胞外か ら細胞内へのシグナル伝達を行なう受容体タンパク質として機能することを指す。 この うな受容体タンパク質として機能としては、 例えば、 NF- kB 活性誘導能が 挙げられる。 NF- kB 活性誘導能は、 実施例 6に記載の方法により検出することが 可能である。 The invention also includes proteins that are functionally equivalent to the mouse and human Troy proteins. Here, “functionally equivalent” means that the target protein is extracellular. To function as a receptor protein that performs signal transduction into cells. The function as such a receptor protein includes, for example, the ability to induce NF-kB activity. The ability to induce NF-kB activity can be detected by the method described in Example 6.
また、 本発明は、 マウス dTroyタンパク質と機能的に同等なタンパク質を包含 する。 ここで 「機能的に同等」 とは、 対象となるタンパク質がデコイ型受容体夕 ンパク質として機能することを指す。 デコイ型受容体タンパク質として機能とし ては、 例えば、 完全型受容体タンパク質 (例えば、 Troyタンパク質) の NF- kB活 性誘導能を阻害する機能が挙げられる。  The invention also includes proteins functionally equivalent to the mouse dTroy protein. Here, “functionally equivalent” means that the target protein functions as a decoy receptor protein. The function as a decoy-type receptor protein includes, for example, a function of inhibiting the ability of a full-type receptor protein (for example, Troy protein) to induce NF-kB activity.
このようなタンパク質を単離するための、当業者によく知られた方法としては、 タンパク質に変異を導入する方法が知られている。 例えば、 当業者であれば、 部 位特異的変異誘発法 (Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nak agawa, M. ( 1995 ) An oligodeoxyribonucleotide - directed dual amber method for site-directed mutagenesis. Gene 152, 27ト 275、 Zoller, MJ, and Smith, As a method well known to those skilled in the art for isolating such a protein, a method of introducing a mutation into the protein is known. For example, those skilled in the art will recognize that site-directed mutagenesis (Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis.Gene 152, 27 275, Zoller, MJ, and Smith,
M. ( 1983 ) Oligonucleotide - directed mutagenesis of DNA fragments cloned i nto M13 vectors. Methods Enzymol . 100, 468 - 500、 Kramer, , Drutsa,V, Janse n具 Kramer, B, Pflugf elder, M, and Fritz,HJ( 1984) The gapped duplex DNA approach to ol igonucleotide - directed mutation construction. Nucleic Aci ds Res. 12, 9441-9456、 Kramer W, and Fritz HJ( 1987) Oligonucleotide- dire cted construction of mutations via gapped duplex DNA Methods. Enzymol . 1 54, 350 - 367、 Kunkel, TA( 1985 ) Rapid and efficient site-specific mutagenes is without phenotypic selection. Proc Natl Acad Sci U S A. 82, 488- 492)な どを用いて、 マウスおよびヒトの Troyタンパク質のアミノ酸配列(配列番号: 2 または 1 1のアミノ酸配列) またはマウス dTroyタンパク質のアミノ酸配列 (配 列番号: 9のアミノ酸配列) に適宜変異を導入することにより、 これらタンパク 質と機能的に同等なタンパク質を調製することができる。 また、 アミノ酸の変異 は自然界においても生じうる。このように、マウスおよびヒ卜の Troyタンパク質 また ίまマゥス dTroyタンパク質のァミノ酸配列において 1もしくは複数のァミノ 酸が変異したアミノ酸配列を有し、 これらタンパク質と機能的に同等なタンパク 質もまた本発明のタンパク質に含まれる。 このような変異体における、 変異する アミノ酸数は、 通常、 30アミノ酸以内であり、 好ましくは、 15アミノ酸以内であ り、 さらに好ましくは 5アミノ酸以内であり、 さらに好ましくは 3アミノ酸以内 であると考えられる。 M. (1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors.Methods Enzymol. 100, 468-500, Kramer,, Drutsa, V, Jansen Kramer, B, Pflugf elder, M, and Fritz, HJ (1984) The gapped duplex DNA approach to ol igonucleotide-directed mutation construction.Nucleic Acids Res. 12, 9441-9456, Kramer W, and Fritz HJ (1987) Oligonucleotide-directed construction of mutations via gapped duplex DNA Methods.Enzymol 1 54, 350-367, Kunkel, TA (1985) Rapid and efficient site-specific mutagenes is without phenotypic selection. Proc Natl Acad Sci US A. 82, 488-492). Functionally equivalent to these proteins by introducing appropriate mutations into the amino acid sequence of the protein (SEQ ID NO: 2 or 11) or the amino acid sequence of mouse dTroy protein (SEQ ID NO: 9) Preparing Various Proteins It is possible. Amino acid mutation Can also occur in nature. Thus, proteins having one or more mutated amino acids in the amino acid sequence of the mouse and human Troy protein or Pomatus dTroy protein, and functionally equivalent to these proteins, are also present in the present invention. Included in the proteins of the invention. The number of amino acids to be mutated in such a mutant is generally considered to be within 30 amino acids, preferably within 15 amino acids, more preferably within 5 amino acids, and further preferably within 3 amino acids. Can be
また、 変異するアミノ酸残基においては、 アミノ酸側鎖の性質が保存されてい る別のアミノ酸に変異されることが望ましい。 例えばァミノ酸側鎖の性質として は、 疎水性アミノ酸 (A、 I、 L、 M、 F、 P、 W、 Y、 V)、 親水性アミノ酸 (R、 D、 N、 C、 E、 Q、 G、 H、 K、 S、 T)、 脂肪族側鎖を有するアミノ酸 (G、 A、 V、 L、 I、 P)、 水酸基含有側鎖を有するアミノ酸 (S、 Τ、 Υ)、 硫黄原子含有側鎖を有するァミノ 酸 ( Μ)、 カルボン酸及びアミ ド含有側鎖を有するアミノ酸 (D、 N、 E、 Q)、 塩 基含有側鎖を有するアミノ酸 (R、 K、 Η)、 芳香族含有側鎖を有するアミノ酸(Η、 F、 Y、 W)を挙げることができる(括弧内はいずれもアミノ酸の一文字標記を表す)。  In addition, it is desirable that the amino acid residue to be mutated is mutated to another amino acid in which the properties of the amino acid side chain are conserved. For example, the properties of the amino acid side chain include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V) and hydrophilic amino acids (R, D, N, C, E, Q, G , H, K, S, T), amino acids having aliphatic side chains (G, A, V, L, I, P), amino acids having hydroxyl-containing side chains (S, Τ, Υ), sulfur atom-containing side Amino acids (D, N, E, Q) with carboxylic acids and amide-containing side chains, amino acids with side chains containing bases (R, K, 塩), aromatic-containing Amino acids having a chain (Η, F, Y, W) can be mentioned (all brackets indicate one letter of amino acids).
1又は複数個のアミノ酸残基が付加された蛋白質としては、 例えば、 本発明の Troy夕ンパク質または dTroy夕ンパク質を含む融合蛋白質が挙げられる。融合蛋 白質は、 これらタンパク質と他のぺプチド又は蛋白質とが融合したものであり、 本発明に含まれる。 融合蛋白質を作製する方法は、 本発明のタンパク質をコード する DNAと他のぺプチド又は蛋白質をコ一ドする DNAをフレームが一致するよう に連結してこれを発現べクタ一に導入し、 宿主で発現させればよく、 すでに公知 の手法を用いることができる。 本発明の蛋白質との融合に付される他のぺプチド 又は蛋白質は特に限定されない。  Examples of the protein to which one or more amino acid residues are added include a fusion protein containing Troy protein or dTroy protein of the present invention. The fusion protein is a fusion of these proteins with other peptides or proteins, and is included in the present invention. The method for producing a fusion protein is as follows: a DNA encoding the protein of the present invention and another peptide or a DNA encoding the protein are ligated so that their frames coincide with each other, and this is introduced into an expression vector; And a known method can be used. Other peptides or proteins to be fused with the protein of the present invention are not particularly limited.
また、 機能的に同等なタンパク質を単離する当業者によく知られた他の方法と しては、 ハイブリダィゼーシヨン技術 (Sambrook,J et al ., Molecular Cloning 2nd ed., 9.47-9.58, Cold Spring Harbor Lab. press, 1989) を利用した方法 が挙げられる。即ち、 当業者であれば、 マウス若しくはヒトの Troyタンパク質ま たはマウス dTroyタンパク質をコードする DNA配列 (配列番号: 1、 8、 または 1 0 ) もしくはその一部を基に、 これと相同性の高い DNAを単離して、 該 DNAか らこれらタンパク質と機能的に同等なタンパク質を単離することも通常行いうる ことである。 このようなハイプリダイゼ一シヨン技術を利用して単離された MA がコードするタンパク質もまた本発明のタンパク質に含まれる。 このようなタン パク質としては、 例えば、 哺乳動物のホモログ (例えば、 ヒトおよびマウス以外 の哺乳動物由来の Troyタンパク質やマウス以外の哺乳動物由来の dTroyタンパク 質) が挙げられる。 Other methods well known to those skilled in the art for isolating functionally equivalent proteins include the hybridization technique (Sambrook, J et al., Molecular Cloning 2nd ed., 9.47-9.58). , Cold Spring Harbor Lab. Press, 1989) Is mentioned. That is, those skilled in the art will recognize, based on the DNA sequence encoding mouse or human Troy protein or mouse dTroy protein (SEQ ID NOS: 1, 8, or 10) or a portion thereof, It is also usually possible to isolate high DNA and to isolate proteins functionally equivalent to these proteins from the DNA. The protein encoded by MA isolated using such a hybridization technique is also included in the protein of the present invention. Such proteins include, for example, mammalian homologs (eg, Troy protein derived from mammals other than human and mouse, and dTroy protein derived from mammals other than mouse).
機能的に同等なタンパク質をコードする DNAを単離するためのハイプリダイゼ ーシヨンは、 例えば、 ストリンジエンシーが、 10%ホルムアミ ド、 5xSSPE、 lxデ ンハルト溶液、 lxサケ精子 MAの条件で行うことができる。より好ましい条件(よ りストリンジェントな条件) としては、 25%ホルムアミ ド、 5xSSPE、 lxデンハル ト溶液、 lxサケ精子 DNAの条件であり、 さらに好ましい条件(さらにストリンジ ェントな条件) としては、 50%ホルムアミ ド、 5xSSPE、 lx デンハルト溶液、 lx サケ精子 DNAの条件である。 但し、 ハイプリダイゼーシヨンのストリンジェンシ 一に影響する要素としては上記ホルムアミ ド濃度以外にも複数考えられ、 当業者 であればこれら要素を適宜選択することで同様のストリンジエンシーを実現する ことが可能である。 また、 ハイプリダイゼーシヨンにかえて、 タンパク質をコ一 ドする DNA (配列番号: 1、 8、 または 1 0 ) の一部をプライマ一に用いる遺伝 子増幅法、 例えば、 PCR法を利用して単離することも可能である。  Hybridization to isolate DNA encoding a functionally equivalent protein can be performed, for example, under conditions where the stringency is 10% formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm MA. . More preferred conditions (more stringent conditions) are 25% formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm DNA, and more preferred conditions (more stringent conditions) are 50% The conditions are formamide, 5xSSPE, lx Denhardt's solution, and lx salmon sperm DNA. However, there may be a plurality of factors affecting the stringency of the hybridization other than the above-mentioned formamide concentration, and those skilled in the art can realize similar stringency by appropriately selecting these factors. Is possible. In addition, instead of using hybridization, a gene amplification method using a part of DNA (SEQ ID NO: 1, 8, or 10) encoding a protein as a primer, for example, a PCR method is used. It is also possible to isolate.
