WO2000068262A1 - Novel pollen antigen - Google Patents

Novel pollen antigen Download PDF

Info

Publication number
WO2000068262A1
WO2000068262A1 PCT/JP2000/003031 JP0003031W WO0068262A1 WO 2000068262 A1 WO2000068262 A1 WO 2000068262A1 JP 0003031 W JP0003031 W JP 0003031W WO 0068262 A1 WO0068262 A1 WO 0068262A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
antibody
derivative
partial peptide
pollen
Prior art date
Application number
PCT/JP2000/003031
Other languages
French (fr)
Japanese (ja)
Inventor
Hironao Ueno
Kenya Shitara
Nobuo Hanai
Seiji Sugimoto
Keiichi Yano
Akiko Furuya
Original Assignee
Kyowa Hakko Kogyo Co., Ltd.
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 Kyowa Hakko Kogyo Co., Ltd. filed Critical Kyowa Hakko Kogyo Co., Ltd.
Priority to AU44316/00A priority Critical patent/AU4431600A/en
Publication of WO2000068262A1 publication Critical patent/WO2000068262A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/16Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to a diagnosis of an allergic disease containing a novel protein having allergen activity in an allergic disease, a partial peptide of the protein or a derivative thereof, the protein, a partial peptide of the protein or a derivative thereof as an active ingredient.
  • Hay fever is one of the most serious allergic diseases in Japan.
  • pollen that cause allergies, such as cedar, cypress, birch, duckweed, oats, rice and ragweed.
  • cedar pollen accounts for about 80% of hay fever.
  • Cedar hay fever cases have increased sharply since the 1970s, accounting for more than 10% of the population.
  • air pollution caused by NOx in vehicle exhaust gas is also considered to be a cause of cedar pollinosis, and the number of cedar pollinosis patients is still increasing.
  • information such as the amount of cedar pollen scattered is taken up in the media every day, and there is a problem that cedar pollinosis can no longer be neglected in public health.
  • Pollinosis is an allergic disease caused mainly by pollen allergen, an antigenic substance in pollen. Allergy is a tissue disorder that occurs locally or systemically when a living body sensitized to a specific antigen comes in contact with the same antigen again. Allergic reactions are classified as type I-IV according to Coombs and Gell Pollinosis is classified as a type I allergy mediated by IgE, which responds specifically to pollen allergens.
  • Its pathological condition is mainly immediate allergic disease with rhinitis and conjunctivitis as the main symptoms, and it causes watery nasal discharge, sneezing, itching, lacrimation, coughing, and sometimes general malaise (atopy Allergy diseases: supervision, supervised by Yuichi Yamamura, Yoshikazu Giri, Nakayama Shoten, 1992, Allergic diseases; edited by Akimasa Motomoto, Takemasa Nakagawa, Yakuhin Yaku Shuppan Co., Ltd., 1985).
  • the pollen allergen eluted from the pollen particles in contact with the mucous membrane of the nasal cavity, mesotrum, and trachea is taken up by antigen presenting cells such as macrophages. Thereafter, upon reaching the antigen-presenting cell campa tissue incorporating the antigen, a specific immunological response to pollen allergen is induced in the lymph tissue, and IgE that specifically reacts with the allergen is produced.
  • the IgE is in the bloodstream Binds to IgE receptors on mast cells and basophils. At this time, when the pollen allergen reenters the living body, IgE specific to the pollen allergen on mast cells and basophils binds to the invading pollen allergen.
  • the mast cells and the basophils are activated, and chemical mediators such as histamine, serotonin, anaphylactic eosinophil chemotactic factor, neutrophil chemotactic factor, prostaglandin, and peptide leukotriene are circulated from within the cell.
  • chemical mediators such as histamine, serotonin, anaphylactic eosinophil chemotactic factor, neutrophil chemotactic factor, prostaglandin, and peptide leukotriene are circulated from within the cell.
  • histamine histamine acts on the sneezing center rather than the sensory terminal via the HI receptor at the sensory nerve terminal, causing sneezing.
  • hay fever is a symptomatic treatment that mainly administers antiallergic agents, anti-inflammatory agents, etc., and desensitization therapy in which an antigen (allergen extract) extracted from pollen is administered little by little as a desensitizing agent. Partially used together.
  • Anti-allergic agents or anti-allergic agents such as anti-allergic agents mainly acting to suppress release of chemical mediators, anti-histamine agents mainly acting to antagonize histamine at HI receptors, and steroid agents mainly acting to suppress the production of inflammatory mediators Inflammatory agents are used for symptomatic treatment to the last of the above-mentioned mechanism of hay fever, so that allergic symptoms caused by hay fever can be temporarily alleviated, but hay fever cannot be cured.
  • allergen extracts used in desensitization therapy are also used for diagnosis and are indispensable for both treatment and diagnosis of hay fever.
  • the currently used allergen extract is a crudely extracted antigen obtained by extracting pollen in an aqueous solvent and appropriately diluting it.Therefore, even if the same extraction method is used, the composition tends to fluctuate greatly depending on the pollen of the raw material. However, there is a problem that it is extremely non-uniform chemically and it is difficult to obtain a certain desensitizing effect. For this reason, antigen administration for a long period of time may be necessary until the desensitizing effect appears, which also has a problem that a heavy burden is imposed on the patient. Furthermore, the fact that allergen extracts currently used contain many impurities also poses a problem in the effectiveness and reliability of treatment and diagnosis.
  • allergenic proteins derived from cedar pollen have been reported as follows. It has been tell.
  • Japanese Patent Application Laid-Open No. 7-188289 reports a polypeptide having a molecular weight of 55,000 Da and a pI of 9.0
  • Japanese Patent Application Laid-Open No. 7-188291 discloses a polypeptide having a molecular weight of 44,000 to 54,000 Da and ⁇ . Glycoproteins have been reported.
  • cedar pollinosis patients are targeted, such as some patients responding to only one of Cry j and Cry j 2, accurate diagnosis or effective treatment of cedar hay fever can be achieved. Is still insufficient with allergens reported so far.
  • the present invention relates to the following (1) to (32).
  • a protein having the following physicochemical properties (1) A protein having the following physicochemical properties.
  • N-terminal amino acid sequence has an amino acid sequence represented by Xaa—Leu—Phe—Pro—Lys—Glu—Ala—Leu—Pro—Thr at the N-terminus (where Xaa is Glu, Asp, His, Gl y, Arg, Trp or Cys).
  • (V) Stability Does not substantially deactivate even when left in an aqueous solution (pH 7.3) at 4 ° C for 2 months.
  • Xaa preferably represents Glu.
  • Protein ⁇ The protein according to the above (1) or (2), which is derived from cedar pollen.
  • a diagnostic agent for an allergic disease comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof as an active ingredient.
  • a therapeutic drug for an allergic disease comprising, as an active ingredient, the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
  • An agent for preventing an allergic disease comprising as an active ingredient the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
  • An allergic disease desensitizing agent comprising, as an active ingredient, the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
  • Desensitization therapy characterized by repeatedly orally or parenterally administering the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof. .
  • An agent for diagnosing an allergic disease comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
  • a therapeutic drug for an allergic disease comprising as an active ingredient the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
  • a prophylactic agent for an allergic disease comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
  • Allergy disease desensitization comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient. Formulation.
  • the protein according to any one of (1) to (5) above, a partial peptide of the protein or a derivative thereof, and a composition containing a known allergen or a derivative thereof are orally or parenterally administered. Desensitization therapy characterized by repeated administration.
  • a diagnostic agent for an allergic disease comprising the antibody according to any one of (22) to (26) above as an active ingredient.
  • a remedy for allergic disease comprising the antibody according to (1) or (22) to (26) as an active ingredient.
  • a prophylactic agent for allergic diseases which comprises the antibody described in the item (1) of (22) to (26) above as an active ingredient.
  • the present inventors have found that an unknown cedar pollen-derived protein has allergen activity and has properties and properties different from those of known cedar pollen-derived proteins. Further, compared to known cedar pollen-derived proteins, the protein of the present invention shows a stronger binding activity to serum IgE of cedar pollinosis patients, and furthermore, serum IgE antibodies against the purified protein of the present invention. The titer showed a high correlation with the IgE antibody titer against cedar pollen-derived allergen extract used in the existing in vitro diagnosis.
  • the allergic disease of the present invention includes all allergic diseases such as an allergic disease caused by one or more homologous allergens and an allergic disease caused by one or more different allergens.
  • insects such as pollen, house dust and mite, and allergens such as food.
  • -Allergen extracts which are crude extracts from allergens and allergens, and major allergens purified from the allergen extract.
  • Major allergens include Cry j 1 and Cry j 2 derived from cedar pollen, Cha 0 KSuzeki derived from hinoki pollen, et al. Molecular Immunology, Vol. 33, 451, 1996), derived from cypress pollen Bet v Bet v 2, Amb a 1, Amb a 2, derived from ragweed pollen, Der f 1, Der f 2, Der pl, Derp 2, derived from mite bodies, Fel d 1, derived from cat hair, etc. Can be
  • Allergic diseases caused by pollen allergens include allergic diseases caused by multiple types of pollen, and allergic diseases caused by allergens other than pollen in addition to pollen.
  • an allergic disease caused by multiple types of pollen Japanese cedar pollinosis patients have an allergic reaction to S-cypress pollen (Science of hay fever, Yozo Saito 'Tidetake Ide, Doujin Kagaku, 1994
  • An example of an allergic disease caused by allergens other than pollen in addition to pollen is an example in which a cedar pollinosis patient shows an allergic reaction to unrelated allergens other than pollen such as mites.
  • the main symptoms of allergic diseases include allergic rhinitis, allergic conjunctivitis, allergic dermatitis and bronchial asthma.
  • the protein of the present invention is obtained by isolating and purifying from a male flower or a flower bud that forms pollen or pollen. Any pollen can be used as long as the protein of the present invention can be isolated and purified.
  • Any pollen can be used as long as the protein of the present invention can be isolated and purified.
  • Japanese cedar (Cryptomeria japonica) pollen which belongs to the cedar family of the cedar family, may be mentioned, for example.
  • Proteins homologous to the protein of the present invention can also be isolated from pollen that causes allergies other than cedar pollen.For example, pollen such as hinoki, birch, duckweed, ryegrass, rice, and grasshopper can be isolated. can give.
  • the protein of the present invention can be produced by extracting the above-mentioned pollen with a solvent and purifying the resulting extract.
  • pollen collected from a male flower is suspended and suspended in a solvent, and immersed while stirring as necessary.
  • Extraction solvents include buffers or water commonly used by those skilled in the art of protein chemistry.
  • the buffer include a phosphate buffer, a citrate buffer, a Tris-HCl buffer, an acetate buffer and the like.
  • An appropriate salt or hydrophilic organic solvent may be added to the above-mentioned solvent for the purpose of increasing the protein extraction efficiency.
  • the salt include sodium chloride and potassium chloride.
  • the hydrophilic organic solvent include methanol, ethanol, butanol, acetone and the like.
  • the suspension obtained above is centrifuged, and the supernatant is collected. Furthermore, the amount of protein recovered can be increased by repeatedly performing the above-described immersion and centrifugation extraction operations on the residue. Usually, most of the protein can be extracted by repeating this extraction operation 1 to 5 times.
  • Purification means usually used for protein separation and purification include salting out, dialysis, filtration, concentration, centrifugation, various types of chromatography, and electrophoresis.
  • Examples of various types of chromatography include ion exchange chromatography, gel filtration chromatography, hydrophobic chromatography, affinity chromatography, and reverse phase chromatography.
  • Examples of the electrophoresis include polyacrylamide gel electrophoresis, isoelectric focusing, and the like.
  • the allergen activity of the protein obtained above can be detected using the presence and reactivity of specific IgE or specific lymphocytes derived from serum or blood cells of hay fever patients as indicators.
  • Methods for detecting specific IgE or a reaction mediated by specific IgE include a stress test method and an in vitro test method.
  • the stress test method is a test method in which the obtained protein is loaded on a hay fever patient and an allergic reaction is observed.
  • In vitro test methods include enzyme immunoassay and radioimmunoassay using serum from hay fever patients, and leukocytes from hay fever patients.
  • a histamine release test that detects histamine release by a full cell, basophil) force.
  • Means for detecting specific lymphocytes include a proliferative response test using lymphocytes from hay fever patients. (Atopy's allergic disease; supervised by Yuichi Yamamura, Yoshikazu Giri, Nakayama Shoten, 1992)
  • serum obtained from peripheral blood collected from hay fever patients is reacted with the obtained protein immobilized on an appropriate carrier, and the serum for the obtained protein is used.
  • This is a method for detecting specific IgE in the medium, such as ELISA (enzyme-linked immunosorbent assay) and RA f (radio-allergosorbent test).
  • the histamine release test is performed using the obtained protein and the leukocyte fraction containing fertilized cells and basophils prepared from the peripheral blood of a hay fever patient, or the peripheral blood of a healthy human subject to the serum of a hay fever patient. This is a method for detecting histamine released by reacting the obtained leukocyte fraction and activating mast cells or basophils via IgE specific to the obtained protein.
  • the obtained protein is reacted with lymphocytes prepared from the peripheral blood of a hay fever patient, and the T cell or the B cell specific activation reaction or proliferation to the obtained protein is performed. This is a method of detecting a response.
  • partially purified proteins contain pollen antigens such as Cry j 1. Therefore, in order to obtain a higher purity protein, gel filtration chromatography, ion exchange chromatography, affinity chromatography, etc.
  • the purified protein of the present invention can be obtained by applying a combination of methods such as reverse phase chromatography, hydrophobic chromatography, polyacrylamide gel electrophoresis, and isoelectric focusing.
  • a partial peptide having allergen activity can be obtained from the protein of the present invention.
  • the partial peptide of the present invention can be produced from the protein separated and purified by the method (1). That is, the protein can be limitedly degraded by a proteolytic enzyme or a chemical technique, and a partial peptide can be obtained by a method similar to the method for purifying the protein from the obtained reaction mixture.
  • proteolytic enzymes include trypsin, chymotrypsin, pepsin, papain, and V8 protease.
  • the chemical method include a decomposition reaction with bromomethane under acidic conditions of formic acid.
  • a peptide having a partial amino acid sequence of the protein can also be produced by a chemical synthesis method such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method).
  • Fmoc method fluorenylmethyloxycarbonyl method
  • tBoc method t-butyloxycarbonyl method.
  • Kuwawa Trading Advanced ChemTech, USA
  • Perkin-Elmer Japan US Perkin-Elmer
  • Aroca US Protein Technology Instrument
  • Kurabo US SyntheceU-Vega
  • Nippon Perceptive 'Limited US PerSeptive
  • Shimadzu Corporation It can also be produced using a peptide synthesizer such as
  • the partial peptide having an allergen activity of the present invention can be obtained by screening a plurality of peptides having a partial sequence of the amino acid sequence of the protein by a method similar to the method for detecting the allergen activity of a protein.
  • Examples of the method include the above-mentioned 1 (1) enzyme immunoassay using sera from hay fever patients and a proliferation response test using lymphocytes from hay fever patients.
  • the present invention includes derivatives of the protein of the present invention or partial peptides thereof.
  • Derivatives refer to modified forms and modified forms aimed at eliminating or attenuating the reactivity with IgE in a living body.
  • Derivatives can be used alone or in combination with existing protein chemistry or protein chemistry techniques [Biochemistry Experiment Course 1 Protein Chemistry IV, edited by the Biochemical Society of Japan, Neogene Chemistry Experiment Course 1 Protein IV, Biochemistry Society], etc. Can be obtained. For example, after cleaving the disulfide bond of the protein of the present invention in the presence of a reducing agent such as dithiothreitol, the SH group of the resulting cysteine residue is blocked with odoacetic acid or the like to modify the higher-order structure.
  • a reducing agent such as dithiothreitol
  • the protein of the present invention a partial peptide of the protein and a derivative thereof can be used alone or in combination as a diagnostic, preventive or therapeutic agent for allergic diseases.
  • At least one of the protein of the present invention, a partial peptide of the protein and a derivative thereof is mixed with the above-mentioned known allergen to be used as a diagnostic, preventive or therapeutic drug for allergic diseases. You can also.
  • the method for preparing a diagnostic agent for pollen allergic disease using the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is not particularly limited, but the protein and the partial protein purified according to the above-mentioned method are used. Peptides and their derivatives can be concentrated and collected as a liquid, or further freeze-dried and collected as a solid, and can be prepared and used alone or as an appropriate combination agent.
  • the diagnostic agent used in the stress test is not only a single form, but also a physiologically acceptable carrier, excipient, diluent, stabilizer, solubilizer, emulsifier, buffer Agent, soothing agent, preservative, coloring agent and the like.
  • a diagnostic agent used in an in vitro test which is one of the diagnostic methods described later, not only a single form, but also a physiologically, immunologically or immunochemically acceptable carrier, excipient, and dilution can be used. It can be used as a compounding agent to which agents, stabilizers, dissolution aids, emulsifiers, buffers, preservatives, coloring agents and the like are added.
  • the method for preparing a therapeutic or prophylactic agent comprising the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is not particularly limited.
  • a protein purified according to the above-described method, a protein purified from the protein, The partial peptides and their derivatives are concentrated and prepared as a liquid, or further freeze-dried and prepared as a solid, and prepared alone or as an appropriate combination, and used as a therapeutic or prophylactic agent for hay fever. Can be used.
  • prophylactic or therapeutic agent for hay fever can be administered by a usual administration route, for example, an oral, subcutaneous, intradermal, intramuscular, intraperitoneal, intravenous, etc. administration method.
  • Dosage forms include, for example, injections, powders, fine granules, granules, tablets, capsules, sprays, eye drops, nasal drops, cataplasms, lotions, troches, sublingual tablets and the like.
  • the desensitizer containing the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is prepared in the same manner as the above-mentioned drugs, and is used in the same manner as a general desensitizer containing an allergen. be able to.
  • the use example is shown below, but it is not limited to this.
  • a diagnostic agent containing the above-mentioned protein of the present invention, a partial peptide of the protein or a derivative thereof as an active ingredient, or a protein of the present invention, a partial peptide of the protein or a derivative thereof and a known allergen or the allergen Appropriate drugs, dosages and usages are determined based on the results of skin tests using a diagnostic drug or the like whose derivative is the active ingredient.
  • an appropriate amount of the diagnostic agent is administered, and if the wheal diameter or redness diameter at the administration site after 15 to 30 minutes is a certain value or more, the result is considered positive.
  • the protein of the present invention For a patient determined to be positive, the protein of the present invention, a partial peptide of the protein Or a derivative thereof, or the protein of the present invention, or a partial peptide of the protein, orally or parenterally administer the derivative and a known allergen or a desensitizer containing the allergen derivative as an active ingredient. .
  • the dose varies depending on the patient's symptoms and the route of administration.In general, the dose should be about 100 // g or less per adult, preferably 10 zg or less, while observing the course after administration. It is appropriately selected [JP-A-7-179494, JP-A-7-133229], and is administered repeatedly once a week or once a month.
  • desensitization containing 1/10 of the minimum concentration of allergen that is positive in the intradermal test, which is the most sensitive skin test
  • the initial dose is 0.05 ml of the drug. While observing the course of the repeated administration described above, repeat the administration once a week to once a month while gradually increasing the dose so that the local swelling of the administration is within about 5 cm in diameter. Repeated administration is continued up to the upper limit of the dose at which the swelling of the administration site is about 5 cm in diameter.