これらハイブリダィゼ一シヨン技術または遺伝子増幅技術により単離される D NAがコードするタンパク質は、 通常、 配列番号: 2、 9、 または 1 1に記載の本 発明のタンパク質とアミノ酸配列において高い相同性を有する。高い相同性とは、 通常、 70%以上の相同性、好ましくは 85%以上の相同性、さらに好ましくは 95% 以上の相同性である。 蛋白質の相同性を決定するには、 文献 (Wi lbur, W. J. an d Lipman, D. J. Proc. Natl. Acad. Sci . USA ( 1983) 80, 726- 730)に記載のァ ルゴサズムにしたがえばよい。 The DNA encoded by the DNA isolated by the hybridization technique or the gene amplification technique usually has a high homology in amino acid sequence with the protein of the present invention described in SEQ ID NO: 2, 9, or 11. High homology usually means a homology of 70% or more, preferably 85% or more, more preferably 95% or more. To determine protein homology, the literature (Wilbur, WJ an d Lipman, DJ Proc. Natl. Acad. Sci. USA (1983) 80, 726-730).
本発明のタンパク質は、 当業者に公知の方法により、 組み換えタンパク質とし て、 また天然のタンパク質として調製することが可能である。 組み換えタンパク 質であれば、 例えば、 本発明のタンパク質の膜結合に必要な領域以外の部分を、 可溶性タンパク質として細胞外に分泌させる。 次いで、 この細胞の培養上清を回 収し、 濃縮した後、 イオン交換、 逆相、 ゲル濾過などのクロマトグラフィー、 あ るいは本発明の夕ンパク質に対する抗体をカラムに固定したァフィニティ一クロ マトグラフィ一にかけることにより、 または、 さらにこれらのカラムを複数組み 合わせることにより精製することが可能である。 また、 本発明のタンパク質をグ ル夕チオン S トランスフェラーゼタンパク質との融合タンパク質として、 あるい はヒスチジンを複数付加させた組み換えタンパク質として宿主細胞 (例えば、 動 物細胞や大腸菌など) 内で発現させ、 発現させた組み換えタンパク質をグル夕チ オンカラム、 あるいはニッケルカラムにより精製する。 その後、 必要に応じて融 合タンパク質のうち目的のタンパク質以外の領域を、 トロンビンまたはファクタ —Xaなどにより切断し、 除去する方法により調製できる。天然のタンパク質であ れば、 例えば、 本発明のタンパク質を発現している細胞の抽出物に対し、 後述す る本発明の抗体が結合したァフィ二ティーカラムを作用させて精製することによ り単離することができる。  The protein of the present invention can be prepared as a recombinant protein or as a natural protein by methods known to those skilled in the art. In the case of a recombinant protein, for example, a portion other than the region required for membrane binding of the protein of the present invention is extracellularly secreted as a soluble protein. Next, the culture supernatant of the cells is collected and concentrated, and then subjected to chromatography such as ion exchange, reverse phase, gel filtration, or affinity chromatography in which the antibody against the protein of the present invention is immobilized on a column. Purification can be carried out by combining them or by combining a plurality of these columns. In addition, the protein of the present invention is expressed in a host cell (for example, an animal cell or Escherichia coli) as a fusion protein with a glutathione S-transferase protein or as a recombinant protein to which a plurality of histidines are added. The recombinant protein is purified using a glutathione column or a nickel column. Thereafter, if necessary, a region other than the target protein in the fusion protein can be prepared by a method of cleaving with thrombin or Factor-Xa and removing it. If the protein is a natural protein, it is purified by, for example, allowing an extract of cells expressing the protein of the present invention to act on an affinity column to which the antibody of the present invention described later is bound. Can be isolated.
本発明は、 また、 本発明の蛋白質の部分ペプチドを包含する。 本発明の部分べ プチドは、 少なくとも 8アミノ酸以上、 好ましくは 15アミノ酸以上、 さらに好ま しくは 50アミノ酸以上 (例えば、 100アミノ酸以上) のアミノ酸配列からなる。 該部分ペプチドは、 例えば、 本発明の蛋白質に対する抗体の作製、 本発明のタン パク質に結合するリガンドなどの化合物のスクリ一ニングゃ、 本発明のタンパク 質の競合阻害剤の調製などに利用し得る。 本発明の部分ペプチドとしては、 例え ば、 本発明の夕ンパク質からシグナルべプチドが除去された形態の夕ンパク質や 膜結合に必要な領域を除去した可溶性タンパク質、 さらには、 細胞内の領域を除 去しご形態のタンパク質が含まれる。 また、 例えば、 リガンドとの結合能を有す るが細胞内へのシグナル伝達を行う能力のない部分べプチド (本発明の受容体の 競合阻害剤として機能する) が含まれる。 本発明の部分ペプチドは、 遺伝子工学 的手法、 公知のペプチド合成法、 あるいは本発明の蛋白質を適切なぺプチダーゼ で切断することによって製造することができる。 The present invention also includes partial peptides of the protein of the present invention. The partial peptide of the present invention has an amino acid sequence of at least 8 amino acids or more, preferably 15 amino acids or more, more preferably 50 amino acids or more (for example, 100 amino acids or more). The partial peptide is used, for example, for preparing an antibody against the protein of the present invention, screening a compound such as a ligand that binds to the protein of the present invention, and preparing a competitive inhibitor of the protein of the present invention. obtain. The partial peptide of the present invention includes, for example, the protein of the present invention in which the signal peptide is removed from the protein and the protein of the present invention. This includes soluble proteins from which the region required for membrane binding has been removed, and proteins in the form of corn by removing the intracellular region. In addition, for example, a partial peptide (which functions as a competitive inhibitor of the receptor of the present invention) that has the ability to bind to a ligand but does not have the ability to transmit signals into cells is included. The partial peptide of the present invention can be produced by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
また、 本発明は、 本発明のタンパク質をコードする DNAを提供する。 本発明の DNA は、 本発明のタンパク質をコードしうるものであれば、 いかなる形態でもよ い。即ち、 mRNAから合成された cDNAであるか、 ゲノム DNAであるか、 化学合成!) NAであるかなどを問わない。 また、 本発明のタンパク質をコードしうる限り、 遺 伝暗号の縮重に基づく任意の塩基配列を有する MAが含まれる。  The present invention also provides a DNA encoding the protein of the present invention. The DNA of the present invention may be in any form as long as it can encode the protein of the present invention. That is, whether it is cDNA synthesized from mRNA, genomic DNA, or chemical synthesis! It does not matter whether it is NA or not. Also, as long as it can encode the protein of the present invention, MA having an arbitrary base sequence based on the degeneracy of the genetic code is included.
本発明の DNAは、 当業者に公知の方法により調製することができる。 例えば、 本発明のタンパク質を発現している細胞より cDNAライブラリ一を作製し、本発明 の DNAの配列 (例えば、 配列番号: 1、 8、 または 1 0に記載の DNA配列) の一 部をプローブにしてハイブリダィゼーシヨンを行うことにより調製できる。また、 本発明のタンパク質を発現している細胞より RNAを調製し、 本発明の DNAの配列 (例えば、 配列番号: 1、 8、 または 1 0に記載の DNA配列) に基づいてオリゴ DNAを合成し、 これをプライマーとして用いて PCR反応を行い、 本発明のタンパ ク質をコードする cDNAを増幅させることにより調製することも可能である。 本発明の DNAは、 本発明の夕ンパク質を組み換えタンパク質として生産するた めに利用することができる。 また、 本発明のタンパク質をコードする DNAに欠陥 がある場合には、 アンチセンスによる機能阻害や、 正常な遺伝子に置き換える遺 伝子治療などへの応用も考えられる。  The DNA of the present invention can be prepared by a method known to those skilled in the art. For example, a cDNA library is prepared from cells expressing the protein of the present invention, and a part of the DNA sequence of the present invention (for example, the DNA sequence of SEQ ID NO: 1, 8, or 10) is probed. And by performing hybridization. In addition, RNA is prepared from cells expressing the protein of the present invention, and oligo DNA is synthesized based on the sequence of the DNA of the present invention (for example, the DNA sequence described in SEQ ID NO: 1, 8, or 10). However, it can also be prepared by performing a PCR reaction using this as a primer and amplifying a cDNA encoding the protein of the present invention. The DNA of the present invention can be used to produce the protein of the present invention as a recombinant protein. If the DNA encoding the protein of the present invention is defective, application to antisense function inhibition, gene therapy for replacement with a normal gene, and the like can be considered.
また、 本発明は、 本発明の DNAが挿入されたベクターを提供する。 本発明のベ クタ一としては、 宿主細胞内において本発明の DNAの発現させうるものであれば 特に制限はない。 例えば、 宿主を大腸菌とする場合には、 ベクターを大腸菌 (例 えば、 JM109、 DH5ひ、 HB10U XLlBlue) などで大量に増幅させ大量調製するため に、 大腸菌で増幅されるための「ori」 をもち、 さらに形質転換された大腸菌の選 抜遺伝子 (例えば、 なんらかの薬剤 (アンピシリンやテトラサイクリン、 カナマ イシン、 クロラムフエ二コール) により判別できるような薬剤耐性遺伝子) を有 すれば特に制限はない。 例えば、 M13系べクタ一、 pUC系べクタ一、 pBR322、 pBl uescript, pCR- Scriptなどが挙げられる。 また cDNAのサブクローニング、 切り 出しを目的とした場合も上記ベクターの他に、 pGEM- T、 pDIRECT、 pT7などがあげ られる。 本発明のタンパク質を生産する目的においてべクタ一を使用する場合に は、 特に、 発現べクタ一が有用である。 発現ベクターとしては、 例えば、 大腸菌 での発現を目的とした場合は、 ベクタ一が大腸菌で増幅されるような上記特徴を 持つほかに、 宿主を JM109、 DH5ひ、 HB10K XLlBlueなどの大腸菌とした場合にお いては、大腸菌で効率よく発現できるようなプロモ一夕一(例えば、 lac , T7など) を持っていることが不可欠である。 このようなベクタ一としては、 上記ベクター の他に pGEX、 pEGFP、 または pET (この場合、宿主は T7 MAポリメラ一ゼを発現し ている BL21が好ましい)などが挙げられる。 The present invention also provides a vector into which the DNA of the present invention has been inserted. The vector of the present invention is not particularly limited as long as it can express the DNA of the present invention in a host cell. For example, when the host is E. coli, the vector is For example, JM109, DH5, and HB10U XLlBlue) have an “ori” to be amplified in Escherichia coli for large-scale amplification and preparation in Escherichia coli, and a selection gene for transformed Escherichia coli (for example, any drug (A drug resistance gene that can be identified by ampicillin, tetracycline, kanamycin, chloramphenicol) is not particularly limited. For example, M13 system vector, pUC system vector, pBR322, pBluescript, pCR-Script, and the like. For the purpose of cDNA subcloning and excision, pGEM-T, pDIRECT, pT7, etc. may also be used in addition to the above vectors. When a vector is used for the purpose of producing the protein of the present invention, an expression vector is particularly useful. As an expression vector, for example, when the purpose is expression in E. coli, in addition to having the above characteristics such that the vector is amplified in E. coli, when the host is E. coli such as JM109, DH5, HB10K XLlBlue, etc. It is essential to have a promoter (eg, lac, T7, etc.) that can be efficiently expressed in E. coli. Examples of such a vector include pGEX, pEGFP, or pET (in this case, the host is preferably BL21 expressing T7 MA polymerase) in addition to the above vectors.
また、 CH0細胞、 COS細胞 、 NIH3T3細胞等の動物細胞での発現を目的とした場 合には、 細胞内で発現させるために必要なプロモ一夕一 (SV40,MMLV- LT1,EF1ひ, CMV プロモーターなど) を持っていることが不可欠であり、 細胞への形質転換を 選抜するための遺伝子 (例えば、 薬剤 (ネオマイシン、 G418など) により判別で きるような薬剤耐性遺伝子) を有すればさらに好ましい。 このような特性を有す るべクタ一としては、 例えば、 p匪、 pDR2、 pBK- RSV、 pBK-CMVヽ pOPRSV, pOP13 などが挙げられる。  In addition, when the expression is intended for expression in animal cells such as CH0 cells, COS cells, and NIH3T3 cells, the promoters required for expression in cells (SV40, MMLV-LT1, EF1, CMV It is essential to have a promoter, etc., and it is more preferable to have a gene for selecting transformation into cells (for example, a drug resistance gene that can be distinguished by a drug (neomycin, G418, etc.)). . Vectors having such characteristics include, for example, bandits, pDR2, pBK-RSV, pBK-CMV ヽ pOPRSV, and pOP13.
さらに、 細胞内でのコピー数の増幅を目的とした宿主ベクター系においては、 安定産生細胞株の場合は、核酸合成経路を欠損した CH0細胞にそれを相補する DH FR遺伝子を有するベクタ一 (例えば、 pCHOIなど) を用いメ トトレキセ一ト (MT X)により増幅させる方法が挙げられ、 また一過性の発現を目的とする場合には、 SV40 T抗原を染色体上に持つ COS細胞を用いて SV40の複製機転を持- (pcD"など) で形質転換する方法が挙げられる。 Furthermore, in a host vector system for the purpose of amplifying the copy number in a cell, in the case of a stable production cell line, a vector having a DHFR gene that complements a nucleic acid synthesis pathway-deficient CH0 cell (eg, , PCHOI, etc.) and amplifying with methotrexate (MTX). In the case of transient expression, There is a method in which COS cells having the SV40 T antigen on the chromosome are used to transform the cells with the replication mechanism of SV40 (such as pcD ").
一方、 動物の生体内で本発明の DNAを発現させる方法としては、 本発明の DNA を適当なベクターに組み込みレトロウイルス法、 リボソーム法、 カチォニックリ ポソ一ム法、 アデノウイルス法などにより生体内に導入する方法などが挙げられ る。用いられるベクターに特に制限はないが、 pAdexlcwや pZIPneoなどが挙げら れる。 ベクターへの本発明の DNAの挿入などの一般的な遺伝子操作は、 常法に従 つて行うことが可能である (Molecular Cloning ,5.6卜 5.63)。  On the other hand, as a method for expressing the DNA of the present invention in an animal living body, the DNA of the present invention is incorporated into an appropriate vector, and the retroviral method, the ribosome method, the cationic liposome method, the adenovirus method, and the like are used to express the DNA in vivo. There is a method for introduction. The vector to be used is not particularly limited, and examples include pAdexlcw and pZIPneo. General genetic manipulation such as insertion of the DNA of the present invention into a vector can be performed according to a conventional method (Molecular Cloning, 5.6, 5.63).
また、 本発明は、 本発明のベクターが導入された宿主細胞を提供する。 本発明 のベクターが導入される宿主細胞としては特に制限はなく、例えば、大腸菌や種々 の動物細胞などを用いることが可能である。大腸菌では、 例えば、 JM109、 DH5ひ、 HB101 等が挙げられ、 動物細胞においては、 例えば、 CH0細胞、 COS細胞、 3T3細 胞、 HeLa細胞などが挙げられる。 動物細胞において、 大量発現を目的とする場合 には特に CH0細胞が好ましい。 宿主細胞へのベクタ一の導入は、 例えば、 リン酸 カルシウム法、 DEAEデキストラン法、 エレクト口ポレーシヨン、 リポフエクショ ンなどの方法で行うことが可能である。  The present invention also provides a host cell into which the vector of the present invention has been introduced. The host cell into which the vector of the present invention is introduced is not particularly limited, and for example, Escherichia coli and various animal cells can be used. Examples of Escherichia coli include JM109, DH5 and HB101, and examples of animal cells include CH0 cells, COS cells, 3T3 cells, and HeLa cells. In animal cells, when large-scale expression is intended, CH0 cells are particularly preferred. The introduction of the vector into a host cell can be carried out by, for example, a calcium phosphate method, a DEAE dextran method, electo-portation, lipofection, and the like.
また、 本発明は、 本発明のタンパク質と結合する抗体を提供する。 本発明の抗 体の形態には、 特に制限はなく、 ポリクローナル抗体の他、 モノクローナル抗体 も含まれる。 また、 ゥサギなどに本発明のタンパク質を免疫して得た抗血清、 す ベてのクラスのポリクロ一ナル抗体およびモノクローナル抗体、 さらにヒト抗体 や遺伝子組み換えによるヒト型化抗体も含まれる。  The present invention also provides an antibody that binds to the protein of the present invention. The form of the antibody of the present invention is not particularly limited, and includes a monoclonal antibody as well as a polyclonal antibody. Also included are antisera obtained by immunizing egrets and the like with the protein of the present invention, all classes of polyclonal and monoclonal antibodies, and human antibodies and humanized antibodies obtained by genetic recombination.
本発明の抗体は、 以下の方法により調製することが可能である。 ポリクロ一ナ ル抗体であれば、 例えば、 本発明のタンパク質をゥサギなどの小動物に免疫し血 清を得て、 これを本発明のタンパク質をカツプリングさせたァフィ二ティーカラ ムにより、 本発明のタンパク質のみを認識する画分を得て、 さらにこの画分から 免疫グロブリン Gあるいは Mを、 プロテイン A、 あるいはプロテイン Gカラムに より精製することにより調製することができる。 また、 モノクローナル抗体であ れば、一本発明のタンパク質をマウスなどの小動物に免疫を行い、 同マウスより脾 臓を摘出し、 これをすりつぶして細胞にし、 マウスミエローマ細胞とポリエチレ ングリコールなどの試薬により融合させ、 これによりできた融合細胞 (ハイプリ ドーマ) の中から、 本発明のタンパク質に対する抗体を産生するクローンを選択 する。 次いで、 得られたハイブリ ド一マをマウス腹腔内に移植し、 同マウスより 腹水を回収し、 得られたモノクローナル抗体を、 例えば、 硫安沈殿、 プロテイン A、 プロテイン Gカラム、 DEAEイオン交換クロマトグラフィー、 本発明のタンパ ク質をカツプリングしたァフィ二ティーカラムなどにより精製することで調製す ることが可能である。 本発明の抗体は、 本発明のタンパク質の精製、 検出に用い られる他、 本発明のタンパク質のァゴニストやアン夕ゴニストの候補になる。 ま た、 本発明の抗体を神経関連疾患の抗体治療へ応用することも考えられる。 抗体 治療に用いる場合には、 免疫原性を低下させるため、 ヒト抗体やヒト型抗体が好 ましい。 The antibody of the present invention can be prepared by the following method. In the case of a polyclonal antibody, for example, a small animal such as a heron is immunized with the protein of the present invention to obtain a serum, which is then subjected to an affinity column in which the protein of the present invention is coupled. Of immunoglobulin G or M from this fraction was applied to a protein A or protein G column. It can be prepared by further purification. In the case of a monoclonal antibody, a small animal such as a mouse is immunized with the protein of the present invention, the spleen is excised from the mouse, and the spleen is crushed into cells, and mouse myeloma cells and reagents such as polyethylene glycol are used. And a clone that produces an antibody against the protein of the present invention is selected from the resulting fused cells (hybridomas). Next, the obtained hybridoma was transplanted into a mouse intraperitoneal cavity, ascites was recovered from the mouse, and the obtained monoclonal antibody was subjected to, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, It can be prepared by purifying the protein of the present invention using a coupled affinity column or the like. The antibody of the present invention is used for purification and detection of the protein of the present invention, and is also a candidate for an agonist and an agonist of the protein of the present invention. It is also conceivable to apply the antibody of the present invention to antibody therapy for neurological diseases. When used for antibody therapy, human or humanized antibodies are preferred to reduce immunogenicity.
また、 ヒト抗体遺伝子のレパートリ一を有するトランスジエニック動物に抗原 となる蛋白質、蛋白質発現細胞又はその溶解物を免疫して抗体産生細胞を取得し、 これをミエローマ細胞と融合させたハイプリ ドーマを用いて蛋白質に対するヒト 抗体を取得してもよい (国際公開番号 W092- 03918、 W093-2227, W094-02602, W09 4-25585、 W096-33735および W096- 34096参照)。  Further, a transgenic animal having a repertoire of human antibody genes is immunized with a protein serving as an antigen, a protein-expressing cell or a lysate thereof to obtain an antibody-producing cell, and a hybridoma obtained by fusing the antibody-producing cell with myeloma cells is used. To obtain a human antibody against the protein (see International Publication Nos. W092-03918, W093-2227, W094-02602, W094-25585, W096-33735 and W096-34096).
さらに、 本発明の抗体は、 本発明の蛋白質に結合するかぎり、 その抗体断片や 抗体修飾物であってよい。例えば、 抗体断片としては、 Fab、 F(abr )2、 Fv又は H 鎖と L鎖の Fvを適当なリンカーで連結させたシングルチェイン Fv(scFv) (Husto n, J. S. et al . , Proc . Natl . Acad. Sci . U. S.A. ( 1988) 85, 5879-5883 ) が 挙げられる。 具体的には、 抗体を酵素、 例えば、 パパイン、 ペプシンで処理し抗 体断片を生成させるか、 又は、 これら抗体断片をコードする遺伝子を構築し、 こ れを発現ベクターに導入した後、 適当な宿主細胞で発現させる (例えば、 Co, M. S. et al ., J. Immunol . ( 1994) 152, 2968-2976 ; Better, M. and Hor itz, A. H.-7 Methods Enzymol . ( 1989) 178, 476-496 ; Pluckthun, A. and Skerra, A. , Methods Enzymol . ( 1989) 178, 497-515 ; Lamoyi, E., Methods Enzymol . ( 1986 ) 121, 652-663 ; Rousseaux, J. et al . , Methods Enzymol . ( 1986 ) 121 ,Furthermore, the antibody of the present invention may be an antibody fragment or modified antibody thereof as long as it binds to the protein of the present invention. For example, as an antibody fragment, Fab, F (ab r ) 2, Fv or a single chain Fv (scFv) in which an Fv of an H chain and an L chain are linked by an appropriate linker (Huston, JS et al., Proc. Natl. Acad. Sci. USA (1988) 85, 5879-5883). Specifically, the antibody is treated with an enzyme, for example, papain or pepsin, to generate an antibody fragment, or a gene encoding these antibody fragment is constructed and introduced into an expression vector. Expression in host cells (eg, Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horitz, AH-7 Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra, A. (1989) 178, 497-515; Lamoyi, E., Methods Enzymol. (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol. (1986) 121,
663-669 ; Bird, R. E. and Walker, B. W., Trends Biotechnol . ( 1991 ) 9, 1 32- 137参照)。 663-669; Bird, R.E. and Walker, B.W., Trends Biotechnol. (1991) 9, 132-137).
抗体修飾物として、 ポリエチレングリコール(PEG)等の各種分子と結合した抗 体を使用することもできる。 本発明の 「抗体」 にはこれらの抗体修飾物も包含さ れる。 このような抗体修飾物を得るには、 得られた抗体に化学的な修飾を施すこ とによって得ることができる。 これらの方法はこの分野において既に確立されて いる。  As the modified antibody, an antibody bound to various molecules such as polyethylene glycol (PEG) can be used. The “antibody” of the present invention also includes these modified antibodies. Such a modified antibody can be obtained by subjecting the obtained antibody to chemical modification. These methods are already established in this field.