  • a desensitizer containing a partial peptide of the protein having such properties or a derivative thereof as an active ingredient has a desensitizing effect similar to a normal desensitizer, and further has the advantage of avoiding an anaphylactic reaction. It is also useful as a sensitizer with low side effects.
  • Methods for diagnosing allergic diseases include a stress test in which a subject is directly loaded with an allergen to observe an allergic reaction, and an in vitro test in which a sample prepared from the subject's tissue is used to detect reactivity with the allergen. can give.
  • Samples prepared from the subject's tissue include serum separated from peripheral blood, leukocytes such as mast cells and basophils, and lymphocytes.
  • Examples of the load test include a skin test and a provocation test.
  • In vitro tests include enzyme immunoassays and radioimmunoassays that detect allergen-specific IgE, histamine release tests that detect histamine release of mast cells and basophils by activation through allergen-specific IgE, Proliferation response tests that detect allergen-specific lymphocyte proliferation responses are examples.
  • Allergen-loaded tissues such as skin tests and challenge tests include skin, nose, ocular mucosa, and respiratory tract.
  • Skin testing methods include prick test, scratch test and intradermal test. I can do it. In the brick test and scratch test, an allergen is loaded on the subject's epithelium, and in the intradermal test, the allergen is loaded on the subject's skin.
  • the provocation test includes a nasal mucosal provocation test, an ocular mucosal provocation test, and an inhalation provocation test. Allergens are loaded into the nasal mucosa of the subject in the nasal mucosa provocation test, to the subject's eye mucosa in the ocular mucosa provocation test, and to the subject's airway in the inhalation provocation test.
  • Stress is the reaction of an allergen while scratching or pecking the epithelium in the case of a brick test or a scratch test.
  • the allergen is injected intradermally.
  • a provocation test Spraying, applying, and inhaling an allergen solution, and sticking an allergen-adhered paper piece to a mucous membrane.
  • test in a test tube is described below.
  • Enzyme immunoassay and radioimmunoassay are methods for detecting IgE bound to allergen by reacting serum in the blood collected from the subject with an allergen immobilized on a suitable carrier.
  • RAST radio-allergosorbent est
  • the histamine release test involves reacting an allergen with a leukocyte fraction, such as mast cells and basophils, prepared from the peripheral blood of a subject, or a leukocyte fraction prepared from the peripheral blood of a healthy subject to which the serum of the subject has been reacted. This is a method for detecting histamine released by the method.
  • the lymphocyte proliferation test is a method for detecting the activation or proliferative response of T cells or B cells that occurs when allergen is added to lymphocytes separated from the peripheral blood of a subject.
  • antibodies of the present invention include monoclonal antibodies, polyclonal antibodies, and antisera containing them.
  • Examples of the monoclonal antibody of the present invention include antibodies produced by hybridomas, humanized antibodies, human antibodies, and the like.
  • a hybridoma is a monoclonal antibody having a desired antigen specificity obtained by cell fusion of B cells obtained by immunizing a non-human mammal with an antigen and myeloma cells derived from a mouse or the like. Means a cell that produces
  • humanized antibody examples include a human-type chimeric antibody, a human-type homology determining region (hereinafter, abbreviated as CDR) -grafted antibody, and the like.
  • CDR human-type homology determining region
  • the human chimeric antibody is composed of an antibody heavy chain variable region (hereinafter, the heavy chain is referred to as an H chain, the variable region is referred to as HV or VH as a V region) and an antibody light chain variable region (hereinafter, referred to as a light chain,
  • the L chain is also called LV or VL, and the heavy chain constant region of a human antibody And the light chain constant region of a human antibody (hereinafter also referred to as CL).
  • any animal can be used as long as hybridomas can be produced, such as mice, rats, hamsters, and rabbits.
  • the human chimeric antibody of the present invention obtains cDNAs encoding VH and VL from a hybridoma that produces a monoclonal antibody that specifically reacts with the pollen antigen of the present invention, and obtains human antibody CH and human antibody CL.
  • a human-type chimeric antibody expression vector can be constructed by inserting each into an expression vector having a gene to be encoded, and can be expressed and produced by introducing into a host cell.
  • any CH can be used as long as it belongs to human immunoglobulin (hereinafter, referred to as hlg).
  • the hlgG class is preferable, and the higGl and hIH belong to the hlgG class.
  • g G2, hIgG3, any subclass such hIgG4 can be used.
  • the CL of the human chimeric antibody any CL belonging to hlg can be used, which can be of ⁇ class or ⁇ class.
  • the human CDR-grafted antibody means an antibody obtained by grafting the amino acid sequences of CDRs of VH and VL of an antibody of a non-human animal at appropriate positions of VH and VL of a human antibody.
  • the human CDR-grafted antibody of the present invention is obtained by grafting VH and VL CDR sequences of a non-human animal antibody specifically reacting with the pollen antigen of the present invention into VH and VL CDR sequences of any human antibody.
  • CDNA encoding the region is constructed and inserted into an expression vector having genes encoding human antibody CH and human antibody CL, respectively, to construct a human CDR-grafted antibody expression vector, and the expression vector Can be expressed into a host cell to express and produce a human CDR-grafted antibody.
  • the CH of human CDR-grafted antibody may be any so long as it belongs to the hlg, are preferred those hlgG class, further hI g Gl belonging to hlgG class, hIgG2, hIgG3, any subclass such hIgG4 Can be used.
  • the CL of the human-type CDR-grafted antibody any CL can be used as long as it belongs to hlg, or a ⁇ class or ⁇ class.
  • Human antibodies originally mean antibodies naturally occurring in the human body, but human antibody phage libraries and human antibody production produced by recent advances in genetic engineering, cell engineering, and developmental engineering techniques. Antibodies and the like obtained from transgenic animals are also included.
  • Antibodies present in the human body can be isolated, for example, by isolating human peripheral blood lymphocytes, infecting and immortalizing EB virus, etc., and cloning the lymphocytes that produce the antibodies.
  • the antibody can be purified.
  • the human antibody phage library is a phage gene encoding antibody gene prepared from human B cells. This is a library in which antibody fragments, such as Fab and single-chain antibodies, are expressed on the phage surface by insertion into a phage. From the library, phages expressing antibody fragments having the desired antigen-binding activity can be recovered using the binding activity to the substrate on which the antigen is immobilized as an index. The antibody fragment can be further converted to a human antibody molecule consisting of two complete H chains and two complete L chains by genetic engineering techniques.
  • a human antibody-producing transgenic animal means an animal in which a human antibody gene has been integrated into cells.
  • a human antibody-producing transgenic animal can be produced by introducing a human antibody gene into mouse ES cells, transplanting the ES cells into an early embryo of another mouse, and then developing the embryo.
  • Production of human antibodies from human antibody-producing transgenic animals can be performed in a culture by obtaining human antibody-producing antibodies and ipridomas by the usual method for producing hybridomas in mammals other than humans, and culturing them. It can be performed by producing and accumulating human antibodies.
  • the antibodies of the present invention also include the antibody fragments described below.
  • antibody fragments include Fab (abbreviation of fragment of antigen binding), Fab ′, F (ab ′) 2 , single chain antibody (hereinafter abbreviated as scFv), disulfide stabilized antibody (disulfide sta bilized). Fv; dsFv), peptides including CDRs, and the like.
  • Fab is a fragment obtained by treating IgG with proteolytic enzyme papain (which is cleaved at amino acid residue 224 of H chain). It is an antibody fragment having a molecular weight of about 50,000 and having antigen-binding activity, which is linked by a disulfide bond.
  • the Fab of the present invention can be obtained by treating an antibody that specifically reacts with the pollen antigen of the present invention with the proteolytic enzyme papain.
  • a DNA encoding the Fab of the antibody is inserted into an expression vector, and the vector is expressed by introducing the vector into a host cell to produce a Fab;
  • F (ab ') 2 is a fragment obtained by treating IgG with the protease pepsin (which is cleaved at the 234th amino acid residue in the H chain).
  • Fab binds via a disulfide bond in the hinge region. This is an antibody fragment having a molecular weight of about 100,000 and having antigen-binding activity, which is slightly larger than that obtained.
  • the F (ab ') 2 of the present invention can be obtained by treating an antibody specifically reacting with the pollen antigen of the present invention with the protease pepsin.
  • the following Fa has a thioether bond! /, Is a disulfide bond and can be made S.
  • Fab ' is an antibody fragment having a molecular weight of about 50,000 and having an antigen-binding activity in which a disulfide bond in the hinge region of F (ab') 2 is cleaved.
  • Fab ′ of the present invention is a reducing agent dithios that reduces F (ab ′) 2 which specifically reacts with the pollen antigen of the present invention. It can be obtained by late processing. Alternatively, Fab ′ can be expressed and produced by inserting a DNA encoding the Fab ′ fragment of the antibody into an expression vector and introducing the vector into a host cell.
  • scFv refers to a VH-P-VL or VL-P-VH polypeptide in which one VH and one VL are linked using an appropriate peptide linker (hereinafter, referred to as P).
  • VH and VL contained in the scFv of the present invention can be used for the antibody, humanized antibody, and human antibody produced by the hybridoma of the present invention.
  • the scFv of the present invention obtains cDNA encoding VH and VL of an antibody that specifically reacts with the pollen antigen of the present invention, constructs a scFv-encoding DNA, and inserts the DNA into an expression vector.
  • the vector can be expressed by introducing the vector into a host cell to produce an scFv.
  • dsFv is obtained by binding a polypeptide in which one amino acid residue in each of VH and VL is replaced with a cysteine residue via a disulfide bond between the cysteine residues.
  • the amino acid residue to be substituted for the cystine residue can be selected based on the prediction of the three-dimensional structure of the antibody according to the method shown by Reiter et al. [Protein Engineering, 7, 697 (1994)].
  • the VH and VL contained in the dsFv of the present invention any of the antibodies, humanized antibodies, and human antibodies produced by the hybridoma of the present invention can be used.
  • the dsFv of the present invention obtains cDNA encoding VH and VL of an antibody that specifically reacts with the pollen antigen of the present invention, constructs a DNA encoding dsFv, and inserts the DNA into an expression vector. Then, the vector can be expressed by introducing the vector into a host cell to produce dsFv.
  • the peptide containing the CDR comprises at least one region of the H chain or L chain CDR. Multiple CDRs can be linked directly or via a suitable peptide linker.
  • the peptide containing the CDR of the present invention can be obtained by obtaining cDNA encoding VH and VH of an antibody specifically reacting with the pollen antigen of the present invention, constructing a DNA encoding the CDR, and expressing the DNA.
  • the peptide can be expressed by introducing the expression vector into a host cell to produce a peptide containing CDR.
  • the peptide containing CDR can also be produced by a chemical synthesis method such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the t Boc method (t-butyloxycarbonyl method).
  • Fmoc method fluorenylmethyloxycarbonyl method
  • t Boc method t-butyloxycarbonyl method
  • the monoclonal antibody of the present invention is prepared by preparing an antigen, immunizing an animal, and immunizing the animal.
  • a hybridoma is prepared by fusing cells with myeloma cells, and the hybridoma is cultured or administered to an animal to cause the animal to undergo ascites carcinoma, and the culture solution or ascites is collected.
  • polyclonal antibodies can be prepared by collecting immune serum from immunized animals.
  • the animals are immunized as follows. That is, an antigen prepared as described below is immunized subcutaneously, intravenously, or intraperitoneally in a non-human mammal, such as a mouse, rat, or hamster, and spleen cells, lymph nodes, and peripheral blood of the animal are immunized. The antibody producing cells are collected.
  • the protein of the present invention is purified by the above-described method, or a recombinant vector containing a DNA encoding the protein of the present invention, a partial peptide of the protein or a derivative thereof is transformed into Escherichia coli or yeast. And a method of expressing a protein or polypeptide by introducing it into a host such as an animal cell or an insect cell. Alternatively, a polypeptide having a partial sequence can also be obtained by synthesizing using an amino acid synthesizer.
  • the obtained antigen is combined with a carrier protein having high antigenicity (keyhole limpet hemocyanin, bovine serum albumin, etc.) by chemical modification or the like, or administered, or an appropriate adjuvant (eg, Freund's). Complete adjuvant (Freund's Complex Adjuvant), aluminum hydroxide gel and B. pertussis vaccine.
  • the antigen is administered after the first dose: 5 to 10 times every 2 weeks. Blood is collected from the fundus venous plexus 3 to 7 days after each administration, and the reaction of the serum with the antigen is determined by an enzyme immunoassay [Antibodidies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988J! I will. A non-human mammal whose serum shows a sufficient antibody titer against the antigen used for immunization is provided as a source of the antibody-producing cells.
  • the polyclonal antibody is obtained by separating and purifying serum obtained from the animal.
  • myeloma cells cell lines obtained from mice are used.
  • 8-azaguanine-resistant mouse derived from BALB / c
  • myeloma cell line P3-X63Ag8-U1 P3-U1
  • P3-U1 Current Topics in Microbiology and Immunology, 18: 1-7, 1978
  • P3_NS1 / 1-Ag41 NS_l
  • SP2 / 0-Agl4 SP2 / 0-Agl4
  • SP-2 SP2 / 0-Agl4
  • P3-X63- Ag8653 6
  • J. Immunology, 123: 1548-1550, 1979 P3-X63-Ag8 (X63) (Nature, 256: 495-497, 1975) and the like
  • P3-X63Ag8 X63
  • the antibody-producing cells and myeloma cells obtained above are mixed, suspended in a HAT medium (a medium in which hypoxanthin, thymidine and aminopterin are added to a normal medium), and cultured for 7 to 14 days. After the culture, a part of the culture supernatant is taken and selected by enzyme immunoassay or the like. Then, the clones were cloned by limiting dilution, and those with a stable high antibody titer determined by enzyme immunoassay were selected as anti-pollen antigen monoclonal antibody-producing hybridoma strains. Select.
  • HAT medium a medium in which hypoxanthin, thymidine and aminopterin are added to a normal medium
  • a monoclonal antibody can be produced by culturing the above-described hybridoma cells or administering the hybridoma cells into the peritoneal cavity of an animal to cause the animal to develop ascites cancer, and collecting and purifying the culture solution or ascites.
  • Methods for purifying polyclonal and monoclonal antibodies include centrifugation, ammonium sulfate precipitation, and hydroprilic acid precipitation, or DEAE-Sepharose column, anion exchange column, protein A or G-column, or gel filtration. Use a column! /, And a method of combining chromatography and the like.
  • the monoclonal antibody of the present invention obtained by the above-mentioned method includes the monoclonal antibody KM2715.
  • the hybridoma KM2715 which produces the monoclonal antibody KM2715, was contacted by the Ministry of International Trade and Industry, Institute of Industrial Science and Technology, Institute of Biotechnology and Industrial Technology (1-3 1-3 Tsukuba East, Ibaraki, Japan) based on the Budapest Treaty. Deposited as FERM BP-7149.
  • the antibody or antibody fragment thereof obtained in the above 5 can be used as a diagnostic, therapeutic or prophylactic agent for allergic diseases.
  • Diagnostic, therapeutic or prophylactic agents comprising the antibody of the present invention or an antibody fragment thereof as an active ingredient are allergic diseases comprising the protein of the present invention described in 2 above, a partial peptide of the protein and a derivative thereof as an active ingredient. It can be prepared using the same method as that for preparing diagnostic, therapeutic and prophylactic agents.
  • a composition comprising the antibody or an antibody fragment thereof of the present invention may be used in a method for treating nose of an allergic disease patient. It can be administered or administered directly to mucosal tissues such as the mouth and respiratory tract, the skin, the eyes, etc., to prevent allergens from entering the body, thereby treating or preventing allergic diseases. Further, it is possible to treat or prevent an allergic disease by removing the allergen that has entered the body by intravenously administering a composition comprising the antibody or the antibody fragment thereof of the present invention to a patient with an allergic disease. it can.
  • Examples of a method for diagnosing an allergic disease using an antibody against the pollen antigen of the present invention or an antibody fragment thereof include a method for immunologically quantifying or detecting the pollen antigen of the present invention.
  • Methods include fluorescent antibody method, immunoenzymatic antibody method (ELISA), radioactive substance-labeled immunological antibody method (RIA), immunocytostaining method, western blotting method, immunoprecipitation method, Enzyme immunoassay, sandwich EL1SA method [Monoclonal antibody experiment manual (Kodansha Scientific, 1987), Seismic Chemistry Laboratory Course 5 Immunobiochemical research method (Tokyo Kagaku Dojin, 1986)] Is raised.
  • the antibody antibody method should be performed using the method described in the literature [Monoclonal Antibodies: Principles and practice, Third edition (Academic Press, 1996), Monoclonal antibody experiment manual (Kodansha Scientific, 1987)], etc. Can be.
  • the antibody of the present invention is applied to tissues exposed to pollen antigens such as nasal mucosa, palatal mucosa, respiratory mucosa, and skin isolated from a living body, or body fluids such as tears and nasal secretions, blood and blood cells, and the like.
  • the fluorescent dye is measured with a fluorometer or flow cytometer. This is a method for measuring or measuring pollen antigens at a time.
  • the immunoenzyme-linked immunosorbent assay is applied to tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, airway mucosa, and skin isolated from the body, or body fluids such as tears and nasal secretions, blood, and blood cells.
  • pollen antigens such as nasal mucosa, palate mucosa, airway mucosa, and skin isolated from the body, or body fluids such as tears and nasal secretions, blood, and blood cells.
  • pollen antigens such as nasal mucosa, palate mucosa, airway mucosa, and skin isolated from the body, or body fluids such as tears and nasal secretions, blood, and blood cells.
  • an enzyme such as peroxidase or biotin
  • Radiolabeled immunoassay is applied to tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, respiratory mucosa and skin isolated from the body, or body fluids such as tears and nasal secretions, blood and blood cells, etc. After reacting the antibody of the present invention or an antibody fragment thereof, and further reacting with an anti-immunoglobulin antibody which has been subjected to radiolabeling, measurement is performed using a scintillation counter or the like to quantify or detect pollen antigens. is there.
  • the antibody of the present invention or an antibody fragment thereof is reacted with intranasally separated nasal mucosa, palate mucosa, respiratory mucosa, skin and tissue exposed to ivy pollen antigen, blood cells, and the like.
  • a fluorescent substance such as fluorescin'isothiosinate (FITC), an anti-immunoglobulin antibody labeled with an enzyme such as peroxidase, and the like are reacted, and then observed using a microscope.
  • the Western blotting method uses SDS-polysaccharide to isolate tissues exposed to pollen antigens, such as nasal mucosa, palate mucosa, respiratory mucosa, and skin, or body fluids such as tears and nasal secretions, blood, and blood cells.
  • pollen antigens such as nasal mucosa, palate mucosa, respiratory mucosa, and skin
  • body fluids such as tears and nasal secretions, blood, and blood cells.
  • acrylamide gel electrophoresis Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988
  • the gel was blotted onto a PVDF membrane or a nitrocellulose membrane, and the antibody of the present invention or its antibody was applied to the membrane.
  • Antibody fragment And then reacting with a fluorescent substance such as FITC, an anti-IgG antibody labeled with an enzyme such as peroxidase, and the like, followed by confirmation.