本発明はまた、 配列番号: 1、 8、 または 1 0に示される塩基配列からなる DN Aまたはその相補鎖とハイブリダィズし、少なくとも 15塩基の鎖長を有するヌク レオチドを提供する。 このようなヌクレオチドは、 好ましくは、 配列番号: 1、 8、 または 1 0に示される塩基配列からなる DNAまたはその相補鎖と特異的にハ イブリダィズするヌクレオチドである。 ここで 「特異的にハイブリダィズ」 する とは、 通常のハイブリダィゼーシヨン条件下、 好ましくはストリンジェントなハ ィブリダィゼーシヨン条件下で、 他の蛋白質をコ一ドする DNAとのクロスハイブ リダィゼーシヨンが有意に生じないことを意味する。 このようなヌクレオチドに は、 本発明のタンパク質をコードする DNA又はその相補鎖と特異的にハイブリダ ィズし得るプローブやプライマー、 ヌクレオチド又はヌクレオチド誘導体 (例え ばアンチセンスオリゴヌクレオチドゃリボザィム等) が含まれる。  The present invention also provides a nucleotide that hybridizes with a DNA consisting of the nucleotide sequence shown in SEQ ID NO: 1, 8, or 10, or a complementary strand thereof, and has a length of at least 15 bases. Such a nucleotide is preferably a nucleotide that specifically hybridizes with a DNA consisting of the nucleotide sequence shown in SEQ ID NO: 1, 8, or 10, or a complementary strand thereof. The term "specifically hybridize" as used herein refers to a state in which hybridization with DNA encoding another protein is carried out under ordinary hybridization conditions, preferably under stringent hybridization conditions. It means that no hybridization occurs significantly. Such nucleotides include probes and primers capable of specifically hybridizing with the DNA encoding the protein of the present invention or its complementary strand, nucleotides or nucleotide derivatives (for example, antisense oligonucleotides and ribozymes). .
また、 本発明は、 本発明のタンパク質を利用した、 本発明のタンパク質に結合 する化合物のスクリーニング方法を提供する。  The present invention also provides a method for screening for a compound that binds to the protein of the present invention, using the protein of the present invention.
スクリーニングに用いられる本発明の蛋白質は組換え型、 天然型又は部分ぺプ チドのいずれであってもよい。 また、 精製した蛋白質やその部分ペプチドであつ ても、 細胞表面に発現させた形態、 膜画分としての形態であってもよい。 The protein of the present invention used for screening may be any of a recombinant type, a natural type and a partial peptide. In addition, purified proteins and their partial peptides Or a form expressed on the cell surface or a form as a membrane fraction.
このスクリーニング方法は、 (a )本発明のタンパク質またはその部分べプチド に被検試料を接触させる工程、 (b )本発明のタンパク質またはその部分べプチド に結合する活性を有する化合物を選択する工程、 を含む。 このスクリーニング方 法に用いられる被験試料としては特に制限はなく、 例えば、 細胞抽出物、 細胞培 養上清、 タンパク質、 ペプチド、 合成低分子化合物が挙げられる。 被験試料を接 触させる本発明のタンパク質は、 例えば、 精製したタンパク質として、 細胞膜上 に発現させた形態として、 また、 細胞膜画分として、 被験試料に接触させること ができる。  This screening method comprises: (a) a step of bringing a test sample into contact with the protein of the present invention or a partial peptide thereof; (b) a step of selecting a compound having an activity of binding to the protein of the present invention or a partial peptide thereof; including. The test sample used in this screening method is not particularly limited, and examples include a cell extract, a cell culture supernatant, a protein, a peptide, and a synthetic low-molecular compound. The protein of the present invention that makes a test sample come into contact with the test sample can be, for example, a purified protein, a form expressed on a cell membrane, or a cell membrane fraction.
本発明のタンパク質を用いて、 該タンパク質に対するリガンドを単離する方法 としては、 当業者に公知の多くの方法を用いることが可能である。 例えば、 リガ ンドを発現していることが予想される細胞、組織、臓器よりファージベクター(入 gtll, ZAPなど) を用いた cDNAライブラリ一を作製し、 これを LB-ァガロース上 で発現させフィルターに発現させたタンパク質を固定し、 本発明の夕ンパク質を ピオチンラベル、 あるいは GSTタンパク質との融合タンパク質として精製し、 こ れを上記フィル夕一と反応させ、結合するタンパク質を発現しているプラークを、 ストレプトアビジン、 あるいは抗 GST抗体により検出する 「ウェストウエスタン ブロッテインク法」 (Skolnik EY, Margolis B, Mohammad i M, Lo enstein E, Fi scher R, Drepps A, Ullrich A, and Schlessinger J ( 1991 )Cloning of PI3 ki 皿 se - associated p85 utilizing a novel method for expression/cloning of t arget proteins for receptor tyrosine kinases. Cell 65, 83 - 90) により調製 することが可能である。 また、 本発明のタンパク質を SRF 結合領域または GAL4 結合領域と融合させて酵母細胞の中で発現させ、 リガンドを発現していることが 予想される細胞より、 VP16 または GAL4転写活性化領域と融合する形で発現する ような cDNAライブラリーを作製し、 これを上記酵母細胞に導入し、検出された陽 性クローンからライブラリー由来 cDNA を単離して大腸菌に導入して発現させる (酵母細胞内で本発明の夕ンパク質と結合する夕ンパク質が発現すると、 両者の 結合 よりレポ一夕一遺伝子が活性化され、 陽性のクローンが確認できる) 「two ハイプリヅ ドシステム」 (「MATCHMARKER Two-Hybrid Systemj , 「Ma腿 alian MATC HMAKER Two-Hybrid Assay Kitj , 「MATC腿 KER One-Hybrid Systemj (いずれも c lontech社製)、 「HybriZAP Two-Hybrid Vector Systemj (stratagene社製)、文献As a method for isolating a ligand to the protein using the protein of the present invention, many methods known to those skilled in the art can be used. For example, a cDNA library using a phage vector (eg, gtll, ZAP, etc.) is prepared from cells, tissues, and organs that are expected to express the ligand, expressed on LB-agarose, and used as a filter. The expressed protein is fixed, the protein of the present invention is purified as a fusion protein with a biotin label or a GST protein, and the purified protein is reacted with the above-mentioned filter, and a plaque expressing the protein to be bound is obtained. "Westwestern blotte ink method" for detection with streptavidin or anti-GST antibody (Skolnik EY, Margolis B, Mohammad iM, Loenstein E, Fischer R, Drepps A, Ullrich A, and Schlessinger J (1991) Cloning of PI3 ki dish se-associated p85 utilizing a novel method for expression / cloning of target proteins for receptor tyrosine kinases.Cell 65, 83-90) Rukoto is possible. In addition, the protein of the present invention is fused with an SRF binding region or GAL4 binding region and expressed in yeast cells, and fused with a VP16 or GAL4 transcription activation region from cells expected to express a ligand. A cDNA library is prepared that can be expressed in the form of a cell, and is introduced into the yeast cells described above.The cDNA derived from the library is isolated from the positive clones detected and introduced into E. coli for expression. (When the protein that binds to the protein of the present invention is expressed in yeast cells, the binding of the two activates the repo overnight gene, and a positive clone can be confirmed.) “Two hybrid system” (“ MATCHMARKER Two-Hybrid Systemj, "Ma thigh alian MATC HMAKER Two-Hybrid Assay Kitj," MATC thigh KER One-Hybrid Systemj (all manufactured by clontech), "HybriZAP Two-Hybrid Vector Systemj (stratagene), literature"
「Dalton S, and Treisman R ( 1992)Characterization of SAP- 1, a protein r ecruited by serum response factor to the c-fos serum response element. C ell 68, 597-612」) に従い調製することも可能である。 "Dalton S, and Treisman R (1992) Characterization of SAP-1, a protein recruited by serum response factor to the c-fos serum response element. Cell 68, 597-612") .
また、 本発明蛋白質と特異的に結合するリガンドのスクリーニングは、 本発明 蛋白質の細胞外ドメインと既知のシグナル伝達能を有するへモポェチン受容体な どの受容体蛋白質の細胞膜貫通ドメインを含む細胞内ドメインとを連結せしめて 作製したキメラ受容体を、 適当な細胞株、 好ましくは適当な増殖因子の存在下で のみ生存および増殖可能な細胞株 (増殖因子依存性細胞株) の細胞表面に発現せ しめた後、 該細胞株を種々の増殖因子、 サイ ト力イン、 または造血因子等を含む ことが期待される材料を添加して培養することにより実施可能である。 この方法 は、 被検材料中に本発明蛋白質の細胞外ドメインと特異的に結合するリガンドが 存在する場合にのみ、 上記増殖因子依存性細胞株が生存および増殖が可能である ことを利用している。 既知のへモポェチン受容体としては、 例えば、 トロンボポ ェチン受容体、 エリスロポエチン受容体、 G- CSF受容体、 gpl30等が挙げられるが、 本発明のスクリーニング系に用いるキメラ受容体のパートナーは、 これら既知の へモポェチン受容体に限定されるものではなく、 細胞質ドメインにシグナル伝達 活性に必要な構造を備えているものであれば何を用いても構わない。 例えば、 本 発明のタンパク質と同じ TNF受容体スーパーフアミリーに属する 0X40受容体をキ メラ受容体のパ一トナーとして選択することができる (Mallet, S. et al ., EMB 0 J. , 19, 1063-1068 ( 1990) )。 0X40受容体は、 細胞内に正のシグナルを伝達す ることができることから、 増殖反応等を指標としてリガンドのスクリーニングを 行うことができる (Baum, P.R. et al ., EMBO J. 5 13, 3992-4001 ( 1994))。増 殖因 衣存性細胞株としては、 例えば、 BaF3や FDC-P1を初めとした IL3依存性 細胞株を利用することが可能である。 In addition, screening for a ligand that specifically binds to the protein of the present invention comprises screening the extracellular domain of the protein of the present invention and the intracellular domain containing the transmembrane domain of a receptor protein such as a hemopoietin receptor having a known signal transduction ability. And expressed on the cell surface of an appropriate cell line, preferably a cell line that can survive and grow only in the presence of an appropriate growth factor (growth factor-dependent cell line). Thereafter, the cell line can be cultured by adding a material expected to contain various growth factors, cytodynamic factors, hematopoietic factors, and the like. This method utilizes the fact that the above-mentioned growth factor-dependent cell line can survive and proliferate only when the test material contains a ligand that specifically binds to the extracellular domain of the protein of the present invention. I have. Known hemopoietin receptors include, for example, thrombopoietin receptor, erythropoietin receptor, G-CSF receptor, gpl30, and the like.Partners of the chimeric receptor used in the screening system of the present invention include those known The receptor is not limited to the hemopoietin receptor, and any receptor having a structure necessary for signal transduction activity in the cytoplasmic domain may be used. For example, the 0X40 receptor belonging to the same TNF receptor superfamily as the protein of the present invention can be selected as a partner of the chimeric receptor (Mallet, S. et al., EMB 0 J., 19, 1063). -1068 (1990)). Since the 0X40 receptor can transmit a positive signal into cells, screening for ligands using proliferative response etc. as an index (Baum, PR et al., EMBO J. 5 13, 3992-4001 (1994)). Proliferation factors As resident cell lines, for example, IL3-dependent cell lines such as BaF3 and FDC-P1 can be used.