  • the immunoprecipitation method refers to the nasal mucosa, palatal mucosa, airway mucosa, tissues exposed to pollen antigens such as skin, or body fluids such as tears and nasal secretions, blood and blood cells, etc. of the present invention.
  • a carrier having a specific binding ability to immunoglobulin such as protein G-sepharose is added to precipitate an antigen-antibody complex.
  • the sandwich ELISA method is a method using two kinds of antibodies having different antigen recognition sites in the antibody of the present invention or an antibody fragment thereof. Specifically, one antibody or antibody fragment is adsorbed on a plate in advance, and the other antibody or antibody fragment is labeled with a fluorescent substance such as FITC or an enzyme such as peroxidase in advance. Tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, respiratory mucosa and skin isolated from the body, body fluids such as tears and nasal secretions, blood and blood cells, etc. were reacted on the antibody adsorption plate and labeled. This is a method in which an antibody or an antibody fragment thereof is reacted and a reaction is performed according to the labeling substance.
  • a fluorescent substance such as FITC or an enzyme such as peroxidase
  • the protein of the present invention, the partial peptide of the protein, and their derivatives can be detected or quantified from the pollen extract using the immunological method described in 8 above.
  • the diagnostic, therapeutic, prophylactic and desensitizing agents for allergic diseases consisting of the allergen extract described in 2 or 3 above must be kept constant in activity and quality.
  • allergen extracts are extracts obtained using a plant-based solvent, it is difficult to maintain constant activity and quality.
  • the novel pollen antigen of the present invention is considered to be a major allergen because it has high allergen activity and excellent storage stability. Therefore, keeping the content and properties of the pollen antigen in the allergen extract constant results in an allergen extract having constant activity and quality.
  • an antibody capable of specifically detecting and quantifying the pollen antigen can be a simple and useful tool for examining the pollen antigen.
  • Examples of the method for quantifying the pollen antigen include the immunological measurement method described in the above item 8, preferably a sandwich ELISA.
  • Examples of a method for measuring the stability of the pollen antigen in an allergen extract include the Western blotting method described in the above item 8 and the like.
  • the antibody of the present invention or the antibody fragment thereof is obtained by extracting the protein from the pollen protein extract. Can be used to isolate and purify.
  • FIG. 1 shows the results obtained by performing sodium dodecyl sulfate-polyacrylamide gel electrophoresis (hereinafter referred to as SDS-PAGE) (7.5% polyacrylamide gel concentration) for the purpose of measuring the molecular weight of the protein of the present invention. It is a figure showing the result of having detected the protein component by staining.
  • SDS-PAGE sodium dodecyl sulfate-polyacrylamide gel electrophoresis
  • FIG. 2 is a diagram showing the results of analysis of the reactivity of IgE in the sera of 12 cedar pollinosis patients and 4 healthy subjects with the protein of the present invention by enzyme immunoassay.
  • IgE bound to the antigen was removed. Detected by anti-human IgE antibody.
  • CE unpurified total protein fraction
  • Fig. 3 shows the IgE antibody titer specific to the cedar pollen antigen by the cap RAST FEIA and the antigens of the protein (New), unpurified total protein fraction (CE), Cry j 1 and Cry j 2 of the present invention. It is a scatter diagram showing a correlation with a specific IgE antibody titer.
  • FIG. 4 is a diagram showing the results of analyzing the reaction specificity of the monoclonal antibody KM2715 of the present invention by an enzyme immunoassay.
  • FIG. 3 is a diagram comparing the reactivity of the monoclonal antibody with the antigen peptide-THY conjugate immobilized at 5 / ig / ml and the control peptide THY conjugate.
  • FIG. 5 shows the results of Western blotting analysis of the specific detection of the protein in the unpurified total protein fraction (CE) by the monoclonal antibody KM2715 of the present invention.
  • FIG. 2 is a diagram showing 2 ⁇ g of CE subjected to SDS-PAGE and Western blotting. BEST MODE FOR CARRYING OUT THE INVENTION
  • PBS phosphate-buffered salin; hereinafter, referred to as PBS
  • soluble components are eluted while stirring at 4 ° C for 4 hours, and centrifuged.
  • the supernatant was recovered.
  • the residue was further washed with 2 L of PBS, the same elution procedure was performed, and the supernatant was recovered and combined with the supernatant obtained the first time to obtain a crude extract fraction.
  • ammonium sulfate was added to the crude extract to 80% saturation, and the mixture was allowed to stand at 4 ° C for 2 hours to salt out protein components.
  • the salted out product collected by centrifugation is dissolved in 100 ml of 50 mM Tris-HC1 buffer (pH 7.8) and dialyzed twice for 4 to 12 hours against 2 L of the same buffer at 4 ° C. After equilibration with 50 mM Tris-HC1 buffer ( ⁇ 7.8), the mixture was passed through 10 ml of DEAE-Sepharose CL6B (Pharmacia). Further, 30 ml of a 50 mM Tris-HC1 buffer (pH 7, 8) was passed, and 130 ml of the non-adsorbed fraction was collected.
  • This non-adsorbed fraction was dialyzed twice at 4 ° C against a 10 mM acetic acid aqueous solution containing 200 mM NaCl (NaC triacetic acid aqueous solution: ⁇ 3.4) for 4 to 12 hours, and then subjected to NaC triacetic acid aqueous solution in advance.
  • the column was passed through 10 ml of equilibrated CM-Sepharose CL6B (Pharmacia).
  • the adsorbed components are separated under a continuous NaCl concentration gradient from 0 to 1 M using an aqueous solution of NaC-to-acetic acid and an aqueous solution of 10 mM acetic acid (PH3.4) containing 1 M NaCl. I let it.
  • the absorbance at 280 nm of each fraction was measured, and the gel was stained with SDS-PAGE to detect protein components.
  • the protein of the present invention was recovered at a NaCl concentration of around 0.45 M. Was. This eluted fraction contained Cry j 1, unidentified protein and the like in addition to the protein.
  • this crude fraction is concentrated by centrifugation using an ultrafiltration membrane Centricon 10 (manufactured by Amicon), equilibrated with PBS in advance, and passed through a column of Super Loose 12 (manufactured by Pharmacia) to gel. Filtration was performed. The fraction having a maximum molecular weight of 110,000 ⁇ 15,000 Da was collected as a fraction containing the protein. This fraction contained the protein and one unidentified protein.
  • the molecular weight markers for gel filtration include glutamate dehydrogenase (290,000 Da), lactate dehydrogenase (140,000 Da;), enolase (67,000 Da), adenylate kinase (32,000 Da), and cytochrome c. (12,400 Da) was used.
  • ion exchange chromatography was performed by a batch method. That is, the fraction collected by gel filtration was dialyzed twice against 10 mM acetate buffer (pH 5.0) containing 200 mM NaCl for 4 to 12 hours, and then equilibrated with the same buffer in a microcentrifuge tube in advance. The mixture was reacted with 50 ⁇ l of CM-Sepharose CL6B (Pharmacia) placed for 30 minutes. After the reaction, the non-adsorbed fraction was collected as a purified sample of the protein. The purified sample was used in the following tests after buffer replacement with ⁇ S as necessary. The yield of the protein was 0.4 ⁇ / g of cedar pollen used as a raw material, as determined by converting it into serum albumin using a BCA kit (Pierce).
  • a major band was detected at a position corresponding to a molecular weight of 51,000 soils and 5,000 Da under reducing conditions of 2-mercaptoethanol-added calo. Under non-reducing conditions, a major band was detected at a position corresponding to a molecular weight of 48,000 ⁇ 5,000 Da.
  • the molecular weight markers of SDS-PAGE include myosin (200,000 Da),] -galactosidase (116,300 Da), phosphorylase b (97,400 Da), and serum albumin (66,300 Da). Aldolase (42,400 Da;), carbonic anhydrase (30,000 Da), trypsin inhibitor (20,100 Da), and lysozyme (14,400 Da) were used.
  • the N-terminal amino acid sequence of the purified protein was analyzed by an ordinary protein chemistry method using an automated protein sequencer (Shimadzu, PPSQ-10) utilizing the Edman degradation method.
  • a sequence represented by Xaa—Leu_Phe_Pro—Lys—Glu—Ala—Leu—Pro—Thr was confirmed.
  • Xaa indicates any of Glu, Asp, His, Gly, Arg, Trp, or Cys
  • the protein having the binding activity to IgE in the serum of patients with cedar pollinosis of the present invention was analyzed by the following IgE-ELISA.
  • the cedar pollinosis patients were those who showed high reactivity (+4 grade) to sugya allergen by testing with a cap RAST FEIA allergen test diagnostic reagent manufactured by Pharmacia Upjohn. It is.
  • the purified protein was adjusted to 3 g / ml in PBS, added to a 96-well mic opening plate at 50/1 / well, allowed to stand at 4 ° C for 16 hours, and immobilized on the plate. After washing each well of the plate with PBS, PBS (B-PBS) containing l% (w / v) BSA was added at 100 1 / well, and the plate was allowed to stand at room temperature for 2 hours. After washing each well with PBS, add 50 ⁇ l / well of cedar pollinosis patient and healthy human serum diluted 2-fold with B-PBS, and react with the immobilized protein at 4 ° C for 16 hours. I let it.
  • T-PBS After washing with PBS containing 0.05% (v / v) Tween20 (T-PBS), 50 ⁇ l of biotin-labeled goat anti-HgE antibody (AmericanQualex, code: A116 BN) diluted 1,000-fold with B-PBS was used. 1 / well was added thereto, and the reaction was further performed at room temperature for 1 hour. After washing with T-PBS, avidin D-conjugated horseradish peroxidase (VEC) diluted 4,000-fold with B-PBS TOR Co., Code; A-2004) was added at 50 / l / well, and left at room temperature for 30 minutes.
  • VEC horseradish peroxidase
  • the solubility of the protein of the present invention in various solvents was examined. It was soluble in water, saline and phosphate buffer.
  • the aqueous solution dissolved in (5) was stored in a refrigerator at 4 ° C. Two months later, when the experiment (4) was performed using the aqueous solution, the same result as that obtained two months ago was obtained.
  • the protein having the above physicochemical properties was not known until now, and was determined to be a novel substance.
  • the purified proteins Cry j 1 and Cry j 2 were prepared in PBS at 3 ⁇ g / ml and CE at 30 / ig / ml in PBS. The solution was added in 50-well portions, and allowed to stand at 4 ° C for 16 hours to immobilize on a plate. After each well was washed with PBS, B-PBS was added at 100 / i1 / well and left at room temperature for 2 hours. After washing each well with PBS again, 50 ⁇ l / well of serum of a Japanese cedar pollinosis patient diluted 2-fold with B-PBS was used. And reacted at 4 ° C. for 16 hours.
  • the ABTS chromogenic substrate solution was added at 100 ⁇ l / ⁇ l at room temperature and reacted at room temperature. After color development, the reaction was stopped by adding a 5% SDS aqueous solution at 100 ⁇ l / well. The absorbance of the developed substrate was measured at a wavelength of 415 wavelengths and used as an index of the antibody titer of IgE specific to the antigen protein.
  • the diagnostic reagent used for the diagnosis of cedar pollinosis uses the total protein fraction extracted from cedar pollen as an antigen and measures the antibody titer of serum IgE specific to cedar pollen allergen Things.
  • a cap RAST FEIA manufactured by Pharmacia's Upjohn Co., Ltd. (a spear allergen binding cap; code No.CT17, an enzyme-labeled antibody reagent, and a reference reagent), which is generally used as this kind of diagnostic method, is used. ) was used as a control.
  • Cap RAST FEIA was performed according to the method recommended by Pharmacia's Upjohn.
  • the sample serum was reacted with the cap on which the total cedar pollen allergen (unpurified total protein fraction) had been previously immobilized, washed, and then reacted with an enzyme-labeled anti-IgE antibody. After further washing, the fluorescent substance generated by adding the substrate solution was detected by a fluorometer, and IgE specifically bound to the allergen was quantified.
  • Fig. 2 shows the results of IgE-ELISA.
  • a specific IgE antibody against the protein was detected, indicating that the protein was an allergen in cedar pollinosis.
  • no specific IgE antibody against the protein was detected in healthy subjects.
  • Anti-New indicates an antibody against the protein of the present invention.
  • a comparison of the ratio obtained by dividing the specific IgE antibody titer against the protein by the specific IgE antibody titer against Cry j 1 (the protein ZCry j 1) showed that 9 out of 24 patients showed 1.3 or more.
  • Fig. 3 shows a two-variable scatter plot of 12 cedar pollinosis patients and 4 healthy subjects.
  • Cap RAST FEIA and IgE-ELISA use different antigen immobilization methods, detection methods, and enzymes / substrates for detection, but specific IgE antibody titers with cap RAST FEIA and unpurified IgE-EL1SA
  • the correlation coefficient with the total protein-specific IgE antibody titer is 0.921. In other words, the evaluation of the specific IgE antibody titer performed this time was not affected by differences in the measurement system.
  • the specific IgE antibody titer against the protein in the serum of cedar pollinosis patients is equivalent to the specific IgE antibody titer against Cry j1 and Cry j2, even though the ratio of the protein to the total protein of cedar pollen is extremely low. It was more expensive. A single protein exhibiting such high allergen activity is not yet known. Therefore, it can be said that the protein has extremely high antigenicity and is a very important causative substance in the onset of cedar pollinosis.
  • a peptide (SEQ ID NO: 2) with Cys added to the C-terminal Thr of the N-terminal amino acid sequence shown in SEQ ID NO: 1 obtained in 2 (2) above for binding to a carrier protein (SEQ ID NO: 2) Tide synthesis was performed.
  • an automated peptide synthesizer P SSM-8 manufactured by Shimadzu Corporation
  • Synthetic peptides and KLH to enhance immunogenicity was used as an immunogen.
  • KLH is dissolved in PBS to adjust to 10 mg / ml, 25 mg / ml MBS [N- (maleimidobenzoyloxy) succini mide, manufactured by Nacalai Tesque, Inc.] of 1/10 volume of the KLH solution is added dropwise and stirred for 30 minutes. I let it. Free MBS was removed using a Sephadex G-25 column (Pharmacia), a gel filtration column equilibrated with PBS in advance, to obtain 2.5 mg of a KLH-MBS conjugate. Furthermore, it was mixed with 1 mg of peptide dissolved in 0.1 M sodium phosphate buffer (PH 7.0), and reacted by stirring at room temperature for 3 hours. After the reaction, what was dialyzed against PBS-0.5 MN & C1 was obtained as an antigen peptide-KLH conjugate, which was used as an immunogen.
  • MBS N- (maleimidobenzoyloxy) succini mide, manufactured by Nacalai Tesque
  • antigen was prepared by the method shown in the peptide - KLH Konjiyuge M00 / g to Mizusani ⁇ Honoré mini ⁇ Moore Ho cement (Antibodies - A Laboratory Manual, Cold Sprin g Harbor Laboratory, P 99, 1988 ) 2 mg and pertussis vaccine (manufactured by Chiba Prefectural Serum Research Institute) were administered to 5-week-old female mice (Balb / c) together with 1 ⁇ 10 9 cells, and after 2 weeks, 100 // g of antigen ⁇ peptide KLH conjugate was administered. It was administered once a week for a total of four doses.
  • Blood was collected from the fundus venous plexus of the mouse, and its serum antibody titer was examined by the enzyme immunoassay described in 4 (3). Three days after the final immunization, spleens were removed from mice with sufficiently high serum antibody titers.
  • the spleen was shredded in MEM (Minimum Essential Medium) medium (manufactured by Nissui Pharmaceutical Co., Ltd.), loosened with forceps, and centrifuged (OO rpm, 5 minutes). ! Resulting tris precipitate fraction - was added Anmoniumu buffer chloride solution (P H7.6),: erythrocytes were removed by treating 1-2 minutes. The resulting precipitate fraction (cell fraction) was washed three times with MEM medium and used for cell fusion.
  • MEM Minimum Essential Medium
  • Anmoniumu buffer chloride solution P H7.6
  • whole blood is collected at the time of spleen extraction, and the serum obtained after standing at room temperature for 2 hours is a peptide-specific antiserum, and the antiserum is specific for the protein of the present invention obtained in the above 1). It was an antiserum.
  • THY Thiroglobulin; thyroglobulin, Sigma
  • the above conjugate prepared at 5 ⁇ g / ml was dispensed at 50 / Z 1 / well onto an EIA plate (Grainer Co., Ltd.) and left at 4 ° C for adsorption. .
  • PBS containing l% (w / v) BSA (B-PBS) was added at 100 ⁇ l / well to block the remaining active groups, and the mixture was reacted at room temperature for 1 hour.
  • the B-PBS was discarded, and the antiserum of the immunized mouse, the supernatant of the hybridoma culture or the purified monoclonal antibody was dispensed at 50 ⁇ l / well and allowed to react for 2 hours.
  • T-PBS peroxidase-labeled ⁇ sagi anti-mouse immunoglobulin
  • P3X63Ag8U.l 8-azaguanine-resistant mouse myeloma cell line P3X63Ag8U.l (P3-U1: purchased from ATCC) was cultured in normal medium (RPI medium supplemented with 10% fetal calf serum), and 2 x 10 7 or more cells were fused at the time of cell fusion. And used for cell fusion as a parent strain.
  • mice spleen cells obtained in 4 (2) and the myeloma cells obtained in 4 (4) were mixed at a ratio of 10: 1, and centrifuged (1, 200 rpm, 5 minutes). Was loosened well resulting precipitated fraction of cell groups, while stirring, at 37 ° C, polyethylene glycol one 1000 (PEG- 1000) 2g, mixture 10 8 of MEM medium 2ml and dimethyl sulfoxide 0.7ml 0.5 ml was added per mouse splenocytes. Further, lml of MEM medium was added several times to the suspension every 1 to 2 minutes, and then MEM medium was added so that the total volume became 50ml.
  • PEG- 1000 polyethylene glycol one 1000
  • the cells in the precipitate fraction obtained are gently loosened by tapping, and 100 ml of HAT medium [10. % Hat fetal serum-supplemented RPMI medium supplemented with HAT Media Supplement (Boehringer Mannheim).
  • HAT medium 10. % Hat fetal serum-supplemented RPMI medium supplemented with HAT Media Supplement (Boehringer Mannheim).
  • the suspension was dispensed into a 96-well culture plate at a volume of 200 ml / well and cultured at 37 ° C. for 10 to 14 days in a 5% CO 2 incubator.
  • the culture supernatant was examined by the enzyme immunoassay described in 4 (3), and cells that reacted with the antigen peptide but did not react with the control peptide were selected. Further, the cells contained in the well were repeatedly cloned twice by the limiting dilution method to obtain a monoclonal antibody-producing hybridoma KM2715 recognizing a partial sequence of the protein of the present invention.
  • the monoclonal The results of analyzing the reactivity of the antibody KM2715 with the antigen peptide and the control peptide by an enzyme immunoassay are shown in FIG.
  • the monoclonal antibody KM2715 showed specific reactivity to an antigen peptide consisting of a partial sequence of the protein of the present invention.
  • Monoclonal antibodies were purified from mouse ascites.
  • An 8-week-old nude female mouse (BALB / c) treated with pristane was intraperitoneally injected with 5 to 20 ⁇ 10 6 cells / animal of the hybridoma and the hybridoma strain obtained in 4 (5). 10 to 21 days later, ascites was collected from mice in which the ascitic carcinoma of the hybridoma was changed (1 to 8 mlZ mice). After the ascites was centrifuged (1, 200 ⁇ g, 5 minutes) to remove solids, the monoclonal antibody was purified by the caprinoleic acid precipitation method [Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988]. The subclass of the monoclonal antibody was determined to be IgGl by ELISA using a subcluster typing kit.
  • the monoclonal antibody KM2715 specifically reacted with a band around 51 kDa corresponding to the molecular weight of the protein in CE.
  • KM511 Japanese Unexamined Patent Publication No. 8-165300
  • KM511 Japanese Unexamined Patent Publication No. 8-165300
  • the monoclonal antibody can specifically detect and quantify the protein of the present invention in pollen allergen extract, confirming its usefulness.
  • allergen immunotherapy a vaccine for the treatment of allergic diseases
  • Allergy 696 (1998)
  • the protein has extremely high antigenicity and can be said to be a very important causative substance in the onset of cedar pollinosis. Therefore, quantification of the allergen extract using the monoclonal antibody, standardization of the allergen extract, and a method thereof are considered to be extremely useful in promoting efficient desensitization therapy.
  • the protein of the present invention has the property of binding to IgE in the serum of hay fever patients, it is useful not only as an antigen in immunochemical diagnostic methods, but also has the property of inducing allergic disease, and It is also useful as a desensitizing agent for diagnosing, treating, or preventing.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Botany (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A novel protein having an allergen activity in pollinosis; peptide fragments of this protein or derivatives thereof; diagnostics, remedies and preventives for pollen allergy containing as the active ingredient the above protein, its peptide fragments or derivatives thereof; a process for producing the above protein, its peptide fragments or derivatives thereof; an antibody against the above protein, its peptide fragments or derivatives thereof; and diagnostics and remedies containing this antibody as the active ingredient.

Description

明 細 書  Specification
新規花粉抗原 技術分野  New pollen antigen technology
本発明は、アレルギー疾患におけるアレルゲン活性を有する新規なタンパク質、該タ ンパク質の部分ペプチドまたはそれらの誘導体、該タンパク質、該タンパク質の部分ぺ プチドまたはそれらの誘導体を有効成分として含有するアレルギー疾患に対する診断 薬、治療薬、および予防薬、該タンパク質、その部分ペプチドまたはそれらの誘導体を 製造するための方法、該タンパク質、部分ペプチドまたはそれらの誘導体に対する抗 体または抗体断片ならびに該抗体またはその抗体断片を有効成分として含有する診 断薬、治療薬および予防薬に関する。  The present invention relates to a diagnosis of an allergic disease containing a novel protein having allergen activity in an allergic disease, a partial peptide of the protein or a derivative thereof, the protein, a partial peptide of the protein or a derivative thereof as an active ingredient. A method for producing a drug, a therapeutic agent, a prophylactic agent, the protein, a partial peptide or a derivative thereof, an antibody or an antibody fragment against the protein, the partial peptide or a derivative thereof, and the antibody or the antibody fragment thereof. It relates to diagnostic, therapeutic and prophylactic agents contained as components.
背景技術 Background art
花粉症は日本にぉレ、て最も重大なアレルギー性疾患のひとつである。アレルギーを 惹起する花粉にはスギ、ヒノキ、シラカバ、カモガヤ、ホソムギ、イネ、ブタクサなど多数 あるが、特にスギ花粉によるアレルギーは花粉症の約 8割を占めるといわれている。ス ギ花粉症患者は 1970年代以降急激に増加し、国民の 10%以上を占める。さらに、 自動 車排気ガス中の NOxなどによる大気汚染などもスギ花粉症の要因とされ、スギ花粉症 患者は依然増加傾向にある。また、スギ花粉の飛散時期には、 日々スギ花粉の飛散 量などの情報がマスコミに取り上げられるなど、公衆衛生上もはやスギ花粉症は放置 できない問題になっている。  Hay fever is one of the most serious allergic diseases in Japan. There are many types of pollen that cause allergies, such as cedar, cypress, birch, duckweed, oats, rice and ragweed. In particular, it is said that allergy caused by cedar pollen accounts for about 80% of hay fever. Cedar hay fever cases have increased sharply since the 1970s, accounting for more than 10% of the population. In addition, air pollution caused by NOx in vehicle exhaust gas is also considered to be a cause of cedar pollinosis, and the number of cedar pollinosis patients is still increasing. At the time of cedar pollen scattering, information such as the amount of cedar pollen scattered is taken up in the media every day, and there is a problem that cedar pollinosis can no longer be neglected in public health.
花粉症は、花粉中の抗原性物質である花粉アレルゲンが主因となって引き起こされ るアレルギー性疾患である。アレルギーとは、ある特定の抗原に感作された生体が再 び同一の抗原と接触したときに、局所的あるいは全身的に起こす組織障害をいう。ァ レルギ一反応は、 Coombsと Gellによると I〜IV型に分類される力 花粉症は、花粉ァレ ルゲンに特異的に反応する IgEによって仲介される I型アレルギーに分類される。その 病態は、鼻炎、結膜炎を主たる症状とする即時型アレルギー性疾患が主であり、水様 性鼻汁、くしゃみ、痒み、流涙、咳、ときに全身性の倦怠感などをもたらす (アトピー'ァ レルギ一性疾患;監修 山村雄一、吉利和、中山書店、 1992、アレルギー性疾患;編 集 宫本昭正、中川武正、医師薬出版株式会社、 1985など)。  Pollinosis is an allergic disease caused mainly by pollen allergen, an antigenic substance in pollen. Allergy is a tissue disorder that occurs locally or systemically when a living body sensitized to a specific antigen comes in contact with the same antigen again. Allergic reactions are classified as type I-IV according to Coombs and Gell Pollinosis is classified as a type I allergy mediated by IgE, which responds specifically to pollen allergens. Its pathological condition is mainly immediate allergic disease with rhinitis and conjunctivitis as the main symptoms, and it causes watery nasal discharge, sneezing, itching, lacrimation, coughing, and sometimes general malaise (atopy Allergy diseases: supervision, supervised by Yuichi Yamamura, Yoshikazu Giri, Nakayama Shoten, 1992, Allergic diseases; edited by Akimasa Motomoto, Takemasa Nakagawa, Yakuhin Yaku Shuppan Co., Ltd., 1985).
すなわち、鼻腔、目艮、気管の粘膜に接触した花粉粒子から溶出した花粉アレルゲン はマクロファージ等の抗原提示細胞に取込まれる。その後、抗原を取込んだ抗原提示 細胞カ^ンパ組織に到達すると、リンパ組織で、花粉アレルゲンに対して特異的な免 疫応答が誘導され、アレルゲンに特異的に反応する IgEが産生される。該 IgEは血流中 で肥満細胞や好塩基球上の IgE受容体と結合する。このときに、花粉アレルゲンが再 び生体内に侵入すると、肥満細胞および好塩基球上の花粉アレルゲンに特異的な Ig Eと侵入した花粉アレルゲンとが結合する。該肥満細胞および該好塩基球は活性化さ れ、細胞内からヒスタミン、セロトニン、アナフィラキシー性好酸球遊走因子、好中球遊 走因子、プロスタグランジン、ペプチドロイコトリェンなどのケミカルメディエーターが周 囲の組織に放出されると、標的細胞を刺激して I型アレルギーの病態が形成される。例 えば、ケミカルメディエーター力ヒスタミンの場合、ヒスタミンは知覚神経終末の HI受容 体を介して、知覚終末よりくしゃみ中枢に作用してくしゃみを起こす。またそのほかに、 平滑筋収縮、血管透過性亢進、浮腫、粘膜分泌亢進なども引き起こし、即時反応の症 状をもたらしたりする (アトピー'アレルギー性疾患;監修 山村雄一、吉利和、中山書 店、 1992など)。 That is, the pollen allergen eluted from the pollen particles in contact with the mucous membrane of the nasal cavity, mesotrum, and trachea is taken up by antigen presenting cells such as macrophages. Thereafter, upon reaching the antigen-presenting cell campa tissue incorporating the antigen, a specific immunological response to pollen allergen is induced in the lymph tissue, and IgE that specifically reacts with the allergen is produced. The IgE is in the bloodstream Binds to IgE receptors on mast cells and basophils. At this time, when the pollen allergen reenters the living body, IgE specific to the pollen allergen on mast cells and basophils binds to the invading pollen allergen. The mast cells and the basophils are activated, and chemical mediators such as histamine, serotonin, anaphylactic eosinophil chemotactic factor, neutrophil chemotactic factor, prostaglandin, and peptide leukotriene are circulated from within the cell. When released into surrounding tissue, it stimulates target cells to form a type I allergic condition. For example, in the case of the chemical mediator histamine, histamine acts on the sneezing center rather than the sensory terminal via the HI receptor at the sensory nerve terminal, causing sneezing. In addition, it also causes smooth muscle contraction, increased vascular permeability, edema, increased secretion of mucous membranes, etc., leading to symptoms of immediate response (Atopy's allergic disease; supervised by Yuichi Yamamura, Yoshikazu Kirishi, Nakayama Shoten, 1992) Such).
現在、花粉症の治療は、主として抗アレルギー剤、抗炎症剤などを投与する対症療 法であり、花粉より抽出した抗原 (アレルゲンエキス)を減感作剤として少量ずつ投与 する減感作療法が一部併用して行われている。  Currently, the treatment of hay fever is a symptomatic treatment that mainly administers antiallergic agents, anti-inflammatory agents, etc., and desensitization therapy in which an antigen (allergen extract) extracted from pollen is administered little by little as a desensitizing agent. Partially used together.
ケミカルメディエーター遊離抑制を主作用とする抗アレルギー剤、 HI受容体でのヒス タミンとの拮抗を主作用とする抗ヒスタミン剤、炎症性メディエーターの産生抑制を主 作用とするステロイド剤など、抗アレルギー剤あるいは抗炎症剤は、上述した花粉症の 発症機構からもあくまでも対症療法で用いているので、花粉症によるアレルギー症状 を一時的に緩和するが、花粉症を根治することはできな 、。  Anti-allergic agents or anti-allergic agents, such as anti-allergic agents mainly acting to suppress release of chemical mediators, anti-histamine agents mainly acting to antagonize histamine at HI receptors, and steroid agents mainly acting to suppress the production of inflammatory mediators Inflammatory agents are used for symptomatic treatment to the last of the above-mentioned mechanism of hay fever, so that allergic symptoms caused by hay fever can be temporarily alleviated, but hay fever cannot be cured.
減感作療法の正確な機序は不明だが、該療法はアレルゲンエキスの投与によりァレ ルゲンに特異的な IgGや IgAの産生を促進することが関係するとレ、われており(免疫生 物学;監訳 笹月健彦、南江堂、 1995)、根治療法として期待されている。また、減感 作療法に用いられるアレルゲンエキスは診断にも使用されており、花粉症の治療、診 断の双方に不可欠なものとなってレ、る。  The exact mechanism of desensitization therapy is unknown, but it has been suggested that the therapy involves the production of allergen-specific IgG and IgA by administration of allergen extract (Immunological Biology). ; Translation by Takehiko Sasazuki, Nankodo, 1995), which is expected as a root treatment. In addition, allergen extracts used in desensitization therapy are also used for diagnosis and are indispensable for both treatment and diagnosis of hay fever.
しかしながら、現在使用されているアレルゲンエキスは、花粉を水性溶媒中で抽出 し、適宜希釈した粗抽出抗原であるため、同一の抽出法を用いたとしても原料の花粉 により組成が大幅に変動し易く、化学的に極めて不均一となり一定の減感作効果が得 られにくいなどの問題点がある。そのため、減感作効果が現われるまで長期間にわた る抗原投与が必要になることがあり、患者に大きな負担を与えるといった問題点をも有 する。さらに、現在使用されているアレルゲンエキスには多くの不純物が含まれること も、治療、診断の有効性、信頼性において問題である。  However, the currently used allergen extract is a crudely extracted antigen obtained by extracting pollen in an aqueous solvent and appropriately diluting it.Therefore, even if the same extraction method is used, the composition tends to fluctuate greatly depending on the pollen of the raw material. However, there is a problem that it is extremely non-uniform chemically and it is difficult to obtain a certain desensitizing effect. For this reason, antigen administration for a long period of time may be necessary until the desensitizing effect appears, which also has a problem that a heavy burden is imposed on the patient. Furthermore, the fact that allergen extracts currently used contain many impurities also poses a problem in the effectiveness and reliability of treatment and diagnosis.
そこで、花粉症の原因となるアレルゲン性タンパク質の分離、精製、同定が試みられ ており、これまで、スギ花粉由来のアレルゲン性タンパク質についても、以下のように報 告されている。 Therefore, attempts have been made to separate, purify, and identify allergenic proteins that cause hay fever. So far, allergenic proteins derived from cedar pollen have been reported as follows. It has been tell.
Yasuedaらは、分子量 41,000〜46,000Daのスギ花粉アレルゲンを単離精製し Sugi Ba sic Protein (SBP)と 名した (Journal of Allergy and Clinical Immunology, Vol.71 , 77-86, 1983)。現在、 WHOの命名法に従レ、 Cry j 1 と呼称されている。その後、 Tani aiらにより Cry j 1 の N末端の 20アミノ酸残基の配列が決定された (FEBS Letters, Vo 1.239, 329-332, 1988)。さらに、最近、 Cry j 1 の cDNAがクローニングされ、全塩基 配列が報告された (WO93/01213, Allergenic Proteins And Peptides Of Japanese Cedar Pollen), (特開平6- 197768、スギ花粉抗原およびそれをコードする遺伝子)。 また、 Sakaguchiらは、 Cry j 1 とは異なる、 Cry j 2と命名される分子量 37,000Daの スギ花粉アレルゲンを報告した (Allergy, Vol.45, 309-312, 1990)。最近、 Cry j 2の 全塩基配列も報告された (W〇94/1 1512、杉花粉由来のアレルゲン性タンパク質及び ヘプテ卜ノ、 (Biochemical and Biophysical Research Communications, Vol.201 , 1021 -1028, 1994)、 (Namba, M., et al., FEBS Letters, Vol.353, 124-128, 1994)。 Yasueda et al. Name cedar pollen allergen molecular weight 41,000~46,000Da the isolated and purified Sugi Ba sic Protein (SBP) ( Journal of Allergy and Clinical Immunology, Vol.71, 77-86, 1 983) . Currently, it is called Cry j 1 according to the WHO nomenclature. Subsequently, the sequence of the N-terminal 20 amino acid residues of Cry j 1 was determined by Tani ai et al. (FEBS Letters, Vo 1.239, 329-332, 1988). Furthermore, recently, a cDNA cloning of Cry j 1, the entire nucleotide sequence has been reported (WO93 / 01213, Allergenic Proteins And Peptides Of Japanese Cedar Pollen), ( JP-A 6 - 1 9 77 6 8, cedar pollen antigen and The gene that encodes it). Sakaguchi et al. Also reported a cedar pollen allergen with a molecular weight of 37,000 Da, which is different from Cry j 1 and is named Cry j 2 (Allergy, Vol. 45, 309-312, 1990). Recently, the entire nucleotide sequence of Cry j 2 was also reported (W〇94 / 1 1512, allergenic protein and heptenono from cedar pollen, (Biochemical and Biophysical Research Communications, Vol. 201, 1021-1028, 1994). (Namba, M., et al., FEBS Letters, Vol. 353, 124-128, 1994).
しかし、 Sakaguchiらの報告 (Allergy, Vol.45, 309-312, 1990)によると、 Cry j 1 と y j 2は免疫学的な交差反応を示さないこと、また、スギ花粉症患者の多くは Cry j 1 および Cry j 2に対する特異的な IgEを有する力';、一部のスギ花粉症患者では Cry j 1 または Cry j 2の一方にしか反応しないことが明らかにされている。  However, according to a report by Sakaguchi et al. (Allergy, Vol. 45, 309-312, 1990), Cry j 1 and yj 2 do not show immunological cross-reactivity, and many cedar pollinosis patients The ability to have specific IgE for j1 and Cry j2 '; it has been shown that some cedar hay fever patients only respond to one of Cry j1 or Cry j2.
さらに、最近、これらとは異なるスギ花粉アレルゲンも報告されている。特開平 7-1882 89号公報には分子量 55,000Da、 pI9.0のポリペプチドが報告されており、また、特開平 7 - 188291号公報には分子量 44,000〜54,000Da、 ρΙδ.5〜9.2の糖タンパク質が報告され ている。  Furthermore, recently, a cedar pollen allergen different from these has been reported. Japanese Patent Application Laid-Open No. 7-188289 reports a polypeptide having a molecular weight of 55,000 Da and a pI of 9.0, and Japanese Patent Application Laid-Open No. 7-188291 discloses a polypeptide having a molecular weight of 44,000 to 54,000 Da and ρΙδ. Glycoproteins have been reported.
このように、レ、くつかのスギ花粉アレルゲンが単離され、その性状や性質が解析され たことから、治療、予防または診断を目的として、精製スギ花粉アレルゲンを治療薬、 予防薬または診断薬としてヒトに投与することが試みられている。 Taniguchiら (Int. Arc h. Allergy Appl. Immunol., Vol.89, 136-142, 1989)は、マウスを用いた実験で、プ ルランを結合した SBP (Cry j 1)は特異的 IgEの産生を抑制することを報告し、減感作 剤としての有用性を示唆した。  In this way, several Japanese cedar pollen allergens were isolated and their properties and properties were analyzed, and purified cedar pollen allergens were used as therapeutic, prophylactic or diagnostic agents for the purpose of treatment, prevention or diagnosis. Has been attempted to be administered to humans. Taniguchi et al. (Int. Arc h. Allergy Appl. Immunol., Vol. 89, 136-142, 1989) reported that in a mouse experiment, pullulan-bound SBP (Cry j 1) produced specific IgE. And reported that it was useful as a desensitizer.
しかし、前述したように一部の患者では Cry j Cry j 2のいずれかにしか反応しな いなど、全スギ花粉症患者を対象とした場合、スギ花粉症の正確な診断あるいは有効 な治療にはこれまで報告されたアレルゲンのみでは未だ不十分である。  However, as described above, if all cedar pollinosis patients are targeted, such as some patients responding to only one of Cry j and Cry j 2, accurate diagnosis or effective treatment of cedar hay fever can be achieved. Is still insufficient with allergens reported so far.
さらに、最近、減感作療法におけるアナフィラキシー反応による副作用を回避するこ とを目的に、遺伝子操作により IgE反応性を減弱化した改変体(Takai, Τ·, et al., Nat ure Biotechnology, Vol.15, 754-758, 1997)、 T細胞が認識するェピトープを含むぺ プチド断片(WO94/01560、 W094/11512,特開平 7- 118295、特開平 8- 47392)の減感 作剤への応用が試みられている。したがって、花粉症の原因となるアレルゲンの分子 レベルでの角军析も必要となっており、ァレノレゲンの同定はさらにその重要性を増してい る。 Furthermore, recently, in order to avoid side effects due to the anaphylactic reaction in desensitization therapy, a variant in which IgE reactivity has been attenuated by genetic manipulation (Takai, Τ ·, et al., Nature Biotechnology, Vol. 15, 754-758, 1997), including epitopes recognized by T cells. Attempts have been made to apply peptide fragments (WO94 / 01560, W094 / 11512, JP-A-7-118295, JP-A-8-47392) to desensitizers. Therefore, molecular analysis of allergens that cause pollinosis at the molecular level is also required, and the identification of allenogens is becoming even more important.