さらに、 本発明のタンパク質を、 そのリガンドを発現していない細胞で発現さ せ、 次いで、 該細胞に、 リガンドを発現していることが予想される細胞より構築 した発現 cDNAライブラリーを COSなどの細胞に導入して得た培養上清を添加し、 そして細胞の機能的あるいは形態的変化を指標にリガンドを探索する 「ダイレク ト発現クローニング法」 (Yokota T, Otsuka T, Mosmann Τ, Banchereau J, DeFr ance T, Blanchard D, De Vries JE, Lee F, and Arai K. ( 1986) Isolation and characterization of a human interleukin cDNA clone, homologous to mouse B - cell stimulatory factor 1, that expresses B-cell- and T-cell-stimulat ing activities. Proc Natl Acad Sci U S A. 83, 5894- 5898)により調製するこ とも可能である。 これらのスクリーニング方法は、 本発明のタンパク質に対する 抗体や、 本発明のタンパク質に結合する化合物を被検試料として用いたァゴニス トゃアン夕ゴニス卜のスクリーニングに用いることも可能である。  Furthermore, the protein of the present invention is expressed in a cell that does not express the ligand, and then an expressed cDNA library constructed from cells that are expected to express the ligand is expressed in the cell. “Direct expression cloning method” in which the culture supernatant obtained by introduction into cells is added, and ligands are searched based on the functional or morphological changes of the cells (Yokota T, Otsuka T, Mosmann II, Banchereau J, DeFrance T, Blanchard D, De Vries JE, Lee F, and Arai K. (1986) Isolation and characterization of a human interleukin cDNA clone, homologous to mouse B-cell stimulatory factor 1, that expresses B-cell- and T- It can also be prepared using cell-stimulating activities. Proc Natl Acad Sci US A. 83, 5894-5898). These screening methods can also be used for screening agonist and gonist using an antibody against the protein of the present invention or a compound that binds to the protein of the present invention as a test sample.
さらにまた、 本発明のタンパク質を固定したァフィ二ティ一カラムに本発明の リガンドを発現していることが予想される細胞の培養上清をのせ、 カラムに特異 的に結合するタンパク質を精製することにより調製することも可能である。 得ら れたタンパク質 (リガンド) のアミノ酸配列を分析し、 それを基にオリゴ DNAを 合成し、該 DNAをプローブとして cDNAライブラリ一をスクリーニングすることに より、 該リガンドをコードする DNAを得ることが可能である。  Furthermore, the culture supernatant of cells expected to express the ligand of the present invention is placed on an affinity column on which the protein of the present invention is immobilized, and the protein that specifically binds to the column is purified. Can also be prepared. The DNA encoding the ligand can be obtained by analyzing the amino acid sequence of the obtained protein (ligand), synthesizing oligo DNA based on the amino acid sequence, and screening a cDNA library using the DNA as a probe. It is possible.
さらにまた、 本発明の蛋白質の細胞外領域と、 抗体 (例えばヒト IgG抗体) の Fc領域とのキメラ蛋白質として発現し、 プロテイン Aカラム等を用いて精製する ことができる。 このような抗体様キメラ蛋白質は、 リガンドの結合活性を有する ことから、 適宜、 放射性同位元素等で標識した後、 リガンドのスクリーニングに 用いることができる (Suda, T. et al ., Cell , 175, 1169-1178 ( 1993) )。 TNFフ アミリー分子はその多くが膜結合型でも存在することから、 各種の細胞と抗体様 キメテ蛋白質を反応させて、 結合活性を示した細胞から、 リガンドを単離する事 ができる。 また、 cDNAライブラリーを導入した細胞を用いて同様にリガンドを単 離することができる。 さらに、 抗体様キメラ蛋白質をアン夕ゴニストとして用い ることも可能である。 Furthermore, it can be expressed as a chimeric protein of the extracellular region of the protein of the present invention and the Fc region of an antibody (for example, human IgG antibody), and purified using a protein A column or the like. Since such an antibody-like chimeric protein has ligand-binding activity, it can be appropriately labeled with a radioisotope or the like and then used for ligand screening (Suda, T. et al., Cell, 175, 1169-1178 (1993)). TNF Since most of the amilly molecules are also present in a membrane-bound form, ligands can be isolated from cells that have exhibited binding activity by reacting various cells with the antibody-like chimete protein. In addition, the ligand can be similarly isolated using cells into which the cDNA library has been introduced. Furthermore, an antibody-like chimeric protein can be used as an agonist.
また、 本発明のタンパク質を用いて、 該タンパク質に対するァゴニスト、 およ びアン夕ゴニストを単離する方法としては、 例えば、 固定した本発明のタンパク 質に、 化合物、 または天然物バンク、 もしくはランダムファージペプチドデイス プレイライブラリ一を作用させ、 結合する分子をスクリーニングする方法や、 コ ンビナトリアルケミストリ一技術によるハイスループットを用いたスクリ一ニン グ方法 (Wrighton NC; Farrell FX; Chang R; Kashyap AK; Barbone FP ; Mu lcahy LS; Johnson DL; Barrett RW; Jolliffe LK; Dower WJ. , Small peptides as potent mimetics of the protein hormone erythropoietin, Science (UNITE D STATES) Jul 26 1996, 273 p458- 64、 Verdine GL. , The combinatorial chemi stry of nature. Nature (ENGLAND) Nov 7 1996, 384 pi卜 13、 Hogan JC Jr. ,Di rected combinatorial chemistry. Nature (ENGLAND) Nov 7 1996, 384 pl7-9) が当業者に公知である。  Methods for isolating an agonist and an engonist against the protein using the protein of the present invention include, for example, a compound, a natural product bank, or a random phage in the immobilized protein of the present invention. Screening for binding molecules using peptide display libraries and high-throughput screening methods using combinatorial chemistry technology (Wrighton NC; Farrell FX; Chang R; Kashyap AK; Barbone FP; Mu lcahy LS; Johnson DL; Barrett RW; Jolliffe LK; Dower WJ., Small peptides as potent mimetics of the protein hormone erythropoietin, Science (UNITE D STATES) Jul 26 1996, 273 p458-64, Verdine GL., The Nature (ENGLAND) Nov 7 1996, 384 pi 13, Hogan JC Jr., Di rected combinatorial chemistry.Nature (ENGLAND) Nov 7 1996, 384 pl 7-9) are known to those skilled in the art.
<NF- kB活性を指標としたマウスおよびヒト Troyの活性を調節する活性を有する 化合物のスクリーニングを追加致しました。 不要であれば削除致します > また、 本発明は、 本発明のタンパク質の活性を調節する化合物をスクリーニン グする方法を提供する。このスクリーニング方法は、 (a )本発明のタンパク質を コードする DNAを発現するべクタ一および NF- kBの結合に応答してレポ一夕一遺 伝子を発現するべクタ一を含む細胞に被検試料を接触させる工程、 (b )該細胞に おけるレポ一夕一活性を検出する工程、 および (c ) 被検試料を該細胞に接触さ せないで検出した場合と比較して、 該レポ一夕一活性を低下または増加させる化 合物を選択する工程、 を含む。 <Screening for compounds that regulate the activity of mouse and human Troy based on NF-KB activity has been added. The present invention also provides a method for screening for a compound that regulates the activity of the protein of the present invention. This screening method comprises the steps of: (a) covering cells containing a vector that expresses a DNA encoding the protein of the present invention and a vector that expresses a repo allele in response to binding of NF-KB; Contacting a test sample, (b) detecting the repo overnight activity in the cells, and (c) comparing the repo activity to the test sample without contacting the cells. Decrease or increase the activity overnight Selecting a compound.
本 ¾fl月の夕ンパク質をコードする DNAを発現させるためのベクタ一としては、 例えば、 pCOSlや pME18を用いることができる。 また、 NF- kBの活性化に応答して レポーター遺伝子を発現するべクタ一としては、 例えば、 実施例に記載の kB-luc を用いることができる。  For example, pCOSl or pME18 can be used as a vector for expressing DNA encoding the protein of the present month. In addition, as a vector that expresses a reporter gene in response to activation of NF-kB, for example, kB-luc described in Examples can be used.
kB - lucは、 以下の原理によりレポーター遺伝子を発現する。 即ち、 本発明の夕 ンパク質にシグナルが伝達されると、 TRAF (TNFR associated factor) を介し、 その下流のキナーゼが活性化する。 このキナーゼにより IkBキナーゼ複合体が活 性化され、 NF-kBに結合している抑制性タンパク質 IkBをリン酸化する。 すると、 IkBが分解され、 NF- kBが遊離し、 核内へ移行する。 この NF- kBが kB- luc上の結 合部位に結合するとそれに応じてレポ一夕一遺伝子が発現する。  kB-luc expresses a reporter gene according to the following principle. That is, when a signal is transmitted to the protein of the present invention, kinases downstream thereof are activated via TRAF (TNFR associated factor). This kinase activates the IkB kinase complex and phosphorylates the inhibitory protein IkB bound to NF-kB. Then, IkB is degraded, and NF-kB is released and translocated into the nucleus. When this NF-kB binds to the binding site on kB-luc, the repo overnight gene is expressed accordingly.
本発明のスクリーニングに用いる細胞としては、 例えば、 HEK293T 細胞株が好 適である。 細胞へのベクタ一の導入は、 当業者に公知の方法で行なうことができ る。  As the cells used for the screening of the present invention, for example, the HEK293T cell line is preferable. Introduction of a vector into a cell can be performed by a method known to those skilled in the art.
本発明のスクリーニング方法においては、 ベクターの導入された細胞に被検試 料を接触させ、 レポ一夕一活性を検出する。被検試料としては、 特に制限はなく、 例えば、 細胞抽出物、 細胞培養上清、 タンパク質、 ペプチド、 合成低分子化合物 が挙げられる。 上記本発明のタンパク質に結合する化合物のスクリーニングによ り単離された化合物を用いることも考えられる。 レポ一夕一活性の検出の結果、 被検試料を細胞に接触させない場合と比較して、 レポ一夕一活性が低下していれ ば、 用いた被検試料は本発明のタンパク質の活性を阻害すると判定され、 一方、 レポーター活性が増加していれば、 用いた被検試料は本発明のタンパク質の活性 を誘導または増加させると判定される。  In the screening method of the present invention, a test sample is brought into contact with cells into which a vector has been introduced, and the repo overnight activity is detected. The test sample is not particularly limited, and includes, for example, a cell extract, a cell culture supernatant, a protein, a peptide, and a synthetic low-molecular compound. It is also conceivable to use a compound isolated by screening for a compound that binds to the protein of the present invention. As a result of the detection of the repo overnight activity, the test sample used inhibits the activity of the protein of the present invention if the repo overnight activity is lower than when the test sample is not brought into contact with the cells. On the other hand, if the reporter activity is increased, it is determined that the test sample used induces or increases the activity of the protein of the present invention.
本発明のスクリーニングにより単離されたリガンド、 ァゴニストあるいはアン 夕ゴニストは、 本発明のタンパク質が関与する疾患 (例えば、 神経関連疾患) の 治療への応用が考えられる。 本発明のスクリーニング法により単離される化合物 を、 薬剤として用いる場合には、 公知の製剤学的製造法により製剤化して用いる こと^能である。 例えば、 薬理学上許容される担体または媒体 (生理食塩水、 植物油、 懸濁剤、 界面活性剤、 安定剤など) とともに患者に投与される。投与は、 化合物の性質に応じて、 経皮的、 鼻腔内的、 経気管支的、 筋内的、 静脈内、 また は経口的に行われると考えられる。 投与量は、 患者の年齢、 体重、 症状、 投与方 法などにより変動するが、 当業者であれば適宜適当な投与量を選択することが可 能である。 The ligand, agonist or aminogonist isolated by the screening of the present invention can be applied to the treatment of a disease associated with the protein of the present invention (for example, a nerve-related disease). Compound isolated by the screening method of the present invention When used as a drug, it can be formulated into a drug by a known pharmaceutical manufacturing method. For example, it is administered to a patient together with a pharmacologically acceptable carrier or vehicle (eg, saline, vegetable oils, suspensions, surfactants, stabilizers, etc.). Administration will be transdermal, intranasal, transbronchial, intramuscular, intravenous, or oral, depending on the nature of the compound. The dose varies depending on the patient's age, body weight, symptoms, administration method, and the like, but those skilled in the art can appropriately select an appropriate dose.