発明の開示 Disclosure of the invention
上述のことから、有効性、安全性、信頼性の点からも、減感作治療薬、予防薬、診断 薬として有用なアレルゲンの同定が期待されている。さらに、アレルギー疾患の迅速か つ正確な診断は、アレルギー疾患の有効な治療を行ううえで極めて重要であり、有用 な診断システムの確立、さらには治療方法が期待されてレ、る。  Based on the above, identification of allergens useful as therapeutic drugs for desensitization, prophylactics, and diagnostics is expected from the viewpoints of efficacy, safety and reliability. Furthermore, rapid and accurate diagnosis of allergic diseases is extremely important for effective treatment of allergic diseases, and the establishment of a useful diagnostic system and further treatment methods are expected.
本発明は、以下の(1)〜(32)に関する。  The present invention relates to the following (1) to (32).
(1) 下記の理化学的性質を有するタンパク質。  (1) A protein having the following physicochemical properties.
(i)分子量: 51, 000 ± 5, OOODa (還元条件下 SDS—ポリアクリルアミドゲル電気泳 動)  (i) Molecular weight: 51,000 ± 5, OOODa (SDS-polyacrylamide gel electrophoresis under reducing conditions)
(ii) N末端アミノ酸配列: N末端に Xaa— Leu一 Phe - Pro一 Lys一 Glu— Ala— Leu — Pro— Thrで表わされるアミノ酸配列を有する(ただし、 Xaaは、 Glu、 Asp、 His, Gl y、 Arg、 Trpまたは Cysのいずれかを示す)。  (ii) N-terminal amino acid sequence: has an amino acid sequence represented by Xaa—Leu—Phe—Pro—Lys—Glu—Ala—Leu—Pro—Thr at the N-terminus (where Xaa is Glu, Asp, His, Gl y, Arg, Trp or Cys).
(iii)紫外吸収スペクトル:溶媒が、水、生理的食塩液またはリン酸緩衝液のとき、 280η m付近に極大吸収を有する。  (iii) Ultraviolet absorption spectrum: when the solvent is water, physiological saline or phosphate buffer, it has a maximum absorption around 280ηm.
(iv)溶剤への溶解性:水、生理的食塩液およびリン酸緩衝液に可溶である。  (iv) Solubility in solvent: soluble in water, physiological saline and phosphate buffer.
(V)安定性:水溶液 (pH7. 3)中、 4°Cで 2か月間放置しても実質的に失活しない。 上記において、 Xaaは、好ましくは Gluを示す。  (V) Stability: Does not substantially deactivate even when left in an aqueous solution (pH 7.3) at 4 ° C for 2 months. In the above, Xaa preferably represents Glu.
(2) 花粉症患者の血清中 IgEとの結合活性を有し;かつアレルゲン活性を有する、 上記(1)記載のタンパク質。  (2) The protein according to the above (1), which has a binding activity to IgE in serum of a hay fever patient; and has an allergen activity.
(3) タンパク質; ^スギ花粉に由来する上記(1)または (2)記載のタンパク質。  (3) Protein; ^ The protein according to the above (1) or (2), which is derived from cedar pollen.
(4) 上記(1 )〜(3)のレ、ずれか 1項に記載のタンパク質の部分ペプチド。  (4) The partial peptide of the protein according to (1) or (3) above.
(5) 上記(1)〜(4)のレ、ずれか 1項に記載のタンパク質または該タンパク質の部分 ペプチドの誘導体。  (5) The protein or the partial peptide derivative of the protein according to (1) or (4).
(6) 花粉より溶媒で抽出した抽出物を、塩析、透析、濾過、濃縮、遠心分離、各種 クロマトグラフィー、電気泳動から選ばれる精製手段と、アレルゲン活性を指標とする 検出手段とを組み合わせることによりタンパク質を採取することを特徴とする上記(1) 〜(3)のいずれか 1項に記載のタンパク質を製造する方法。  (6) Combining a purification method selected from salt extraction, dialysis, filtration, concentration, centrifugation, various types of chromatography, and electrophoresis of an extract extracted from pollen with a solvent, and a detection method using allergen activity as an index The method for producing a protein according to any one of the above (1) to (3), wherein the protein is collected by:
(7) 上記(1)〜(3)のレ、ずれか 1項に記載のタンパク質を酵素もしくは化学分解ま たは合成することにより、上記(4)または(5)に記載の部分ペプチドまたは該部分ぺプ チド誘導体を製造する方法。 (7) The partial peptide according to (4) or (5) or the protein according to (4) or (5) above, by enzymatically or chemically decomposing or synthesizing the protein according to (1) to (3). Partial mapping A method for producing a tide derivative.
(8) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を有効成分として含有するアレルギー疾患診断薬。  (8) A diagnostic agent for an allergic disease comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof as an active ingredient.
(9) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を有効成分として含有するアレルギー疾患治療薬。  (9) A therapeutic drug for an allergic disease comprising, as an active ingredient, the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
(10) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を有効成分として含有するアレルギー疾患予防薬。  (10) An agent for preventing an allergic disease, comprising as an active ingredient the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
(11) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を有効成分として含有するアレルギー疾患減感作剤。  (11) An allergic disease desensitizing agent comprising, as an active ingredient, the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
(12) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体に特異的に結合する IgEの定量方法。  (12) A method for quantifying IgE that specifically binds to the protein according to any one of the above (1) to (5), a partial peptide of the protein, or a derivative thereof.
(13) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体に特異的に結合する IgEの検出方法。  (13) A method for detecting IgE that specifically binds to the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
(14) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を用いる、アレルギ一疾患の診断方法。  (14) A method for diagnosing an allergic disease using the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof.
(15) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を経口または非経口で反復投与することを特徴とする減感 作療法。  (15) Desensitization therapy characterized by repeatedly orally or parenterally administering the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof. .
(16) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分 として含有するアレルギー疾患診断薬。  (16) An agent for diagnosing an allergic disease, comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
(17) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分 として含有するアレルギー疾患治療薬。  (17) A therapeutic drug for an allergic disease, comprising as an active ingredient the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
(18) 上記(1)〜(5)のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分 として含有するアレルギー疾患予防薬。  (18) A prophylactic agent for an allergic disease comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient .
(19) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分 として含有するアレルギー疾患減感作剤。  (19) Allergy disease desensitization comprising the protein according to any one of the above (1) to (5), a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient. Formulation.
(20) 上記(1)〜 )のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を含む組成 物を用いる、アレルギー疾患の診断方法。 (21) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を含む組成 物を経口あるいは非経口で反復投与することを特徴とする減感作療法。 (20) A method for diagnosing an allergic disease using the protein according to any one of the above (1) to (1), a partial peptide of the protein or a derivative thereof, and a composition containing a known allergen or a derivative thereof. . (21) The protein according to any one of (1) to (5) above, a partial peptide of the protein or a derivative thereof, and a composition containing a known allergen or a derivative thereof are orally or parenterally administered. Desensitization therapy characterized by repeated administration.
(22) 上記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプ チドまたはそれらの誘導体を認識する抗体。  (22) An antibody that recognizes the protein according to any one of (1) to (5) above, a partial peptide of the protein, or a derivative thereof.
(23) 上記(1)〜(5)のいずれか 1項に記載のタンパク質の N末端アミノ酸配列に特 異的に反応する、上記 (22)に記載の抗体。  (23) The antibody according to (22), which specifically reacts with an N-terminal amino acid sequence of the protein according to any one of (1) to (5).
(24) 抗体が、配列番号 1記載のアミノ酸配列を認識する上記 (22)記載の抗体。  (24) The antibody according to (22), wherein the antibody recognizes the amino acid sequence of SEQ ID NO: 1.
(25) 抗体が、モノクローナル抗体である上記(22)〜(24)に記載の抗体。  (25) The antibody according to the above (22) to (24), wherein the antibody is a monoclonal antibody.
(26) モノクローナル抗体力 KM2715である上記(25)記載の抗体。  (26) The antibody according to the above (25), wherein the antibody has a monoclonal antibody activity of KM2715.
(27) 上記 (22)〜(26)のレ、ずれか 1項に記載の抗体を有効成分として含有するァ レルギ一疾患診断薬。  (27) A diagnostic agent for an allergic disease comprising the antibody according to any one of (22) to (26) above as an active ingredient.
(28) 上記 (22)〜(26)のレ、ずれか 1項に記載の抗体を有効成分として含有するァ レルギ一疾患治療薬。  (28) A remedy for allergic disease comprising the antibody according to (1) or (22) to (26) as an active ingredient.
(29) 上記 (22)〜(26)のレ、ずれ力 1項に記載の抗体を有効成分として含有するァ レルギ一疾患予防薬。  (29) A prophylactic agent for allergic diseases, which comprises the antibody described in the item (1) of (22) to (26) above as an active ingredient.
(30) 上記 (22)〜(26)のいずれか 1項に記載の抗体を用いて、花粉抗原を定量 する方法。  (30) A method for quantifying a pollen antigen using the antibody according to any one of (22) to (26).
(31) 上記(22)〜(26)のいずれか 1項に記載の抗体を用いて、花粉抗原中の上 記(1)〜(5)のいずれ力 1項に記載のタンパク質、該タンパク質の部分ペプチドまたは それらの誘導体を定量する方法。  (31) The protein according to any one of (1) to (5) above in a pollen antigen, using the antibody according to any one of (22) to (26) above, A method for quantifying a partial peptide or a derivative thereof.
(32) 上記 (22)〜(26)のいずれか 1項に記載の抗体または抗体断片を用いる、ァ レルギ一疾患の診断方法。 本発明者らは、未知のスギ花粉由来のタンパク質がアレルゲン活性を有し、既知の スギ花粉由来タンパク質とは異なる性状および性質を有することを見出した。また、既 知のスギ花粉由来タンパク質と比較して、本発明のタンパク質がスギ花粉症患者の血 清中 IgEと強い結合活性を示すこと、さらに、本発明の精製タンパク質に対する血清中 I gEの抗体価が、既存の体外診断で用いたスギ花粉由来アレルゲンエキスに対する IgE 抗体価と高い相関性を示すことを明らかにした。以上のことより、本発明のタンパク質ま たは該タンパク質の部分ペプチドを用いて、アレルギー疾患の診断、治療および予防 を行うことができる。 本発明のアレルギー疾患とは、単一または複数の同種アレルゲンにより引き起こされ るアレルギー性疾患、単一または複数の異種のアレルゲンにより引き起こされるアレル ギー性疾患など、すべてのアレルギー疾患を包含する。 (32) A method for diagnosing an allergic disease using the antibody or antibody fragment according to any one of the above (22) to (26). The present inventors have found that an unknown cedar pollen-derived protein has allergen activity and has properties and properties different from those of known cedar pollen-derived proteins. Further, compared to known cedar pollen-derived proteins, the protein of the present invention shows a stronger binding activity to serum IgE of cedar pollinosis patients, and furthermore, serum IgE antibodies against the purified protein of the present invention. The titer showed a high correlation with the IgE antibody titer against cedar pollen-derived allergen extract used in the existing in vitro diagnosis. As described above, diagnosis, treatment and prevention of allergic diseases can be performed using the protein of the present invention or the partial peptide of the protein. The allergic disease of the present invention includes all allergic diseases such as an allergic disease caused by one or more homologous allergens and an allergic disease caused by one or more different allergens.
アレルゲンとしては、花粉、ハウスダスト、ダニ虫体などの昆虫、食物などのアレルギ Allergens include insects such as pollen, house dust and mite, and allergens such as food.
—誘因物およびアレルギー誘因物からの粗抽出物であるアレルゲンエキス、該アレル ゲンエキスより精製した主要アレルゲンなどがあげられる。 -Allergen extracts, which are crude extracts from allergens and allergens, and major allergens purified from the allergen extract.
主要アレルゲンとしては、スギ花粉に由来する Cry j 1、 Cry j 2、ヒノキ花粉に由来 する Cha 0 KSuzeki, Μ·, et al. Molecular Immunology, Vol.33, 451, 1996)、カノ ノキ花粉に由来する Bet v Bet v 2、ブタクサ花粉に由来する Amb a 1、 Amb a 2、ダニ虫体に由来する Der f 1、 Der f 2、 Der pl、 Derp2、ネコ毛に由来する Fel d 1 などがあげられる。  Major allergens include Cry j 1 and Cry j 2 derived from cedar pollen, Cha 0 KSuzeki derived from hinoki pollen, et al. Molecular Immunology, Vol. 33, 451, 1996), derived from cypress pollen Bet v Bet v 2, Amb a 1, Amb a 2, derived from ragweed pollen, Der f 1, Der f 2, Der pl, Derp 2, derived from mite bodies, Fel d 1, derived from cat hair, etc. Can be
花粉アレルゲンの引き起こすアレルギー疾患(花粉症)としては、複数種の花粉によ り引き起こされるアレルギー疾患、花粉に加えて花粉以外のアレルゲンにより引き起こ されるアレルギー疾患があげられる。  Allergic diseases caused by pollen allergens (hay fever) include allergic diseases caused by multiple types of pollen, and allergic diseases caused by allergens other than pollen in addition to pollen.
具体的には、複数種の花粉により引き起こされるアレルギー疾患として、スギ花粉症 患者力 Sヒノキ花粉に対してもアレルギー反応を示す例 (花粉症の科学、斎藤洋三 '井 手武、科学同人、 1994)などがあげられ、花粉に加えて花粉以外のアレルゲンにより引 き起こされるアレルギー疾患として、スギ花粉症患者がダニなどの花粉以外の類縁性 のないアレルゲンに対してアレルギー反応を示す例などがあげられる。  Specifically, as an allergic disease caused by multiple types of pollen, Japanese cedar pollinosis patients have an allergic reaction to S-cypress pollen (Science of hay fever, Yozo Saito 'Tidetake Ide, Doujin Kagaku, 1994 An example of an allergic disease caused by allergens other than pollen in addition to pollen is an example in which a cedar pollinosis patient shows an allergic reaction to unrelated allergens other than pollen such as mites. Can be
アレルギー疾患の主な症状としては、アレルギー性鼻炎、アレルギー性結膜炎、ァレ ルギー性皮膚炎、気管支喘息などがあげられる。  The main symptoms of allergic diseases include allergic rhinitis, allergic conjunctivitis, allergic dermatitis and bronchial asthma.
以下に、本発明について詳細に述べる。  Hereinafter, the present invention will be described in detail.
1 .新規花粉抗原の製造方法  1. Method for producing a new pollen antigen
( 1 ) タンパク質の製造方法  (1) Protein production method
本発明のタンパク質は、花粉、あるいは花粉を形成 *保持している雄花あるいは花芽 より単離、精製して取得する。花粉は、本発明のタンパク質を単離、精製可能であれ ばいずれも用いられる。例えば、アレルギ一を惹起するスギ花粉があげられ、好ましく はスギ科スギ属に属する日本スギ(クリプトメリア-ジャポニカ)の花粉があげられる。ま た、スギ花粉以外のアレルギーを惹起する花粉からも本発明のタンパク質に相同なタ ンパク質を単離することができ、例えば、ヒノキ、シラカバ、カモガヤ、ホソムギ、イネ、ブ タクサ等の花粉があげられる。  The protein of the present invention is obtained by isolating and purifying from a male flower or a flower bud that forms pollen or pollen. Any pollen can be used as long as the protein of the present invention can be isolated and purified. For example, Japanese cedar (Cryptomeria japonica) pollen, which belongs to the cedar family of the cedar family, may be mentioned, for example. Proteins homologous to the protein of the present invention can also be isolated from pollen that causes allergies other than cedar pollen.For example, pollen such as hinoki, birch, duckweed, ryegrass, rice, and grasshopper can be isolated. can give.
以下に、花粉由来のタンパク質の製造方法について述べる。方法は、特に断りのな い限り、 [ジャーナル'ォブ 'アレルギー 'アンド'クリニカル 'ィムノロジー(Journal of All ergy and Clinical Immunology), Vol.71 , 77- 86, 1983]に準じて行っている。 The method for producing pollen-derived protein is described below. Unless otherwise noted, the method is described in [Journal of Allergy] and [Clinical] Immunology. ergy and Clinical Immunology), Vol.71, 77-86, 1983].
本発明のタンパク質は、上述の花粉を溶媒で抽出し、得られる抽出液を精製すること により製造することができる。  The protein of the present invention can be produced by extracting the above-mentioned pollen with a solvent and purifying the resulting extract.
花粉から該タンパク質を抽出するには、雄花より採取した花粉を溶媒中に浮遊 ·懸濁 させ、必要に応じて攪拌させながら、浸漬させる。  To extract the protein from pollen, pollen collected from a male flower is suspended and suspended in a solvent, and immersed while stirring as necessary.
浸漬は、室温以下、好ましくは 4°Cで、 1時間以上、好ましくは 8時間から 12時間行う。 抽出溶媒としては、タンパク質化学において当業者が一般的に用いる緩衝液または 水などがあげられる。緩衝液としては、リン酸緩衝液、クェン酸緩衝液、トリス塩酸緩衝 液、酢酸緩衝液などがあげられる。  The immersion is performed at room temperature or lower, preferably at 4 ° C., for 1 hour or longer, preferably for 8 to 12 hours. Extraction solvents include buffers or water commonly used by those skilled in the art of protein chemistry. Examples of the buffer include a phosphate buffer, a citrate buffer, a Tris-HCl buffer, an acetate buffer and the like.
上記溶媒に、タンパク質の抽出効率を高める目的で適宜の塩、親水性有機溶媒を 加えてもよい。塩としては、塩化ナトリウム、塩化カリウムなどがあげられる。親水性有機 溶媒としては、メタノール、エタノール、ブタノール、アセトンなどがあげられる。  An appropriate salt or hydrophilic organic solvent may be added to the above-mentioned solvent for the purpose of increasing the protein extraction efficiency. Examples of the salt include sodium chloride and potassium chloride. Examples of the hydrophilic organic solvent include methanol, ethanol, butanol, acetone and the like.
ついで上記で得られた懸濁液を遠心分離し、上清を回収する。さらに、残渣に対し て上述の浸漬および遠心分離による抽出操作を繰り返し行うことにより、タンパク質の 回収量を上昇させることができる。通常、この抽出操作を 1〜5回繰り返すことにより大 部分のタンパク質を抽出することができる。  Then, the suspension obtained above is centrifuged, and the supernatant is collected. Furthermore, the amount of protein recovered can be increased by repeatedly performing the above-described immersion and centrifugation extraction operations on the residue. Usually, most of the protein can be extracted by repeating this extraction operation 1 to 5 times.
上述したタンパク質抽出液より、タンパク質の分離精製に通常用いられる精製手段 [蛋白質'酵素の基礎実験法 改訂第 2版 堀尾武ー編(南江堂) 1994]と、アレルゲ ン活性を指標とする検出手段とを組み合わせることにより本発明の部分精製されたタ ンパク質を得ることができる。  Purification methods usually used for protein separation and purification from the protein extract described above [Basic Experimental Method for Protein 'Enzyme Rev. 2nd Edition, Takeo Horio (Nan-Edo) 1994] and a detection method using allergen activity as an index By combining these, the partially purified protein of the present invention can be obtained.
タンパク質の分離精製に通常用いられる精製手段としては、塩析、透析、濾過、濃 縮、遠心分離、各種クロマトグラフィー、電気泳動などがあげられる。各種クロマトグラフ ィ一としては、イオン交換クロマトグラフィー、ゲルろ過クロマトグラフィー、疎水性クロマ トグラフィ一、ァフィ二ティ一クロマトグラフィー、逆相クロマトグラフィーなどがあげられ る。電気泳動としては、ポリアクリルアミドゲル電気泳動、等電点電気泳動などがあげら れる。  Purification means usually used for protein separation and purification include salting out, dialysis, filtration, concentration, centrifugation, various types of chromatography, and electrophoresis. Examples of various types of chromatography include ion exchange chromatography, gel filtration chromatography, hydrophobic chromatography, affinity chromatography, and reverse phase chromatography. Examples of the electrophoresis include polyacrylamide gel electrophoresis, isoelectric focusing, and the like.
上述で取得されたタンパク質のアレルゲン活性は、花粉症患者血清または血液細胞 に由来する特異的 IgEあるいは特異的リンパ球の存在とその反応性を指標に検出する こと力できる。  The allergen activity of the protein obtained above can be detected using the presence and reactivity of specific IgE or specific lymphocytes derived from serum or blood cells of hay fever patients as indicators.
特異的 IgEあるいは特異的 IgEを介した反応を検出する方法は、負荷試験法、試験 管内試験法があげられる。負荷試験法は、取得されたタンパク質を花粉症患者に負 荷してアレルギー反応を観察する試験方法である。試験管内試験法として、花粉症患 者の血清を用いる酵素免疫測定法や放射性免疫測定法、花粉症患者の白血球 (月巴 満細胞、好塩基球)力 のヒスタミン遊離を検出するヒスタミン遊離試験などがあげられ る。 Methods for detecting specific IgE or a reaction mediated by specific IgE include a stress test method and an in vitro test method. The stress test method is a test method in which the obtained protein is loaded on a hay fever patient and an allergic reaction is observed. In vitro test methods include enzyme immunoassay and radioimmunoassay using serum from hay fever patients, and leukocytes from hay fever patients. A histamine release test that detects histamine release by a full cell, basophil) force.
特異的リンパ球を検出する手段には、花粉症患者リンパ球を用いた増殖応答試験な どがあげられる。(アトピー 'アレルギー性疾患;監修 山村雄一、吉利和、中山書店、 1992)  Means for detecting specific lymphocytes include a proliferative response test using lymphocytes from hay fever patients. (Atopy's allergic disease; supervised by Yuichi Yamamura, Yoshikazu Giri, Nakayama Shoten, 1992)
酵素免疫測定法、放射性免疫測定法は、適当な担体に固相化した取得したタンパ ク質に対し、花粉症患者から採取した末梢血より調製した血清を反応させ、取得したタ ンパク質に対する血清中の特異的 IgEを検出する方法であり、 ELISA(enzyme-linked i mmunosorbent assay), RA f(radio-allergosorbent test)なとがあけられる。  In the enzyme immunoassay and the radioimmunoassay, serum obtained from peripheral blood collected from hay fever patients is reacted with the obtained protein immobilized on an appropriate carrier, and the serum for the obtained protein is used. This is a method for detecting specific IgE in the medium, such as ELISA (enzyme-linked immunosorbent assay) and RA f (radio-allergosorbent test).
ヒスタミン遊離試験は、取得したタンパク質に、花粉症患者の末梢血より調製した肥 満細胞、好塩基球などを含む白血球画分、あるいは、花粉症患者血清を反応させた 健常人の末梢血より調製した白血球画分を反応させて、取得したタンパク質に特異的 な IgEを介して肥満細胞あるいは好塩基球などを活性化することにより放出されるヒスタ ミンを検出する方法である。  The histamine release test is performed using the obtained protein and the leukocyte fraction containing fertilized cells and basophils prepared from the peripheral blood of a hay fever patient, or the peripheral blood of a healthy human subject to the serum of a hay fever patient. This is a method for detecting histamine released by reacting the obtained leukocyte fraction and activating mast cells or basophils via IgE specific to the obtained protein.
リンパ球増殖試験は、取得したタンパク質に、花粉症患者の末梢血より調製したリン パ球を反応させて、取得したタンパク質に対する T細胞あるレ、は B細胞の特異的な活 性化反応または増殖応答を検出する方法である。  In the lymphocyte proliferation test, the obtained protein is reacted with lymphocytes prepared from the peripheral blood of a hay fever patient, and the T cell or the B cell specific activation reaction or proliferation to the obtained protein is performed. This is a method of detecting a response.
通常、部分精製タンパク質は、 Cry j 1などの花粉抗原を含んでいるので、より高純 度のタンパク質を得るために、さらに、ゲルろ過クロマトグラフィー、イオン交換クロマト グラフィ一、ァフィ二ティ一クロマトグラフィ一、逆相クロマトグラフィー、疎水性クロマトグ ラフィ一、ポリアクリルアミドゲル電気泳動、等電点電気泳動などの方法を組み合わせ て適用し、本発明の精製タンパク質を得ることができる。  Normally, partially purified proteins contain pollen antigens such as Cry j 1. Therefore, in order to obtain a higher purity protein, gel filtration chromatography, ion exchange chromatography, affinity chromatography, etc. The purified protein of the present invention can be obtained by applying a combination of methods such as reverse phase chromatography, hydrophobic chromatography, polyacrylamide gel electrophoresis, and isoelectric focusing.
(2)部分ペプチドの調製方法  (2) Method for preparing partial peptide
本発明のタンパク質よりアレルゲン活性を有する部分ペプチドを得ることができる。 本発明の部分ペプチドは、上記 1 (1 )の方法により分離 ·精製したタンパク質より製造 することができる。すなわち、該タンパク質をタンパク質分解酵素または化学的手法に より限定分解し、得られた反応混合液より上述のタンパク質を精製する方法と同様の 方法を用いて部分ペプチドを得ることができる。タンパク質分解酵素としては、トリプシ ン、キモトリブシン、ペプシン、パパイン、 V8プロテアーゼなどがあげられる。化学的手 法としては、ぎ酸酸性条件下におけるプロモシアンによる分解反応などがあげられる。 あるいは、該タンパク質の部分アミノ酸配列を有するペプチドは、 Fmoc法(フルォレ ニルメチルォキシカルボニル法)、 tBoc法(t-ブチルォキシカルボニル法)等の化学合 成法によっても製造することができる。また、桑和貿易(米国 Advanced chemTech社 製)、パーキンエルマ一ジャパン(米国 Perkin- Elmer社製)、ァロカ(米国 Protein Tech nology Instrument社製)、クラボウ(米国 SyntheceU-Vega社製)、 日本パーセプティブ' リミテッド (米国 PerSeptive社製)、島津製作所等のペプチド合成機を用いても製造す ることがでさる。 A partial peptide having allergen activity can be obtained from the protein of the present invention. The partial peptide of the present invention can be produced from the protein separated and purified by the method (1). That is, the protein can be limitedly degraded by a proteolytic enzyme or a chemical technique, and a partial peptide can be obtained by a method similar to the method for purifying the protein from the obtained reaction mixture. Examples of proteolytic enzymes include trypsin, chymotrypsin, pepsin, papain, and V8 protease. Examples of the chemical method include a decomposition reaction with bromomethane under acidic conditions of formic acid. Alternatively, a peptide having a partial amino acid sequence of the protein can also be produced by a chemical synthesis method such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method). Kuwawa Trading (Advanced ChemTech, USA) Perkin-Elmer Japan (US Perkin-Elmer), Aroca (US Protein Technology Instrument), Kurabo (US SyntheceU-Vega), Nippon Perceptive 'Limited (US PerSeptive), Shimadzu Corporation It can also be produced using a peptide synthesizer such as
本発明のアレルゲン活性を有する部分ペプチドは、該タンパク質のアミノ酸配列の部 分配列を有する複数のペプチドより、タンパク質のアレルゲン活性の検出方法と同様 の方法によりスクリーニングして得ることができる。方法としては、上記 1 (1)の花粉症患 者の血清を用いた酵素免疫測定法、花粉症患者リンパ球を用いた増殖応答試験など があげられる。  The partial peptide having an allergen activity of the present invention can be obtained by screening a plurality of peptides having a partial sequence of the amino acid sequence of the protein by a method similar to the method for detecting the allergen activity of a protein. Examples of the method include the above-mentioned 1 (1) enzyme immunoassay using sera from hay fever patients and a proliferation response test using lymphocytes from hay fever patients.
(3)タンパク質またはその部分ペプチドの誘導体の製造方法  (3) Method for producing derivative of protein or partial peptide thereof
本発明は、本発明のタンパク質またはその部分ペプチドの誘導体を包含する。 誘導体とは、生体内の IgEとの反応性を消失あるいは減弱することを目的とした修飾 体および改変体をいう。誘導体は、既存のタンパク質化学的手法またはタンパク質ェ 学的手法 [生化学実験講座 1 タンパク質の化学 IV 日本生化学会編、新生化学実験 講座 1 タンパク質 IV 本生化学会編]などを単独あるいは組み合わせることにより、 取得することができる。例えば、本発明のタンパク質をジチオスレィトールなどの還元 剤存在下、ジスルフイド結合を開裂した後、生じるシスティン残基の SH基をョード酢酸 などによりブロックして高次構造を改変することなどがあげられる。  The present invention includes derivatives of the protein of the present invention or partial peptides thereof. Derivatives refer to modified forms and modified forms aimed at eliminating or attenuating the reactivity with IgE in a living body. Derivatives can be used alone or in combination with existing protein chemistry or protein chemistry techniques [Biochemistry Experiment Course 1 Protein Chemistry IV, edited by the Biochemical Society of Japan, Neogene Chemistry Experiment Course 1 Protein IV, Biochemistry Society], etc. Can be obtained. For example, after cleaving the disulfide bond of the protein of the present invention in the presence of a reducing agent such as dithiothreitol, the SH group of the resulting cysteine residue is blocked with odoacetic acid or the like to modify the higher-order structure. Can be
2.アレルギー疾患の診断薬、予防薬および治療薬  2.Diagnostic, preventive and therapeutic drugs for allergic diseases
本発明のタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体は、単独ま たは混合して、アレルギー疾患の診断薬、予防薬または治療薬として使用することが できる。  The protein of the present invention, a partial peptide of the protein and a derivative thereof can be used alone or in combination as a diagnostic, preventive or therapeutic agent for allergic diseases.
また、本発明のタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体の少 なくとも 1種と、上述した既知アレルゲンとを混合することにより、アレルギー疾患の診 断薬、予防薬または治療薬として使用することもできる。  Further, at least one of the protein of the present invention, a partial peptide of the protein and a derivative thereof is mixed with the above-mentioned known allergen to be used as a diagnostic, preventive or therapeutic drug for allergic diseases. You can also.
以下に、診断薬、予防薬および治療薬の調製方法を述べるが、本発明のタンパク 質、該タンパク質の部分ペプチドおよびそれらの誘導体のみならず既知アレルゲンに ついても、該方法を使用することにより調製することができる。  Hereinafter, methods for preparing diagnostic, prophylactic and therapeutic agents will be described. Not only the protein of the present invention, partial peptides of the protein and their derivatives but also known allergens can be prepared by using the method. can do.
本発明のタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体を有効成 分とする花粉アレルギー疾患の診断薬の調製方法には特に制限はないが、前述の方 法に準じて精製されたタンパク質および部分ペプチドおよびそれらの誘導体を濃縮し て液状として採取する力、あるいはさらに凍結乾燥して固状として採取して、単独ある いは適宜の配合剤として、調製し、使用することができる。 後述する診断方法のひとつである、負荷試験に使用する診断薬としては単独の形態 はもとより、生理学的に許容される担体、賦形剤、希釈剤、安定剤、溶解補助剤、乳化 剤、緩衝剤、無痛化剤、保存剤、着色剤などを添加した配合剤として使用することがで きる。 The method for preparing a diagnostic agent for pollen allergic disease using the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is not particularly limited, but the protein and the partial protein purified according to the above-mentioned method are used. Peptides and their derivatives can be concentrated and collected as a liquid, or further freeze-dried and collected as a solid, and can be prepared and used alone or as an appropriate combination agent. One of the diagnostic methods described below, the diagnostic agent used in the stress test is not only a single form, but also a physiologically acceptable carrier, excipient, diluent, stabilizer, solubilizer, emulsifier, buffer Agent, soothing agent, preservative, coloring agent and the like.
また、後述する診断方法のひとつである、試験管内試験に使用する診断薬として は、単独の形態はもとより、生理学的、免疫学的あるいは免疫化学的に許容される担 体、賦形剤、希釈剤、安定剤、溶解補助剤、乳化剤、緩衝剤、保存剤、着色剤などを 添加した配合剤として使用することができる。  In addition, as a diagnostic agent used in an in vitro test, which is one of the diagnostic methods described later, not only a single form, but also a physiologically, immunologically or immunochemically acceptable carrier, excipient, and dilution can be used. It can be used as a compounding agent to which agents, stabilizers, dissolution aids, emulsifiers, buffers, preservatives, coloring agents and the like are added.
本発明のタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体を有効成 分とする治療薬および予防薬の調製方法には特に制限はないが、前述の方法に準じ て精製されたタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体を濃縮 して液状として調製するか、あるいはさらに凍結乾燥して固状として調製して、単独あ るいは適宜の配合剤として調製し、花粉症に対する治療薬および予防薬に用いること ができる。すなわち単独の形態はもとより、一般的に使用されるアジュバントや各種の 添加剤、賦形剤、希釈剤、安定剤、溶解補助剤、乳化剤、緩衝剤、無痛化剤、保存 剤、着色剤などを添加した配合剤として調製することができる。また、単純糖質、複合 糖質またはそれらの誘導体との複合体として調製することができる。花粉症の予防薬 あるいは治療薬は通常の投与経路、例えば、経口、皮下、皮内、筋肉内、腹腔内、静 脈内等の投与方法により、投与することができる。投薬形態としては、例えば、注射 剤、散剤、細粒剤、顆粒剤、錠剤、カプセル剤、噴霧剤、点眼剤、点鼻剤、パップ剤、 ローション剤、トローチ、舌下錠などがあげられる。  The method for preparing a therapeutic or prophylactic agent comprising the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is not particularly limited. However, a protein purified according to the above-described method, a protein purified from the protein, The partial peptides and their derivatives are concentrated and prepared as a liquid, or further freeze-dried and prepared as a solid, and prepared alone or as an appropriate combination, and used as a therapeutic or prophylactic agent for hay fever. Can be used. That is, in addition to the single form, commonly used adjuvants, various additives, excipients, diluents, stabilizers, dissolution aids, emulsifiers, buffers, soothing agents, preservatives, coloring agents, etc. It can be prepared as an added compounding agent. Further, it can be prepared as a complex with simple saccharide, complex saccharide, or a derivative thereof. The prophylactic or therapeutic agent for hay fever can be administered by a usual administration route, for example, an oral, subcutaneous, intradermal, intramuscular, intraperitoneal, intravenous, etc. administration method. Dosage forms include, for example, injections, powders, fine granules, granules, tablets, capsules, sprays, eye drops, nasal drops, cataplasms, lotions, troches, sublingual tablets and the like.
3.アレルギー疾患の減感作剤およびその使用方法  3. Desensitizers for allergic diseases and their use
本発明のタンパク質、該タンパク質の部分ペプチドおよびそれらの誘導体を有効成 分とする減感作剤は、上記の薬剤と同様に調製され、アレルゲンを含む通常一般の減 感作剤と同様に使用することができる。以下にその使用例を示すが、何らこれに限定 されるものではない。  The desensitizer containing the protein of the present invention, a partial peptide of the protein and a derivative thereof as an active ingredient is prepared in the same manner as the above-mentioned drugs, and is used in the same manner as a general desensitizer containing an allergen. be able to. The use example is shown below, but it is not limited to this.
通常、前述の本発明のタンパク質、該タンパク質の部分ペプチドあるいはそれらの誘 導体を有効成分とする診断薬、または本発明のタンパク質、該タンパク質の部分ぺプ チドあるいはそれらの誘導体と既知アレルゲンあるいは該アレルゲン誘導体を有効成 分とする診断薬などを用いた皮膚テストにおける診断結果に基づいて適切な薬剤、用 量および用法を決定する。皮膚テストにおいて診断薬の適量を投与し、 15〜30分間経 過後の投与部位での膨疹径あるいは発赤径が一定以上の値を示した場合を陽性とす る。陽性と判断された患者について、本発明のタンパク質、該タンパク質の部分ぺプチ ドおよびそれらの誘導体、または本発明のタンパク質、該タンパク質の部分ペプチドあ るレ、はそれらの誘導体と既知アレルゲンあるいは該アレルゲン誘導体を有効成分とす る減感作剤を経口あるいは非経口に投与する。投与量は、患者の症状、投与経路に より異なる力 通常、投与後の経過を観察しながら、成人 1回あたり約 100 // g以下とな るよう、好ましくは 10 z g以下を目安として用量を適宜に選択〔特開平 7 - 179494、特開 平 7-133229〕し、毎週 1回ないし毎月 1回の頻度で、反復投与する。例えば通常行わ れている減感作剤の皮下投与の場合、皮膚テストの中で最も高感度なテストである、 皮内テストで陽性を示す最少濃度の 10分の 1濃度のアレルゲンを含む減感作剤 0.05 mlを初回投与量とする。上述の反復投与による経過を観察しながら、投与局所の膨張 が直径約 5cm以内になるように徐々に用量を増やしながら、週 1回から月 1回の反復投 与を行う。投与局所の膨張が直径約 5cmとなる投与量を上限として、以後反復投与を 継続する。 Usually, a diagnostic agent containing the above-mentioned protein of the present invention, a partial peptide of the protein or a derivative thereof as an active ingredient, or a protein of the present invention, a partial peptide of the protein or a derivative thereof and a known allergen or the allergen Appropriate drugs, dosages and usages are determined based on the results of skin tests using a diagnostic drug or the like whose derivative is the active ingredient. In the skin test, an appropriate amount of the diagnostic agent is administered, and if the wheal diameter or redness diameter at the administration site after 15 to 30 minutes is a certain value or more, the result is considered positive. For a patient determined to be positive, the protein of the present invention, a partial peptide of the protein Or a derivative thereof, or the protein of the present invention, or a partial peptide of the protein, orally or parenterally administer the derivative and a known allergen or a desensitizer containing the allergen derivative as an active ingredient. . The dose varies depending on the patient's symptoms and the route of administration.In general, the dose should be about 100 // g or less per adult, preferably 10 zg or less, while observing the course after administration. It is appropriately selected [JP-A-7-179494, JP-A-7-133229], and is administered repeatedly once a week or once a month. For example, in the case of the usual subcutaneous administration of a desensitizing agent, desensitization containing 1/10 of the minimum concentration of allergen that is positive in the intradermal test, which is the most sensitive skin test The initial dose is 0.05 ml of the drug. While observing the course of the repeated administration described above, repeat the administration once a week to once a month while gradually increasing the dose so that the local swelling of the administration is within about 5 cm in diameter. Repeated administration is continued up to the upper limit of the dose at which the swelling of the administration site is about 5 cm in diameter.