例えば、 本発明の蛋白質と結合活性を有する化合物の投与量は、 症状により差 異はあるが、 経口投与の場合、 一般的に成人 (体重 60kgとして) においては、 1 日あたり約 0. 1から 100mg、 好ましくは約 1.0から 50mg、 より好ましくは約 1.0 から 20mgであると考えられる。  For example, the dosage of the compound having the binding activity to the protein of the present invention may vary depending on the condition. It is believed to be 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
非経口的に投与する場合は、 その 1回投与量は投与対象、 対象臓器、 症状、 投 与方法によっても異なるが、 例えば注射剤の形では通常成人 (体重 60kgとして) においては、 1 日あたり約 0.01から 30mg、 好ましくは約 0. 1から 20mg、 より好 ましくは約 0. 1から 1 Omg程度を静脈注射により投与するのが好都合であると考え られる。他の動物の場合も、 体重 60kg当たりに換算した量、 あるいは、 体表面積 あたりに換算した量を投与することができる。 図面の簡単な説明  For parenteral administration, the single dose varies depending on the subject of administration, target organ, symptoms, and administration method. For example, in the case of parenteral injections, usually for adults (with a body weight of 60 kg), It may be convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 Omg by intravenous injection. In the case of other animals, the dose can be administered in terms of the amount converted per 60 kg body weight or the amount converted per body surface area. BRIEF DESCRIPTION OF THE FIGURES
図 1は、マウス Troyおよび dTroyの NF-kB活性誘導能の解析を行なった結果を 示す。 発明を実施するための最良の形態  FIG. 1 shows the results of analyzing the ability of mouse Troy and dTroy to induce NF-kB activity. BEST MODE FOR CARRYING OUT THE INVENTION
以下、 本発明を実施例により具体的に説明するが、 本発明はこれら実施例に制 限されるものではない。  Hereinafter, the present invention will be described specifically with reference to Examples, but the present invention is not limited to these Examples.
[実施例 1 ] シグナルシーケンストラップ (SST)法によるマウス脳 cDNA ライ ブラリーのスクリーニング [Example 1] Mouse brain cDNA library by signal sequence trap (SST) method Bally Screening
マ ス脳より TRIZOL (GIBCO BRL)を用いて RNAを抽出し、 さらにこれを oligo dTカラム (Pharmacia)に通塔することにより polyA( + )RNAを調製した。 SuperSc ript Choice System (GIBCO BRL )のランダムへキサマ一を用いて二本鎖 cDNA を 合成した。 cDNAの末端平滑処理後 BstXIアダプタ一 (Invitrogen)を付加し、 Siz eSep 400 Spun Column (Pharmacia)を用いて 400bp以上の cDNAを分画した。 別 に BstXI (宝酒造) 及び BAP処理した pMX GM (- ) v-mplM 2 (特願平 9- 324912号参 照) と混合後 T4 DNA ligaseを作用させて連結した。 これを Gene Pulser (Bio Rad)を用いて電気穿孔法にて大腸菌 DH10B (GIBCO BRL)に導入して cDNA ライブ ラリーを構築した。 RNA was extracted from the mouse brain using TRIZOL (GIBCO BRL) and passed through an oligo dT column (Pharmacia) to prepare polyA (+) RNA. Double-stranded cDNA was synthesized using a random hexamer of the SuperScript Choice System (GIBCO BRL). After blunt-ending the cDNA, BstXI adapter-1 (Invitrogen) was added, and a cDNA of 400 bp or more was fractionated using a SizeSep 400 Spun Column (Pharmacia). Separately, the mixture was mixed with BstXI (Takara Shuzo) and pMXGM (-) v-mpl M2 (see Japanese Patent Application No. 9-324912) treated with BAP, and ligated with T4 DNA ligase. This was introduced into Escherichia coli DH10B (GIBCO BRL) by electroporation using Gene Pulser (Bio Rad) to construct a cDNA library.
ライブラリ一組換え大腸菌から抽出したプラスミ ドを JETstar カラム (GEN0M ED) を用いて精製した。 パッケージング細胞 B0SC23 (Proc. Natl . Acad. Sci . USA, vol .90: 8392-8396, 1993) に LipofectMINE (LIFE TECHNOLOGIES)を用い てプラスミ ド cDNAライブラリーをトランスフエクトした。 B0SC23を 10% ゥシ胎 児血清 (FCS, JRH BIOSCIENCES)を含む Dulbecco' s Modified Eagel Medium (DM EM, LIFE TECHNOLOGIES)で 6cm dish (Corning) に播き込み 16時間後、 DMEMで洗 浄し、先に 200〃1 DMEMで希釈した 18〃1 LipofectMINEと 200〃1 DMEMで希釈 したプラスミ ド 3 gを混ぜて 15分間室温で放置したものに 1.6ml DMEMを混ぜ て細胞に加えた。 5時間後 2ml の 20%FCSを含む DMEMを加え 19 時間培養した。 その後 3mlの 10% FCSを含む DMEMに置換し 24時間後にその培養上清を回収し た。この組み換えウィルスを含む培養上清にマウス interleukin- 3 ( IL-3) 及び 10 g/ml hexadimethrine bromideを加え、 これに Ba/F3細胞を懸濁して感染さ せた。感染させて 24時間後細胞をリン酸緩衝液にて 3回洗浄し 10% FCSを含む R PMI1640にて培養を続けた。  Plasmids extracted from the recombinant Escherichia coli were purified using a JETstar column (GEN0M ED). Acad. Sci. USA, vol. 90: 8392-8396, 1993) was used to transfect a plasmid cDNA library using LipofectMINE (LIFE TECHNOLOGIES). B0SC23 is seeded on a 6 cm dish (Corning) with Dulbecco's Modified Eagel Medium (DM EM, LIFE TECHNOLOGIES) containing 10% fetal calf serum (FCS, JRH BIOSCIENCES), and after 16 hours, washed with DMEM and washed first. Then, 18 g of LipofectMINE diluted with 200 希 釈 1 DMEM and 3 g of plasmid diluted with 200〃1 DMEM were mixed, left at room temperature for 15 minutes, mixed with 1.6 ml of DMEM, and added to the cells. Five hours later, 2 ml of DMEM containing 20% FCS was added, and the cells were cultured for 19 hours. Then, it was replaced with 3 ml of DMEM containing 10% FCS, and the culture supernatant was collected 24 hours later. Mouse interleukin-3 (IL-3) and 10 g / ml hexadimethrine bromide were added to the culture supernatant containing the recombinant virus, and Ba / F3 cells were suspended and infected with the mixture. Twenty-four hours after the infection, the cells were washed three times with a phosphate buffer, and cultured with RPMI1640 containing 10% FCS.
IL-3非存在下で増殖してきたクローンから染色体 DNAを抽出し、 cDNA挿入部位 を挟むように設定したブラィマー(5, -gggggtggaccatcctcta-3' /配列番号: 3、 および 5, -cgcgcagctgtaaacggtag-3, /配列番号: 4 ) を用いて PCRを行い cDNA 断片を—回収した。 PCRは 500ng染色体 DNA、 500pM各プライマ一、 TaKaRa LA Taq (宝酒造) 2.5単位、 2.5mM MgCl2、 0.3mM dNTPs 及び酵素添付緩衝液を含む反応 液 50〃1について GeneAmpPCR System 2400で以下の条件にて行った。 98°C、 60 秒の変成後、 「98°C,20秒、 68°C, 120秒」 のサイクルを 30回行った。 PCR反応物 をァガロース電気泳動にかけ増幅された断片を含む部分を切り出し、 DNA を精製 した。 得られた DNA断片についての塩基配列を決定したところ、 TNF受容体ス一 パーフアミリ一をコ一ドする cDNAの一部であることが明らかとなった。 A chromosomal DNA was extracted from a clone grown in the absence of IL-3, and a primer (5, -gggggtggaccatcctcta-3 '/ SEQ ID NO: 3, which was set so as to sandwich the cDNA insertion site, And 5, -cgcgcagctgtaaacggtag-3, / SEQ ID NO: 4), and a cDNA fragment was recovered by PCR. PCR: 500 ng chromosomal DNA, 500 pM each primer, TaKaRa LA Taq (Takara Shuzo) 2.5 units, 2.5 mM MgCl 2 , 0.3 mM dNTPs and enzyme attached buffer went. After denaturation at 98 ° C for 60 seconds, 30 cycles of “98 ° C, 20 seconds, 68 ° C, 120 seconds” were performed. The PCR reaction product was subjected to agarose gel electrophoresis to cut out a portion containing the amplified fragment, and the DNA was purified. The nucleotide sequence of the obtained DNA fragment was determined, and it was revealed that the DNA fragment was a part of cDNA encoding the TNF receptor superfamily.
[実施例 2 ] 全長 cDNAのクロ一ニングと塩基配列決定 [Example 2] Cloning of full-length cDNA and determination of nucleotide sequence
完全長 cDNAを得るためにマウス脳 cDNAライブラリ一を oligo dTプライマー で合成し、 上記 cDNA断片をプローブにスクリーニングした。 マウス脳 polyA( + ) RNAから Superscript I I逆転写酵素 (GIBCO BRい、 Sai lアダプタープライマ一(5 ' -gagagagagagagagagagaaacgcgtcgacgcggccgccctttttttttttttttttt-3' /酉己歹1 J番 号: 5 )及び 0.3mM 5 -methyl dCTP、 0.6mM dATP、 dGTP、 dTTPを用いて cDNAを合 成した。 ここに E. coli RNaseH、 E. coli DNA polymerase I及び 0.25mM dATP、 dGTPs dTTP, 0.375mM dCTPを反応させ二本鎖 cDNAを合成し、 K0D DNA polymera se (東洋紡)で cDNAの末端平滑処理後 EcoRIアダプター (Pharmacia)を付加し、 Sai l処理した。 これを SizeSep 400 Spun Column (Pharmacia)に通塔して 400bp 以上の cDNAを分画した。 別に Notl (宝酒造) 及び BAP処理した後さらに EcoRI 処理した pGEM-llZf( + ) (Promega) と混合後 T4 DNA ligaseを作用させて連結し た。 これを Gene Pulser (Bio Had)を用いて電気穿孔法にて大腸菌 DH10B ( GIBC0 BRL)に導入して cDNAライブラリーを構築した。 ライブラリー組換え大腸菌から 抽出したプラスミ ドを JETstarカラム (GEN0MED )を用いて精製した。 To obtain a full-length cDNA, a mouse brain cDNA library was synthesized using oligo dT primers, and the above cDNA fragment was screened as a probe. There Superscript II reverse transcriptase (GIBCO BR from mouse brain polyA (+) RNA, Sai l adapter primer one (5 '-gagagagagagagagagagaaacgcgtcgacgcggccgccctttttttttttttttttt-3' / Rooster himself歹1 J NO: 5) and 0.3mM 5 -methyl dCTP, CDNA was synthesized using 0.6 mM dATP, dGTP, and dTTP, and double-stranded cDNA was synthesized by reacting E. coli RNaseH, E. coli DNA polymerase I, and 0.25 mM dATP, dGTPs dTTP, and 0.375 mM dCTP. After the cDNA was blunt-ended with K0D DNA polymerase (Toyobo), EcoRI adapter (Pharmacia) was added, and the mixture was treated with Sail, which was passed through a SizeSep 400 Spun Column (Pharmacia) to fractionate cDNA of 400bp or more. Separately, after Notl (Takara Shuzo) and BAP treatment, and further mixed with EcoRI-treated pGEM-llZf (+) (Promega), the mixture was ligated by the action of T4 DNA ligase using Gene Pulser (Bio Had). It was introduced into E. coli DH10B (GIBC0 BRL) by electroporation to construct a cDNA library. Plasmids extracted from the library recombinant E. coli were purified using a JETstar column (GEN0MED).