該タンパク質の部分ペプチドまたはその誘導体の中には、 IgEとの結合反応を有さ ず、かつ T細胞ェピトープを含んだものがある。それらの性質を有する該タンパク質の 部分ペプチドまたはその誘導体を有効成分とする減感作剤は、通常の減感作剤と同 様に減感作作用を有するほか、さらにアナフィラキシー反応を回避できる利点をも有 するため、副作用の少なレヽ減感作剤として有用である。  Some partial peptides of the protein or derivatives thereof have no binding reaction with IgE and contain T cell epitopes. A desensitizer containing a partial peptide of the protein having such properties or a derivative thereof as an active ingredient has a desensitizing effect similar to a normal desensitizer, and further has the advantage of avoiding an anaphylactic reaction. It is also useful as a sensitizer with low side effects.
4.アレルギー疾患の診断方法  4.How to diagnose allergic diseases
アレルギー疾患の診断方法には、被験者に直接アレルゲンを負荷してアレルギー反 応を観察する負荷試験と、被験者の組織より調製した試料を用いて、アレルゲンとの 反応性を検出する試験管内試験などがあげられる。被験者の組織より調製した試料に は、末梢血より分離した血清、肥満細胞や好塩基球などの白血球、およびリンパ球な どがあげられる。  Methods for diagnosing allergic diseases include a stress test in which a subject is directly loaded with an allergen to observe an allergic reaction, and an in vitro test in which a sample prepared from the subject's tissue is used to detect reactivity with the allergen. can give. Samples prepared from the subject's tissue include serum separated from peripheral blood, leukocytes such as mast cells and basophils, and lymphocytes.
負荷試験としては、皮膚テストや誘発試験などがあげられる。  Examples of the load test include a skin test and a provocation test.
試験管内試験としては、アレルゲン特異的 IgEを検出する酵素免疫測定法や放射性 免疫測定法、アレルゲン特異的 IgEを介した活性化による肥満細胞や好塩基球力 の ヒスタミン遊離を検出するヒスタミン遊離試験、アレルゲン特異的なリンパ球増殖応答 を検出する増殖応答試験などがあげられる。  In vitro tests include enzyme immunoassays and radioimmunoassays that detect allergen-specific IgE, histamine release tests that detect histamine release of mast cells and basophils by activation through allergen-specific IgE, Proliferation response tests that detect allergen-specific lymphocyte proliferation responses are examples.
上述の試験は、アレルゲンの特定や治療経過の効果判定を目的として行われる(ァ トビー 'アレルギー性疾患;監修 山村雄一、吉利和、中山書店、 1992) o  The above-mentioned studies are conducted to identify allergens and determine the effects of the course of treatment (Artby's Allergic Diseases; supervised by Yuichi Yamamura, Yoshikazu Giri, Nakayama Shoten, 1992) o
皮膚テスト、誘発試験などの負荷試験にぉレ、てアレルゲンを負荷する組織としては、 皮膚、鼻、眼粘膜、気道などがあげられる。  Allergen-loaded tissues such as skin tests and challenge tests include skin, nose, ocular mucosa, and respiratory tract.
皮膚テストの方法としては、プリックテスト、スクラッチテストおよび皮内テストなどがあ げられる。ブリックテスト、スクラッチテストでは、アレルゲンを被験者の上皮に負荷し、 皮内テストでは、アレルゲンを被験者の皮内に負荷する。 Skin testing methods include prick test, scratch test and intradermal test. I can do it. In the brick test and scratch test, an allergen is loaded on the subject's epithelium, and in the intradermal test, the allergen is loaded on the subject's skin.
誘発試験としては、鼻粘膜誘発試験、眼粘膜誘発試験、吸入誘発試験などがあげら れる。鼻粘膜誘発試験では被験者の鼻粘膜に、眼粘膜誘発試験では被験者の眼粘 膜に、吸入誘発試験では被験者の気道にアレルゲンを負荷する。  The provocation test includes a nasal mucosal provocation test, an ocular mucosal provocation test, and an inhalation provocation test. Allergens are loaded into the nasal mucosa of the subject in the nasal mucosa provocation test, to the subject's eye mucosa in the ocular mucosa provocation test, and to the subject's airway in the inhalation provocation test.
負荷とは、ブリックテスト、スクラッチテストの場合、上皮をひっかいたり、つついたりし ながらアレルゲンを反応させることであり、皮内テストの場合、アレルゲンを皮内注射す ることであり、誘発試験の場合、アレルゲン液を噴霧、塗布、吸入させたり、アレルゲン 付着紙片を粘膜に貼付することである。  Stress is the reaction of an allergen while scratching or pecking the epithelium in the case of a brick test or a scratch test.In the case of an intradermal test, the allergen is injected intradermally.In the case of a provocation test, Spraying, applying, and inhaling an allergen solution, and sticking an allergen-adhered paper piece to a mucous membrane.
試験管内試験について、下記に述べる。  The test in a test tube is described below.
酵素免疫測定法、放射性免疫測定法としては、適当な担体に固相化したアレルゲン に、被験者力 採取した血液中の血清を反応させてアレルゲンに結合した IgEを検出 す 方法である、 ELISA、enzyme— imKed immunosorbent assay入 RAST(radio-allergoso rbent est)などがあげられる。  Enzyme immunoassay and radioimmunoassay are methods for detecting IgE bound to allergen by reacting serum in the blood collected from the subject with an allergen immobilized on a suitable carrier. — RAST (radio-allergosorbent est) with imKed immunosorbent assay.
ヒスタミン遊離試験は、被験者の末梢血より調製した肥満細胞、好塩基球などの白血 球画分、あるいは、被験者血清を反応させた健常人の末梢血より調製した白血球画 分に、アレルゲンを反応させることにより放出されるヒスタミンを検出する方法である。リ ンパ球増殖試験とは、被験者の末梢血より分離したリンパ球に対してアレルゲンを添 加した際に起こる T細胞あるいは B細胞の活性化反応または増殖応答を検出する方 法である。  The histamine release test involves reacting an allergen with a leukocyte fraction, such as mast cells and basophils, prepared from the peripheral blood of a subject, or a leukocyte fraction prepared from the peripheral blood of a healthy subject to which the serum of the subject has been reacted. This is a method for detecting histamine released by the method. The lymphocyte proliferation test is a method for detecting the activation or proliferative response of T cells or B cells that occurs when allergen is added to lymphocytes separated from the peripheral blood of a subject.
5.タンパク質、該タンパク質の部分ペプチドまたはそれらの誘導体を認識する抗体 本発明の抗体としては、モノクローナル抗体、ポリクローナル抗体、およびそれらを 含む抗血清などがあげられる。  5. Antibodies Recognizing Proteins, Partial Peptides of the Proteins or Their Derivatives Examples of the antibodies of the present invention include monoclonal antibodies, polyclonal antibodies, and antisera containing them.
本発明のモノクロ一ナル抗体としては、ハイプリドーマが産生する抗体、ヒト化抗体お よびヒト抗体などがあげられる。  Examples of the monoclonal antibody of the present invention include antibodies produced by hybridomas, humanized antibodies, human antibodies, and the like.
ハイプリドーマとは、ヒト以外の哺乳動物に抗原を免疫して取得された B細胞と、マウ ス等に由来するミエローマ細胞とを細胞融合させて得られる、所望の抗原特異性を有 したモノクローナル抗体を産生する細胞を意味する。  A hybridoma is a monoclonal antibody having a desired antigen specificity obtained by cell fusion of B cells obtained by immunizing a non-human mammal with an antigen and myeloma cells derived from a mouse or the like. Means a cell that produces
ヒト化抗体としては、ヒト型キメラ抗体、ヒト型相同性決定領域(complementarity deter mining region:以下、 CDRと略記する)移植抗体などがあげられる。  Examples of the humanized antibody include a human-type chimeric antibody, a human-type homology determining region (hereinafter, abbreviated as CDR) -grafted antibody, and the like.
ヒト型キメラ抗体は、ヒト以外の動物の抗体重鎖可変領域(以下、重鎖は H鎖として、 可変領域は V領域として HVまたは VHとも称す)および抗体軽鎖可変領域 (以下、軽鎖 は L鎖として LVまたは VLとも称す)とヒト抗体の重鎖定常領域(以下、定常領域は C領 域として CHとも称す)およびヒト抗体の軽鎖定常領域 (以下、 CLとも称す)とからなる抗 体を意味する。ヒト以外の動物としては、マウス、ラット、ハムスター、ラビット等、ハイブリ ドーマを作製することが可能であれば、いかなるものも用いることができる。 The human chimeric antibody is composed of an antibody heavy chain variable region (hereinafter, the heavy chain is referred to as an H chain, the variable region is referred to as HV or VH as a V region) and an antibody light chain variable region (hereinafter, referred to as a light chain, The L chain is also called LV or VL, and the heavy chain constant region of a human antibody And the light chain constant region of a human antibody (hereinafter also referred to as CL). As animals other than humans, any animal can be used as long as hybridomas can be produced, such as mice, rats, hamsters, and rabbits.
本発明のヒト型キメラ抗体は、本発明の花粉抗原に特異的に反応するモノクローナ ル抗体を生産するハイブリドーマより、 VHおよび VLをコードする cDNAを取得し、ヒト抗 体 CHおよびヒト抗体 CLをコードする遺伝子を有する発現ベクターにそれぞれ挿入し てヒト型キメラ抗体発現べクタ一を構築し、宿主細胞へ導入することにより発現させ、製 造することができる。  The human chimeric antibody of the present invention obtains cDNAs encoding VH and VL from a hybridoma that produces a monoclonal antibody that specifically reacts with the pollen antigen of the present invention, and obtains human antibody CH and human antibody CL. A human-type chimeric antibody expression vector can be constructed by inserting each into an expression vector having a gene to be encoded, and can be expressed and produced by introducing into a host cell.
ヒト型キメラ抗体の CHとしては、ヒトイムノグロブリン(以下、 hlgと表記する)に属すれ ばいかなるものでもよレ、が、 hlgGクラスのものが好適であり、更に hlgGクラスに属する hi gGl、 hIgG2、 hIgG3、 hIgG4といったサブクラスのいずれも用いることができる。また、ヒト 型キメラ抗体の CLとしては、 hlgに属すればいかなるものでもよぐ κクラスあるいは λ クラスのものを用レ、ること力 Sできる。 As the CH of the human chimeric antibody, any CH can be used as long as it belongs to human immunoglobulin (hereinafter, referred to as hlg). However, the hlgG class is preferable, and the higGl and hIH belong to the hlgG class. g G2, hIgG3, any subclass such hIgG4 can be used. As the CL of the human chimeric antibody, any CL belonging to hlg can be used, which can be of κ class or λ class.
ヒト型 CDR移植抗体は、ヒト以外の動物の抗体の VHおよび VLの CDRのアミノ酸配列 をヒト抗体の VHおよひ VLの適切な位置に移植した抗体を意味する。  The human CDR-grafted antibody means an antibody obtained by grafting the amino acid sequences of CDRs of VH and VL of an antibody of a non-human animal at appropriate positions of VH and VL of a human antibody.
本発明のヒト型 CDR移植抗体は、本発明の花粉抗原に特異的に反応するヒト以外の 動物の抗体の VHおよび VLの CDR配列を任意のヒト抗体の VHおよび VLの CDR配列 に移植した V領域をコードする cDNAを構築し、ヒト抗体の CHおよびヒト抗体の CLをコ —ドする遺伝子を有する発現ベクターにそれぞれ揷入してヒト型 CDR移植抗体発現べ クタ一を構築し、該発現ベクターを宿主細胞へ導入することによりヒト型 CDR移植抗体 を発現させ、製造することができる。  The human CDR-grafted antibody of the present invention is obtained by grafting VH and VL CDR sequences of a non-human animal antibody specifically reacting with the pollen antigen of the present invention into VH and VL CDR sequences of any human antibody. CDNA encoding the region is constructed and inserted into an expression vector having genes encoding human antibody CH and human antibody CL, respectively, to construct a human CDR-grafted antibody expression vector, and the expression vector Can be expressed into a host cell to express and produce a human CDR-grafted antibody.
ヒト型 CDR移植抗体の CHとしては、 hlgに属すればいかなるものでもよいが、 hlgGクラ スのものが好適であり、更に hlgGクラスに属する hIgGl、 hIgG2、 hIgG3、 hIgG4といった サブクラスのいずれも用いることができる。また、ヒト型 CDR移植抗体の CLとしては、 hlg に属すればいかなるものでもよぐ κクラスあるいはえクラスのものを用いることができ る。 The CH of human CDR-grafted antibody may be any so long as it belongs to the hlg, are preferred those hlgG class, further hI g Gl belonging to hlgG class, hIgG2, hIgG3, any subclass such hIgG4 Can be used. As the CL of the human-type CDR-grafted antibody, any CL can be used as long as it belongs to hlg, or a κ class or え class.
ヒト抗体は、元来、ヒト体内に天然に存在する抗体を意味するが、最近の遺伝子工学 的、細胞工学的、発生工学的な技術の進歩により作製されたヒト抗体ファージライブラ リーおよびヒト抗体産生トランスジエニック動物から得られる抗体等も含まれる。  Human antibodies originally mean antibodies naturally occurring in the human body, but human antibody phage libraries and human antibody production produced by recent advances in genetic engineering, cell engineering, and developmental engineering techniques. Antibodies and the like obtained from transgenic animals are also included.
ヒト体内に存在する抗体は、例えば、ヒト末梢血リンパ球を単離し、 EBウィルス等を感 染させ不死化、クローニングすることにより、該抗体を産生するリンパ球を培養でき、培 養物中より該抗体を精製することができる。  Antibodies present in the human body can be isolated, for example, by isolating human peripheral blood lymphocytes, infecting and immortalizing EB virus, etc., and cloning the lymphocytes that produce the antibodies. The antibody can be purified.
ヒト抗体ファージライブラリ一は、ヒト B細胞から調製した抗体遺伝子をファージ遺伝 子に揷入することにより Fab、一本鎖抗体等の抗体断片をファージ表面に発現させたラ イブラリーである。該ライブラリーより、抗原を固定化した基質に対する結合活性を指標 として所望の抗原結合活性を有する抗体断片を発現しているファージを回収すること ができる。該抗体断片は、更に遺伝子工学的手法により、 2本の完全な H鎖および 2本 の完全な L鎖からなるヒト抗体分子へも変換することができる。 The human antibody phage library is a phage gene encoding antibody gene prepared from human B cells. This is a library in which antibody fragments, such as Fab and single-chain antibodies, are expressed on the phage surface by insertion into a phage. From the library, phages expressing antibody fragments having the desired antigen-binding activity can be recovered using the binding activity to the substrate on which the antigen is immobilized as an index. The antibody fragment can be further converted to a human antibody molecule consisting of two complete H chains and two complete L chains by genetic engineering techniques.
ヒト抗体産生トランスジヱニック動物は、ヒト抗体遺伝子が細胞内に組込まれた動物を 意味する。具体的には、マウス ES細胞ヘヒト抗体遺伝子を導入し、該 ES細胞を他のマ ウスの初期胚へ移植後、発生させることによりヒト抗体産生トランスジエニック動物を作 製することができる。ヒト抗体産生トランスジエニック動物からのヒト抗体の作製は、通常 のヒト以外の哺乳動物で行われているハイプリドーマ作製方法によりヒト抗体産生ノ、ィ プリドーマを得、培養することで培養物中にヒト抗体を産生蓄積させることにより行うこと ができる。  A human antibody-producing transgenic animal means an animal in which a human antibody gene has been integrated into cells. Specifically, a human antibody-producing transgenic animal can be produced by introducing a human antibody gene into mouse ES cells, transplanting the ES cells into an early embryo of another mouse, and then developing the embryo. Production of human antibodies from human antibody-producing transgenic animals can be performed in a culture by obtaining human antibody-producing antibodies and ipridomas by the usual method for producing hybridomas in mammals other than humans, and culturing them. It can be performed by producing and accumulating human antibodies.
本発明の抗体は、以下に述べる抗体断片も包含する。  The antibodies of the present invention also include the antibody fragments described below.
抗体断片としては、 Fab (fragment of antigen bindingの略)、 Fab'、 F(ab')2、一本鎖 抗体(single chain Fv :以下、 scFvと略記する)、ジスルフイド安定化抗体(disulfide sta bilized Fv;dsFv)、 CDRを含むペプチドなどがあげられる。 Examples of antibody fragments include Fab (abbreviation of fragment of antigen binding), Fab ′, F (ab ′) 2 , single chain antibody (hereinafter abbreviated as scFv), disulfide stabilized antibody (disulfide sta bilized). Fv; dsFv), peptides including CDRs, and the like.
Fabは、 IgGをタンパク質分解酵素パパインで処理して得られる断片のうち(H鎖の 22 4番目のアミノ酸残基で切断される)、 H鎖の N末端側約半分のアミノ酸と L鎖全体がジ スルフイド結合で結合した分子量約 5万の抗原結合活性を有する抗体断片である。 本発明の Fabは、本発明の花粉抗原に特異的に反応する抗体をタンパク質分解酵 素パパインで処理して得ることができる。または、該抗体の Fabをコードする DNAを発 現べクタ一に挿入し、該ベクターを宿主細胞へ導入することにより発現させ、 Fabを製 造すること;^できる。  Fab is a fragment obtained by treating IgG with proteolytic enzyme papain (which is cleaved at amino acid residue 224 of H chain). It is an antibody fragment having a molecular weight of about 50,000 and having antigen-binding activity, which is linked by a disulfide bond. The Fab of the present invention can be obtained by treating an antibody that specifically reacts with the pollen antigen of the present invention with the proteolytic enzyme papain. Alternatively, a DNA encoding the Fab of the antibody is inserted into an expression vector, and the vector is expressed by introducing the vector into a host cell to produce a Fab;
F(ab')2は、 IgGをタンパク質分解酵素ペプシンで処理して得られる断片のうち(H鎖 の 234番目のアミノ酸残基で切断される)、 Fabがヒンジ領域のジスルフイド結合を介し て結合されたものよりやや大きい、分子量約 10万の抗原結合活性を有する抗体断片 である。 F (ab ') 2 is a fragment obtained by treating IgG with the protease pepsin (which is cleaved at the 234th amino acid residue in the H chain). Fab binds via a disulfide bond in the hinge region. This is an antibody fragment having a molecular weight of about 100,000 and having antigen-binding activity, which is slightly larger than that obtained.
本発明の F(ab')2は、本発明の花粉抗原に特異的に反応する抗体をタンパク質分解 酵素ペプシンで処理して得ることができる。または、下記の Fa をチォエーテル結合あ る!/、はジスルフイド結合させ、作製すること力 Sできる。 The F (ab ') 2 of the present invention can be obtained by treating an antibody specifically reacting with the pollen antigen of the present invention with the protease pepsin. Or, the following Fa has a thioether bond! /, Is a disulfide bond and can be made S.
Fab'は、上記 F(ab')2のヒンジ領域のジスルフイド結合を切断した分子量約 5万の抗原 結合活性を有する抗体断片である。 Fab 'is an antibody fragment having a molecular weight of about 50,000 and having an antigen-binding activity in which a disulfide bond in the hinge region of F (ab') 2 is cleaved.
本発明の Fab'は、本発明の花粉抗原に特異的に反応する F(ab')2を還元剤ジチォス レイト一ル処理して得ることができる。または、該抗体の Fab'断片をコードする DNAを発 現ベクターに挿入し、該ベクターを宿主細胞へ導入することにより Fab'を発現させ、製 造することができる。 Fab ′ of the present invention is a reducing agent dithios that reduces F (ab ′) 2 which specifically reacts with the pollen antigen of the present invention. It can be obtained by late processing. Alternatively, Fab ′ can be expressed and produced by inserting a DNA encoding the Fab ′ fragment of the antibody into an expression vector and introducing the vector into a host cell.
scFvは、一本の VHと一本の VLとを適当なペプチドリンカ一(以下、 Pと称す)を用レヽ て連結した、 VH— P—VLないしは VL— P— VHポリペプチドを示す。本発明の scFvに 含まれる VHおよび VLは、本発明のハイプリドーマが産生する抗体、ヒト化抗体、ヒト抗 体のレ、ずれをも用レ、ること力;できる。  scFv refers to a VH-P-VL or VL-P-VH polypeptide in which one VH and one VL are linked using an appropriate peptide linker (hereinafter, referred to as P). VH and VL contained in the scFv of the present invention can be used for the antibody, humanized antibody, and human antibody produced by the hybridoma of the present invention.
本発明の scFvは、本発明の花粉抗原に特異的に反応する抗体の VHおよび VLをコ ードする cDNAを取得し、 scFvをコードする DNAを構築し、該 DNAを発現ベクターに揷 入し、該ベクターを宿主細胞へ導入することにより発現させ、 scFvを製造することがで きる。  The scFv of the present invention obtains cDNA encoding VH and VL of an antibody that specifically reacts with the pollen antigen of the present invention, constructs a scFv-encoding DNA, and inserts the DNA into an expression vector. The vector can be expressed by introducing the vector into a host cell to produce an scFv.
dsFvは、 VHおよび VL中のそれぞれ 1アミノ酸残基をシスティン残基に置換したポリ ペプチドを該システィン残基間のジスルフイド結合を介して結合させたものをレ、う。シス ティン残基に置換するアミノ酸残基は Reiterらにより示された方法 [Protein Engineerin g, 7, 697 (1994)]に従って、抗体の立体構造予測に基づいて選択することができる。 本発明の dsFvに含まれる VHおよび VLは本発明のハイブリドーマが産生する抗体、ヒト 化抗体、ヒト抗体のいずれをも用いることができる。  dsFv is obtained by binding a polypeptide in which one amino acid residue in each of VH and VL is replaced with a cysteine residue via a disulfide bond between the cysteine residues. The amino acid residue to be substituted for the cystine residue can be selected based on the prediction of the three-dimensional structure of the antibody according to the method shown by Reiter et al. [Protein Engineering, 7, 697 (1994)]. As the VH and VL contained in the dsFv of the present invention, any of the antibodies, humanized antibodies, and human antibodies produced by the hybridoma of the present invention can be used.
本発明の dsFvは、本発明の花粉抗原に特異的に反応する抗体の VHおよび VLをコ ードする cDNAを取得し、 dsFvをコードする DNAを構築し、該 DNAを発現ベクターに揷 入し、該ベクタ一を宿主細胞へ導入することにより発現させ、 dsFvを製造することがで きる。  The dsFv of the present invention obtains cDNA encoding VH and VL of an antibody that specifically reacts with the pollen antigen of the present invention, constructs a DNA encoding dsFv, and inserts the DNA into an expression vector. Then, the vector can be expressed by introducing the vector into a host cell to produce dsFv.
CDRを含むペプチドは、 H鎖または L鎖 CDRの少なくとも 1領域以上を含んで構成さ れる。複数の CDRは、直接または適当なペプチドリンカ一を介して結合させることがで きる。  The peptide containing the CDR comprises at least one region of the H chain or L chain CDR. Multiple CDRs can be linked directly or via a suitable peptide linker.
本発明の CDRを含むペプチドは、本発明の花粉抗原に特異的に反応する抗体の V Hおよび Vしをコードする cDNAを取得した後、 CDRをコードする DNAを構築し、該 DNA を発現べクタ一に挿入し、該発現べクタ一を宿主細胞へ導入することにより発現させ、 CDRを含むペプチドを製造することができる。  The peptide containing the CDR of the present invention can be obtained by obtaining cDNA encoding VH and VH of an antibody specifically reacting with the pollen antigen of the present invention, constructing a DNA encoding the CDR, and expressing the DNA. The peptide can be expressed by introducing the expression vector into a host cell to produce a peptide containing CDR.
また、 CDRを含むペプチドは、 Fmoc法(フルォレニルメチルォキシカルボニル法)、 t Boc法(t-ブチルォキシカルボニル法)等の化学合成法によって製造することもできる。 以下、これらの作製方法のうち、ハイプリドーマが産生するモノクローナル抗体の作 製方法について述べる。  Further, the peptide containing CDR can also be produced by a chemical synthesis method such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the t Boc method (t-butyloxycarbonyl method). Hereinafter, of these production methods, a method for producing a monoclonal antibody produced by a hybridoma will be described.
本発明のモノクローナル抗体は、抗原を調製して、動物に免疫し、免疫した動物の 細胞と、骨髄腫細胞とを融合させてハイプリドーマを作製し、該ハイブリドーマを培養 するか、動物に投与して該動物を腹水癌化させ、該培養液または腹水を採取すること により作製することができる。また、免疫した動物の免疫血清を採取することによりポリ クローナル抗体を作製することができる。 The monoclonal antibody of the present invention is prepared by preparing an antigen, immunizing an animal, and immunizing the animal. A hybridoma is prepared by fusing cells with myeloma cells, and the hybridoma is cultured or administered to an animal to cause the animal to undergo ascites carcinoma, and the culture solution or ascites is collected. Can be. In addition, polyclonal antibodies can be prepared by collecting immune serum from immunized animals.
動物の免疫は、次のように行う。すなわち、マウス、ラット、ハムスターなどの非ヒト哺 乳動物の皮下、静脈内あるいは腹腔内に、後述のように調製した抗原を免疫して、そ の動物の脾細胞、リンパ節、末梢血中の抗体産生細胞を採取する。  The animals are immunized as follows. That is, an antigen prepared as described below is immunized subcutaneously, intravenously, or intraperitoneally in a non-human mammal, such as a mouse, rat, or hamster, and spleen cells, lymph nodes, and peripheral blood of the animal are immunized. The antibody producing cells are collected.
抗原の調製方法としては、本発明のタンパク質を上述の方法で精製するか、または 本発明のタンパク質、該タンパク質の部分ペプチドまたはそれらの誘導体をコードする DNAを含む組換えべクタ一を大腸菌、酵母、動物細胞、昆虫細胞などの宿主に導入 して、タンパク質あるいはポリペプチドを発現させる方法などがあげられる。または、部 分配列を有するポリペプチドは、アミノ酸合成機を用いて合成することによつても得ら れる。免疫の際に、得られた抗原は、抗原性の高いキャリアタンパク質(キーホールリン ペットへモシァニンや牛血清アルブミンなど)と化学修飾などにより結合させ投与した り、あるいは適当なアジュバント〔例えば、フロインドの完全アジュバント (Freund's Com plete Adjuvant)や水酸化アルミニウムゲルと百日咳菌ワクチンなど〕とともに投与する。 抗原の投与は、 1回目の投与の後:!〜 2週間おきに 5〜10回行う。各投与後 3〜7日 目に眼底静脈叢より採血し、その血清が抗原と反応することを酵素免疫測定法〔Antib odies - A Laboratory Manual, Cold Spring Harbor Laboratory, 1988J どで!^ベ る。免疫に用いた抗原に対し、その血清が十分な抗体価を示した非ヒト哺乳動物を抗 体産生細胞の供給源として提供する。ポリクローナル抗体は、該動物から得られた血 清を分離、精製して得られる。  As a method for preparing an antigen, the protein of the present invention is purified by the above-described method, or a recombinant vector containing a DNA encoding the protein of the present invention, a partial peptide of the protein or a derivative thereof is transformed into Escherichia coli or yeast. And a method of expressing a protein or polypeptide by introducing it into a host such as an animal cell or an insect cell. Alternatively, a polypeptide having a partial sequence can also be obtained by synthesizing using an amino acid synthesizer. At the time of immunization, the obtained antigen is combined with a carrier protein having high antigenicity (keyhole limpet hemocyanin, bovine serum albumin, etc.) by chemical modification or the like, or administered, or an appropriate adjuvant (eg, Freund's). Complete adjuvant (Freund's Complex Adjuvant), aluminum hydroxide gel and B. pertussis vaccine. The antigen is administered after the first dose: 5 to 10 times every 2 weeks. Blood is collected from the fundus venous plexus 3 to 7 days after each administration, and the reaction of the serum with the antigen is determined by an enzyme immunoassay [Antibodidies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988J! I will. A non-human mammal whose serum shows a sufficient antibody titer against the antigen used for immunization is provided as a source of the antibody-producing cells. The polyclonal antibody is obtained by separating and purifying serum obtained from the animal.
骨髄腫細胞としては、マウス力 得られた株化細胞を使用する。たとえば、 8—ァザグ ァニン耐性マウス(BALB/c由来)骨髄腫細胞株 P3- X63Ag8- U 1(P3- U l)(Current Top ics in Microbiology and Immunology, 18 :1-7, 1978)、P3_NS1/1 - Ag41(NS_l)(Europe an J. Immunology, 6: 51 1—519, 1976)、 SP2/0-Agl4(SP-2)(Nature,276: 269-270, 19 78)、 P3-X63-Ag8653(653) (J. Immunology, 123:1548- 1550,1979)、P3-X63-Ag8(X63) (Nature, 256:495-497, 1975)などが用いられる。  As myeloma cells, cell lines obtained from mice are used. For example, 8-azaguanine-resistant mouse (derived from BALB / c) myeloma cell line P3-X63Ag8-U1 (P3-U1) (Current Topics in Microbiology and Immunology, 18: 1-7, 1978), P3_NS1 / 1-Ag41 (NS_l) (Europe an J. Immunology, 6: 51 1-519, 1976), SP2 / 0-Agl4 (SP-2) (Nature, 276: 269-270, 1978), P3-X63- Ag8653 (653) (J. Immunology, 123: 1548-1550, 1979), P3-X63-Ag8 (X63) (Nature, 256: 495-497, 1975) and the like are used.
上述で得られた抗体産生細胞と骨髄腫細胞を混合し、 HAT培地〔正常培地にヒポキ サンチン、チミジンおよびアミノプテリンを加えた培地〕に懸濁したのち、 7〜14日間培 養する。培養後、培養上清の一部をとり酵素免疫測定法などにより、選択する。つい で、限界希釈法によりクロ一ニングを行レ、、酵素免疫測定法により安定して高い抗体 価の認められたものを抗花粉抗原モノクローナル抗体産生ハイプリドーマ株として選 択する。 The antibody-producing cells and myeloma cells obtained above are mixed, suspended in a HAT medium (a medium in which hypoxanthin, thymidine and aminopterin are added to a normal medium), and cultured for 7 to 14 days. After the culture, a part of the culture supernatant is taken and selected by enzyme immunoassay or the like. Then, the clones were cloned by limiting dilution, and those with a stable high antibody titer determined by enzyme immunoassay were selected as anti-pollen antigen monoclonal antibody-producing hybridoma strains. Select.
モノクローナル抗体は、上述の該ハイブリドーマ細胞を培養するか、ハイプリドーマ細 胞を動物の腹腔内に投与して該動物を腹水癌化させ、該培養液または腹水を採取、 精製することにより作製できる。  A monoclonal antibody can be produced by culturing the above-described hybridoma cells or administering the hybridoma cells into the peritoneal cavity of an animal to cause the animal to develop ascites cancer, and collecting and purifying the culture solution or ascites.
なお、ポリクローナル抗体およびモノクローナル抗体の精製方法としては、遠心分 離、硫安沈殿、力プリル酸沈殿法、または DEAE—セファロースカラム、陰イオン交換力 ラム、プロテイン Aまたは G—カラムあるレ、はゲル濾過カラムなどを用!/、たクロマトグラフ ィーなどを組み合わせる方法があげられる。  Methods for purifying polyclonal and monoclonal antibodies include centrifugation, ammonium sulfate precipitation, and hydroprilic acid precipitation, or DEAE-Sepharose column, anion exchange column, protein A or G-column, or gel filtration. Use a column! /, And a method of combining chromatography and the like.
上述の方法で得られる本発明のモノクローナル抗体としては、モノクローナル抗体 K M2715があげられる。モノクローナル抗体 KM2715を産生するハイブリドーマ KM2715 は、平成 12年 4月 27日付でブダペスト条約に基づき通商産業省工業技術院生命工学 工業技術研究所(日本国茨城県つくば巿東 1丁目 1番 3号)に FERM BP- 7149として 寄託されている。  The monoclonal antibody of the present invention obtained by the above-mentioned method includes the monoclonal antibody KM2715. Based on the Budapest Treaty, the hybridoma KM2715, which produces the monoclonal antibody KM2715, was contacted by the Ministry of International Trade and Industry, Institute of Industrial Science and Technology, Institute of Biotechnology and Industrial Technology (1-3 1-3 Tsukuba East, Ibaraki, Japan) based on the Budapest Treaty. Deposited as FERM BP-7149.
6.抗体を有効成分とするアレルギー疾患の診断薬、治療薬および予防薬  6.Diagnostic, therapeutic and prophylactic agents for allergic diseases using antibodies as active ingredients
上記 5で得られた抗体またはその抗体断片は、アレルギー疾患の診断薬、治療薬ま たは予防薬として用いることができる。  The antibody or antibody fragment thereof obtained in the above 5 can be used as a diagnostic, therapeutic or prophylactic agent for allergic diseases.
本発明の抗体またはその抗体断片を有効成分とする診断薬、治療薬または予防薬 は、上記 2に記載した本発明のタンパク質、該タンパク質の部分ペプチドおよびそれら の誘導体を有効成分とするアレルギー疾患の診断薬、治療薬ならびに予防薬の調製 方法と同様の方法を用いて調製することができる。  Diagnostic, therapeutic or prophylactic agents comprising the antibody of the present invention or an antibody fragment thereof as an active ingredient are allergic diseases comprising the protein of the present invention described in 2 above, a partial peptide of the protein and a derivative thereof as an active ingredient. It can be prepared using the same method as that for preparing diagnostic, therapeutic and prophylactic agents.
7.抗体を有効成分とするアレルギー疾患の治療方法および予防方法  7. Method for treating and preventing allergic diseases using antibodies as active ingredients
本発明の花粉抗原に対する抗体またはその抗体断片を用レ、た、アレルギー疾患を 治療または予防する方法としては、本発明の抗体またはその抗体断片を含有してなる 組成物を、アレルギー疾患患者の鼻、口、気道などの粘膜組織、皮膚、 目などに直接 投与させて、アレルゲンが体内に侵入することを阻害することにより、アレルギー疾患 を治療または予防することができる。また、本発明の抗体またはその抗体断片を含有し てなる組成物を、アレルギー疾患患者に静脈内投与させて、体内に進入したアレルゲ ンを除去することにより、アレルギー疾患を治療または予防することができる。  As a method for treating or preventing an allergic disease using an antibody against a pollen antigen or an antibody fragment thereof of the present invention, a composition comprising the antibody or an antibody fragment thereof of the present invention may be used in a method for treating nose of an allergic disease patient. It can be administered or administered directly to mucosal tissues such as the mouth and respiratory tract, the skin, the eyes, etc., to prevent allergens from entering the body, thereby treating or preventing allergic diseases. Further, it is possible to treat or prevent an allergic disease by removing the allergen that has entered the body by intravenously administering a composition comprising the antibody or the antibody fragment thereof of the present invention to a patient with an allergic disease. it can.
8.抗体を有効成分とするアレルギー疾患の診断方法  8. Diagnosis method of allergic disease using antibody as active ingredient
本発明の花粉抗原に対する抗体またはその抗体断片を用いた、アレルギー疾患を 診断する方法としては、本発明の花粉抗原を免疫学的に定量または検出する方法が あげられる。方法としては、蛍光抗体法、免疫酵素抗体法 (ELISA)、放射性物質標識 免疫抗体法(RIA)、免疫細胞染色法、ウエスタンブロッテイング法、免疫沈降法、上記 に記した酵素免疫測定法、サンドイッチ EL1SA 法 [単クローン抗体実験マニュアル (講 談社サイエンティフィック、 1987年)、続生化学実験講座 5 免疫生化学研究法 (東京 化学同人、 1986年)]などがあげられる。 Examples of a method for diagnosing an allergic disease using an antibody against the pollen antigen of the present invention or an antibody fragment thereof include a method for immunologically quantifying or detecting the pollen antigen of the present invention. Methods include fluorescent antibody method, immunoenzymatic antibody method (ELISA), radioactive substance-labeled immunological antibody method (RIA), immunocytostaining method, western blotting method, immunoprecipitation method, Enzyme immunoassay, sandwich EL1SA method [Monoclonal antibody experiment manual (Kodansha Scientific, 1987), Seismic Chemistry Laboratory Course 5 Immunobiochemical research method (Tokyo Kagaku Dojin, 1986)] Is raised.
资光抗体法は、文献 [Monoclonal Antibodies: Principles and practice, Third editi on (Academic Press, 1996), 単クローン抗体実験マニュアル(講談社サイェンティフィ ック、 1987) ]等に記載された方法を用いて行うことができる。具体的には、生体内から 分離された鼻粘膜、口蓋粘膜、気道粘膜、皮膚といった花粉抗原にさらされる組織、 あるいは涙液、鼻汁などの体液、血液および血液細胞などに、本発明の抗体またはそ の抗体断片を反応させ、さらにフルォレシン'イソチオシァネート(FITC)またはフィコェ リスリンなどの蛍光物質でラベルした抗ィムノグロブリン抗体などを反応させた後、蛍光 色素を蛍光光度計またはフローサイトメータ一で測定し、花粉抗原を定量または検出 する方法である。  The antibody antibody method should be performed using the method described in the literature [Monoclonal Antibodies: Principles and practice, Third edition (Academic Press, 1996), Monoclonal antibody experiment manual (Kodansha Scientific, 1987)], etc. Can be. Specifically, the antibody of the present invention is applied to tissues exposed to pollen antigens such as nasal mucosa, palatal mucosa, respiratory mucosa, and skin isolated from a living body, or body fluids such as tears and nasal secretions, blood and blood cells, and the like. After reacting the antibody fragment, and further reacting with an anti-immunoglobulin antibody labeled with a fluorescent substance such as fluorescin'isothiocyanate (FITC) or phycoerythrin, the fluorescent dye is measured with a fluorometer or flow cytometer. This is a method for measuring or measuring pollen antigens at a time.
免疫酵素抗体法 (ELISA)は、生体内から分離された鼻粘膜、 口蓋粘膜、気道粘膜、 皮膚といった花粉抗原にさらされる組織、あるいは涙液、鼻汁などの体液、血液およ び血液細胞などに、本発明の抗体またはその抗体断片を反応させ、さらにペルォキシ ダーゼ、ビォチンなどの酵素標識などを施した抗ィムノグロブリン抗体などを反応させ た後、発色色素を吸光光度計で測定し、花粉抗原の定量または検出を行う方法であ る。  The immunoenzyme-linked immunosorbent assay (ELISA) is applied to