先に SST法にて得られた DNA断片 0.1〃gを錶型にその内部プライマー(50pmol e) (55S; 5,一 caaggtcctacctctacaca— 3, /酉己列番号: 6、 および 511A; 5' -aaggtt caccttgctggtac- 3, /配列番号: 7 )、 2.5mM MgCl2、 0.2mM dATP, dGTP, dCTPヽ 0. ImM dT P、 10/ M Biotin- 21-dUTP、 2.5単位1& 1^ LA Taq (宝酒造) 及び酵素 添付緩衝液を含む反応液 50〃1について GeneAmpPCR System2400で 98°C、 60秒 の変成後、 「98°C , 20秒、 56°C, 20秒、 74°C, 60秒」 のサイクルを 30回行いビ ォチン化した。このピオチン化 DNA lOOngを含む 14.9 1水溶液を 100°C5分変成 急冷後、 2mM CoCl2、 30mM TrisHCl pH8.0、 8mM MgCl2、 1.6mM ATP [ァ S]、 80 /M A TP (それそれ最終濃度) 及び 4〃g recAを加え、 37°C15分間保温後上記プラスミ ド cDNAライブラリーを 5〃g加え(最終容量 30 / 1 )更に 20分間保温した。 Hindl l I消化え DNAを 50ng加え 5分後、 10%SDSを 0.6 1、 14mg/ml ProteinaseKを 0. 加え 10分間保温後 50mM PMSFを 2〃1加えて反応を停止させた。ここに鮭精 巣 DNAで blocking処理した Streptavidin MagneSphere (Promega)を加え、 20分 間吸着させた。 0.05%Tween20 を含む 10mM TrisHCl pH7.5, ImM EDTA, 1M NaCl で 1回、 lOmM TrisHCl pH7.5, ImM EDTA, 2M NaClで 3回洗浄後、 さらに蒸留水 で 1回洗浄し ImM EDTA, 0.1N NaOHで DNAを溶出させた。 0.1 μg of the DNA fragment previously obtained by the SST method was converted into type III and its internal primer (50 pmoles) (55S; 5, one caaggtcctacctctacaca—3 // Rooster column numbers: 6, and 511A; 5′-aaggtt caccttgctggtac-3, / SEQ ID NO: 7), 2.5 mM MgCl 2 , 0.2 mM dATP, dGTP, dCTP ヽ 0. ImM dTP, 10 / M Biotin-21-dUTP, 2.5 units 1 & 1 ^ LA Taq (Takara Shuzo) and Enzyme Reaction of 50〃1 containing the buffer attached to the sample After denaturation at 98 ° C for 60 seconds using GeneAmpPCR System2400, cycle “98 ° C, 20 seconds, 56 ° C, 20 seconds, 74 ° C, 60 seconds”. Performed 30 times for biotinylation. Denature the 14.91 aqueous solution containing this biotinylated DNA 100% for 5 minutes at 100 ° C. After quenching, 2 mM CoCl 2 , 30 mM TrisHCl pH 8.0, 8 mM MgCl 2 , 1.6 mM ATP [αS], 80 / MATP (each final concentration) ) And 4 µg recA were added, and the mixture was incubated at 37 ° C for 15 minutes. Then, 5 µg of the plasmid cDNA library was added (final volume: 30/1), and the mixture was further incubated for 20 minutes. After adding 50 ng of HindIII digested DNA and adding 5 ng of the DNA, 5 minutes later, 0.61 of 10% SDS and 0.1 of 14 mg / ml ProteinaseK were added, and the mixture was incubated for 10 minutes, and the reaction was stopped by adding 2〃1 of 50 mM PMSF. To this was added Streptavidin MagneSphere (Promega) blocked with salmon testis DNA and allowed to adsorb for 20 minutes. Wash once with 10mM TrisHCl pH7.5, ImM EDTA, 1M NaCl containing 0.05% Tween20, 3 times with lOmM TrisHCl pH7.5, ImM EDTA, 2M NaCl, then wash once with distilled water and ImM EDTA, 0.1N The DNA was eluted with NaOH.
これをエタノール沈殿後 Gene Pulser (Bio Rad)を用いて電気穿孔法にて大腸 菌 DH10B (GIBC0 BRL)に導入し、 50 g/mlアンピシリン含有培地上にて組み換え 体を選択した。  This was precipitated with ethanol, introduced into E. coli DH10B (GIBC0 BRL) by electroporation using a Gene Pulser (Bio Rad), and recombinants were selected on a medium containing 50 g / ml ampicillin.
得られたコロニーからプラスミ ドを抽出し、上記の recA反応を再度行い、アン ビシリン含有培地上にて組み換え体を選択した。上記の 55S及び 511Aを用いて同 様の条件にてコロニー PCRを行った。 約 460塩基のバンドが得られた陽性コロニ 一からプラスミ ドを抽出して挿入 cDNAの塩基配列を決定した。 該 cDNAは、 4089 塩基からなり、 416 アミノ酸残基のオープンリーディングフレーム(127- 1377)が 認められた。 1〜29アミノ酸がシグナル配列、 171〜193アミノ酸が膜貫通ドメィ ンと推定される。 また細胞外ドメインについては 0X40 (P15725) 等の TNF受容体 スーパ一ファミリ一間と高い相同性を示した。 また、 皮膚の毛包 (hair follicl e)や汗腺や歯の発達に関与すると考えられている Edar(Nature Genetics 1999 v 01.22. 370-374, 366-369)と 0X40以上の高い相同性を示した。本発明者等は、単 離したクロ一ンを 「マウス TRoy」 と命名した。 Plasmid was extracted from the obtained colonies, the recA reaction was performed again, and a recombinant was selected on an ambicilin-containing medium. Colony PCR was performed using 55S and 511A described above under the same conditions. Plasmid was extracted from the positive colony where a band of about 460 bases was obtained, and the base sequence of the inserted cDNA was determined. The cDNA was composed of 4089 bases, and an open reading frame of 416 amino acid residues (127-1377) was recognized. It is estimated that amino acids 1-29 are signal sequences and amino acids 171-193 are transmembrane domains. In addition, the extracellular domain showed high homology with one superfamily of TNF receptors such as 0X40 (P15725). In addition, Edar (Nature Genetics 1999 v) is considered to be involved in the development of hair follicles, sweat glands and teeth. 01.22. 370-374, 366-369) and 0X40 or higher. The present inventors have named the isolated clone "mouse TRoy".
[実施例 3 ] ノーザンブロッテイング [Example 3] Northern blotting
マウス TRoy遺伝子の発現の組織特異性を調べるために、マウス Multiple Tiss ue Northern (MTN) Blot(clontech)を用いて得られた cDNA断片をプロ一ブにハ イブリダィゼイシヨンを行ったところ、 脳において強いシグナルが、 心臓 '肺 ' 肝臓において弱いシグナル、 骨格筋 ·腎臓 '精巣においても極々弱いシグナルが 認められ、 脳特異的に発現する受容体であると考えられた。  In order to examine the tissue specificity of mouse TRoy gene expression, a cDNA fragment obtained using a mouse Multiple Tissue Northern (MTN) Blot (clontech) was hybridized to a probe, A strong signal was observed in the brain, a weak signal was observed in the heart 'lung' and liver, and an extremely weak signal was also observed in the skeletal muscle and kidney 'testes', suggesting that the receptor was expressed specifically in the brain.
さらに、 ノーザンプロット解析により検討を加えたところ、 マウス胚、 特に皮 膚において強いシグナルが認められた。  Furthermore, when examined by Northern blot analysis, a strong signal was observed in mouse embryos, particularly in the skin.
[実施例 4 ] 細胞内ドメインを殆ど含まないクローン 「マウス dTroyj の単離 ランダムへキサマ一をプライマ一として用いて 1 7日目マウス胚の皮膚 cDNA ラィブラリ一を構築し、マウス cDNAの細胞外ドメイン部分をプローブにして RecA 法にてクローンを分離した。 その結果、 細胞内ドメインを殆ど含まないクローン dTroyが単離された。 dTroy cDNAの塩基配列を配列番号: 8に、 該 cDNAがコ一ド する蛋白質のアミノ酸配列を配列番号: 9に示す。 [Example 4] A clone containing almost no intracellular domain “Isolation of mouse dTroyj” A skin cDNA library of a mouse embryo on day 17 was constructed using a random hexamer as a primer, and the extracellular domain of mouse cDNA was constructed. Using the portion as a probe, clones were isolated by the RecA method, and as a result, a clone dTroy containing almost no intracellular domain was isolated, and the nucleotide sequence of the dTroy cDNA was represented by SEQ ID NO: 8, and the cDNA was coded. The amino acid sequence of the protein is shown in SEQ ID NO: 9.
[実施例 5 ] ヒト Troy cDNAの単離 [Example 5] Isolation of human Troy cDNA
ヒト神経膠肉腫細胞株 (gliosarcoma) GI-1においてマウス Troy遺伝子のヒ ト相同分子の発現が、 ノーザンプロッ トにて認められた。 そこで、 ヒト神経膠肉 腫細胞株 GI- 1由来の cDNAライブラリーをランダムへキサマ一をプライマーとし て用いて構築し、 マウス cDNAの細胞外ドメイン部分をプローブにして RecA法を 実施した。その結果、マウス Troy遺伝子に対応するヒトクローンが単離された。 ヒト Troy cDNAの塩基配列を配列番号: 1 0に、 該 cDNAがコードする蛋白質のァ ミノ酸配列を配列番号: 1 1に示す。 In a human gliosarcoma cell line (gliosarcoma) GI-1, expression of a human homologous molecule of the mouse Troy gene was observed in a Northern plot. Therefore, a cDNA library derived from the human glial sarcoma cell line GI-1 was constructed using random hexamers as primers, and the RecA method was performed using the extracellular domain portion of mouse cDNA as a probe. As a result, a human clone corresponding to the mouse Troy gene was isolated. The nucleotide sequence of human Troy cDNA is shown in SEQ ID NO: 10, and the sequence of the protein encoded by the cDNA is shown in FIG. The amino acid sequence is shown in SEQ ID NO: 11.
[実施例 6 ] マウス Troyおよび dTroyの NF-kB活性誘導能の解析 [Example 6] Analysis of the ability of mouse Troy and dTroy to induce NF-kB activity
マウス Troyの細胞内ドメインに TNF 受容体 I I 型のシグナル伝達分子である TNFR- associated factor (TRAF ) 2 の結合配列 TLQE が見出されることから、 その NF-kB活性誘導能を検討した。 6ゥエルプレートに 10%FCSを含む DMEMで HEK293T 細胞株を蒔き込み 16時間後に FuGene6 (Roche)6. 1を用いてマウス Troy cDNAま たはマウス dTroy cDNAが挿入されたプラスミ ドをトランスフエクシヨンした。 ト ランスフエクション効率を標準化するための /5ガラク トシダ一ゼ発現べク夕一 pTK-lacZ lOOng, レポ一夕一プラスミ ド lOOng と他のプラスミ ド (マウス Troy cDNAまたはマウス dTroy cDNAが揷入された pCOSI) 1.3. gをトランスフエクショ ンした。 なお、 モックとしては、 Troyおよび dTroyを発現させるために用いた発 現ベクター pCOSIを用いた。  Since the binding sequence TLQE of TNFR-associated factor (TRAF) 2, a TNF receptor II type signaling molecule, was found in the intracellular domain of mouse Troy, its ability to induce NF-kB activity was examined. Incubate the HEK293T cell line in DMEM containing 10% FCS on a 6-well plate and transfer the plasmid into which mouse Troy cDNA or mouse dTroy cDNA has been inserted using FuGene6 (Roche) 6.1 16 hours later. did. / 5 galactosidase expression vector pTK-lacZ lOOng, repo overnight plasmid lOOng and other plasmids (mouse Troy cDNA or mouse dTroy cDNA were inserted) to standardize the transfection efficiency. 1.3. G was transfected. As a mock, the expression vector pCOSI used for expressing Troy and dTroy was used.
レポ一夕一プラスミ ド kB- lucは、 HTLV-1プロモーターの基本プロモーター dN55 (癌, 79号, 800- 804( 1988) ) の上流に 5つの NF- kB結合サイ トを、 下流にルシフ エラーゼ遺伝子を有する。 24時間後、 細胞を PBSで洗い集め、 250mM Tris緩衝 液 (PH7.8)中に懸濁し、凍結と融解を 3回繰り返して細胞を壊した。遠心上精をサ ンプルとして 0-二 トロフェニル -. -D-ガラク トビラノシ ドを基質に文献 (Molecular Cloning) の記載に従い/?ガラクトシダーゼ活性を測定した。  The repo overnight plasmid kB-luc contains five NF-kB binding sites upstream of the basic HTLV-1 promoter dN55 (cancer, 79, 800-804 (1988)) and a luciferase gene downstream. Having. After 24 hours, the cells were washed with PBS, suspended in 250 mM Tris buffer (PH7.8), and frozen and thawed three times to break the cells. Using 0-nitrophenyl -.- D-galactobyranoside as a substrate using the centrifuged supernatant as a sample, the galactosidase activity was measured as described in the literature (Molecular Cloning).
また、、 ルシフヱラーゼ活性はピツカジーン (東洋インキ製)を用いて測定した。 その結果、 Mockに比べてマウス Troyを発現させた際、 3.5倍以上のルシフエラー ゼ活性が誘導され、 該受容体による NF-kB活性化が示唆された。 一方、 短い分子 dTroyを発現させても殆どルシフヱラ一ゼ活性は誘導されなかった (図 1 )。 産業上の利用の可能性  The luciferase activity was measured using Pitka Gene (manufactured by Toyo Ink). As a result, when mouse Troy was expressed compared to Mock, luciferase activity was induced 3.5 times or more, suggesting that NF-kB was activated by the receptor. On the other hand, expression of the short molecule dTroy hardly induced luciferase activity (Fig. 1). Industrial applicability
本発明により、新規な TNF受容体様夕ンパク質およびその遺伝子が提供された。 これらタンパク質や遺伝子は、 脳機能や皮膚の発達などに関与する新たな因子の 精製ゃクローニング、 さらには医薬品候補化合物のスクリーニングのための有用 なツールとして利用しうる。 According to the present invention, a novel TNF receptor-like protein and its gene have been provided. These proteins and genes can be used as useful tools for purifying and cloning new factors involved in brain function and skin development, and for screening drug candidate compounds.

Claims

請求の範囲 The scope of the claims
1. 下記 (a) から (d) のいずれかに記載の DNA。 1. DNA described in any one of (a) to (d) below.
(a) 配列番号: 2または 1 1に記載のアミノ酸配列からなるタンパク質をコー ドする DNAo  (a) DNAo encoding a protein consisting of the amino acid sequence of SEQ ID NO: 2 or 11
(b) 配列番号: 1または 10に記載の塩基配列のコード領域を含む DNA。  (b) DNA containing the coding region of the nucleotide sequence of SEQ ID NO: 1 or 10.
(c) 配列番号: 2または 1 1に記載のアミノ酸配列において 1若しくは複数の アミノ酸が置換、 欠失、 挿入、 および/または付加したアミノ酸配列からなり、 配列番号: 2または 1 1に記載のアミノ酸配列からなるタンパク質と機能的に同 等なタンパク質をコ一ドする DNA。  (c) an amino acid sequence according to SEQ ID NO: 2 or 11, which comprises an amino acid sequence in which one or more amino acids have been substituted, deleted, inserted and / or added in the amino acid sequence according to SEQ ID NO: 2 or 11; DNA that encodes a protein that is functionally equivalent to a sequenced protein.
( d ) 配列番号: 1または 10に記載の塩基配列からなる DNAとハイブリダイズ する DNAであって、 配列番号: 2または 1 1に記載のアミノ酸配列からなるタン パク質と機能的に同等な夕ンパク質をコードする DNA。  (d) DNA that hybridizes with the DNA having the nucleotide sequence of SEQ ID NO: 1 or 10, and is functionally equivalent to the protein having the amino acid sequence of SEQ ID NO: 2 or 11. DNA that encodes protein.
2. 下記 (a) から (d) のいずれかに記載の DNA。  2. DNA described in any one of (a) to (d) below.
(a)配列番号: 8に記載のアミノ酸配列からなるタンパク質をコ一ドする DNA。  (a) DNA encoding a protein consisting of the amino acid sequence of SEQ ID NO: 8.
(b) 配列番号: 9に記載の塩基配列のコード領域を含む DNA。  (b) DNA containing the coding region of the nucleotide sequence of SEQ ID NO: 9.
(c) 配列番号: 8に記載のアミノ酸配列において 1若しくは複数のアミノ酸が 置換、 欠失、 挿入、 および/または付加したアミノ酸配列からなり、 配列番号: 9に記載のアミノ酸配列からなるタンパク質と機能的に同等なタンパク質をコー ドする DNAo  (c) a protein consisting of the amino acid sequence of SEQ ID NO: 8 with one or more amino acids substituted, deleted, inserted, and / or added, and a protein comprising the amino acid sequence of SEQ ID NO: 9; DNAo that encodes the equivalent protein
(d) 配列番号: 8に記載の塩基配列からなる DNAとハイブリダィズする DNAで あって、 配列番号: 9に記載のアミノ酸配列からなるタンパク質と機能的に同等 なタンパク質をコ一ドする DNA。  (d) DNA that hybridizes with the DNA consisting of the nucleotide sequence of SEQ ID NO: 8 and encodes a protein functionally equivalent to the protein consisting of the amino acid sequence of SEQ ID NO: 9.
3. 請求項 1または 2に記載の DNAが挿入されたべク夕一。  3. A vector into which the DNA according to claim 1 or 2 has been inserted.
4. 請求項 3に記載のベクターが導入された宿主細胞。  4. A host cell into which the vector according to claim 3 has been introduced.
5. 請求項 1または 2に記載の DNAによりコードされるタンパク質。 5. A protein encoded by the DNA according to claim 1 or 2.
6 . 請求項 4に記載の宿主細胞を培養し、 該宿主細胞またはその培養上清から 発現させたタンパク質を回収する工程を含む、 請求項 5に記載の夕ンパク質の製 造方法。 6. The method for producing a protein according to claim 5, comprising a step of culturing the host cell according to claim 4, and recovering the expressed protein from the host cell or a culture supernatant thereof.
7 . 請求項 5に記載の夕ンパク質に対する抗体。  7. An antibody against the protein of claim 5.
8 . 請求項 5に記載の夕ンパク質の部分べプチド。  8. The partial peptide of evening protein according to claim 5.
9 . 配列番号: 1、 8、 または 1 0に記載の塩基配列からなる DNAまたはその 相補鎖とハイブリダィズし、 少なくとも 1 5塩基の鎖長を有するヌクレオチド。 9. A nucleotide having a length of at least 15 bases, which hybridizes with the DNA consisting of the nucleotide sequence of SEQ ID NO: 1, 8, or 10 or a complementary strand thereof.
1 0 . 請求項 4に記載のタンパク質に結合する活性を有する化合物のスクリ一 ニング方法であって、 10. A method for screening a compound having an activity of binding to a protein according to claim 4, wherein
( a ) 請求項 4に記載のタンパク質またはその部分べプチドに被検試料を接触さ せる工程、  (a) contacting a test sample with the protein or the partial peptide thereof according to claim 4,
( b ) 請求項 4に記載のタンパク質またはその部分べプチドに結合する活性を有 する化合物を選択する工程、 を含む方法。  (b) selecting a compound having an activity of binding to the protein according to claim 4 or a partial peptide thereof.
1 1 . 請求項 1 0に記載の方法により単離されうる、 請求項 4に記載のタンパ ク質に結合する化合物。  11. A compound that binds to the protein of claim 4, which can be isolated by the method of claim 10.
1 2 . 請求項 1に記載の DNAによりコ一ドされるタンパク質の活性を調節する 化合物のスクリ一ニング方法であって、  12. A method for screening a compound that regulates the activity of a protein encoded by the DNA according to claim 1,
( a ) 請求項 1に記載の DNAを発現するべクターおよび NF- kBの活性化に応答し てレポ一夕一遺伝子を発現するベクターを含む細胞に被検試料を接触させる工程、  (a) contacting the test sample with a cell containing the vector expressing the DNA according to claim 1 and a vector expressing the repo overnight gene in response to activation of NF-kB;
( b ) 該細胞におけるレポ一夕一活性を検出する工程、 および (b) detecting repo overnight activity in the cells, and
( c ) 被検試料を該細胞に接触させないで検出した場合と比較して、 該レポ一夕 一活性を低下または増加させる化合物を選択する工程、 を含む方法。  (c) a step of selecting a compound that reduces or increases the repo overnight activity as compared to a case where a test sample is detected without contacting the cells.
1 3 . 請求項 1 2に記載の方法により単離されうる、 請求項 1に記載の DNAに よりコードされるタンパク質の活性を調節する化合物。  13. A compound that modulates the activity of a protein encoded by the DNA of claim 1, which can be isolated by the method of claim 12.
1 4 . 天然由来である、 請求項 1 1または 1 3に記載の化合物。  14. The compound according to claim 11 or 13, which is of natural origin.
1 5 . リガンド、 ァゴニスト、 またはアン夕ゴニストである、 請求項 1 1また は 1 3に記載の化合物 c 15. The claim 11, wherein the ligand, agonist, or angelic gonist. Is the compound c described in 13
PCT/JP2000/000940 1999-02-19 2000-02-18 Novel tnf receptor-like proteins WO2000049149A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU25742/00A AU2574200A (en) 1999-02-19 2000-02-18 Novel tnf receptor-like proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP11/41935 1999-02-19
JP4193599 1999-02-19

Publications (2)

Publication Number Publication Date
WO2000049149A1 true WO2000049149A1 (en) 2000-08-24
WO2000049149A9 WO2000049149A9 (en) 2001-03-01

Family

ID=12622094

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2000/000940 WO2000049149A1 (en) 1999-02-19 2000-02-18 Novel tnf receptor-like proteins

Country Status (2)

Country Link
AU (1) AU2574200A (en)
WO (1) WO2000049149A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001038526A1 (en) * 1999-11-23 2001-05-31 Glaxo Group Limited Human tnf receptor
JP2006230356A (en) * 2005-02-28 2006-09-07 Chugai Pharmaceut Co Ltd Transgenic non-human animal inhibiting troy signal
US7223852B2 (en) 1997-09-12 2007-05-29 Biogen Idec Ma Inc. Nucleic acids encoding TRAIN-R: a cysteine rich member of the TNF-receptor family
US7846438B2 (en) 2004-08-03 2010-12-07 Biogen Idec Ma Inc. Methods of promoting neurite outgrowth with soluble TAJ polypeptides

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998038304A1 (en) * 1997-02-27 1998-09-03 Ono Pharmaceutical Co., Ltd. Novel polypeptide, dna encoding the same and use thereof
WO1999011791A2 (en) * 1997-09-05 1999-03-11 University Of Washington Tumor necrosis factor family receptors and ligands, encoding nucleic acids and related binding agents
WO1999013078A1 (en) * 1997-09-12 1999-03-18 Biogen, Inc. Cysteine rich receptors-train
WO1999020644A1 (en) * 1997-10-18 1999-04-29 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO1999033980A2 (en) * 1997-12-30 1999-07-08 Chiron Corporation Members of tnf and tnfr families
WO1999033967A2 (en) * 1997-12-29 1999-07-08 Regeneron Pharmaceuticals, Inc. Novel nucleic acid and polypeptide with homology to the tnf-receptors
WO1999037818A1 (en) * 1998-01-27 1999-07-29 Millennium Pharmaceuticals, Inc. Novel molecules of the tnf receptor superfamily and uses therefor

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998038304A1 (en) * 1997-02-27 1998-09-03 Ono Pharmaceutical Co., Ltd. Novel polypeptide, dna encoding the same and use thereof
WO1999011791A2 (en) * 1997-09-05 1999-03-11 University Of Washington Tumor necrosis factor family receptors and ligands, encoding nucleic acids and related binding agents
WO1999013078A1 (en) * 1997-09-12 1999-03-18 Biogen, Inc. Cysteine rich receptors-train
WO1999020644A1 (en) * 1997-10-18 1999-04-29 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO1999033967A2 (en) * 1997-12-29 1999-07-08 Regeneron Pharmaceuticals, Inc. Novel nucleic acid and polypeptide with homology to the tnf-receptors
WO1999033980A2 (en) * 1997-12-30 1999-07-08 Chiron Corporation Members of tnf and tnfr families
WO1999037818A1 (en) * 1998-01-27 1999-07-29 Millennium Pharmaceuticals, Inc. Novel molecules of the tnf receptor superfamily and uses therefor

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HAMADA, T. ET. AL.: "Isolation and characterization of a novel secretory protein, stromal cell-derived factor-2(SDF-2)using the signal sequence trap method", GENE, vol. 176, 1996, pages 211 - 214, XP002928735 *
PAN, G. ET. AL.: "Identification and functional characterization of DR6, a novel death domain-containing TNF receptor", FEBS LETT., vol. 431, 1998, pages 351 - 356, XP002928733 *
TAN, K. B. ET. AL.: "Characterization of a novel TNF-like ligand and recently described TNF ligand and TNF receptor superfamily genes and their constitutive and inducible expression in hematopoietic and non-hematopoietic cells", GENE, vol. 204, 1997, pages 35 - 46, XP002928734 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7223852B2 (en) 1997-09-12 2007-05-29 Biogen Idec Ma Inc. Nucleic acids encoding TRAIN-R: a cysteine rich member of the TNF-receptor family
US7402658B2 (en) 1997-09-12 2008-07-22 Biogen Idec Ma Inc. TRAIN-R: a cysteine-rich member of the TNF-receptor family
US7838639B2 (en) 1997-09-12 2010-11-23 Biogen Idec Ma Inc. Antibodies to train R: a cysteine-rich member of the TNF-receptor family, and methods of treating tumors expressing said receptor
WO2001038526A1 (en) * 1999-11-23 2001-05-31 Glaxo Group Limited Human tnf receptor
US7846438B2 (en) 2004-08-03 2010-12-07 Biogen Idec Ma Inc. Methods of promoting neurite outgrowth with soluble TAJ polypeptides
JP2006230356A (en) * 2005-02-28 2006-09-07 Chugai Pharmaceut Co Ltd Transgenic non-human animal inhibiting troy signal

Also Published As

Publication number Publication date
AU2574200A (en) 2000-09-04
WO2000049149A9 (en) 2001-03-01

Similar Documents

Publication Publication Date Title
JP4185512B2 (en) TGF-β type receptor cDNA and use thereof
EP1188830B1 (en) Novel hemopoietin receptor protein ,nr10
JP4185070B2 (en) IL-13 receptor polypeptide
AU2020210185C1 (en) Chimeric antigen receptors and uses thereof
JP3615220B2 (en) Fibroblast growth factor receptor
JPH09504030A (en) CNTF family antagonist
JP4044970B2 (en) TNF ligand
US20070243584A1 (en) Tetramerizing polypeptides and methods of use
AU2005318086A1 (en) BCMA polypeptides and uses thereof
CA2299619A1 (en) Human orphan receptor ntr-1
WO2002077230A1 (en) Nr10 splicing variants
NL1010602C2 (en) Nucleic acid encoding a nerve tissue sodium channel protein.
WO2000049149A1 (en) Novel tnf receptor-like proteins
US20220135971A1 (en) Core master regulators of glioblastoma stem cells
WO2000049148A1 (en) Novel cytokine recfptor-like protein
JP2008099690A (en) Novel hemopoietin receptor protein, nr12
JP2839837B2 (en) DNA encoding the ligand-binding domain protein of granulocyte colony-stimulating factor receptor
JP2002531112A (en) Tumor necrosis factor receptor homolog-1
WO2001016298A1 (en) Cloning of mouse gpr10 receptor
WO2002064786A1 (en) Novel gene tcif
US20040137595A1 (en) Mfq-111, a novel human gtpase like protein
US20030138802A1 (en) Novel protein having signal sequence
JP2002000279A (en) Ys 68 gene participating in primary hemopoiesis
JPH05219959A (en) New dna and protein coded therewith

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: C2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGE 18, DESCRIPTION, REPLACED BY A NEW PAGE 18; AFTER RECTIFICATION OF OBVIOUS ERRORS AS AUTHORIZED BY THE INTERNATIONAL SEARCHING AUTHORITY

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 599874

Kind code of ref document: A

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase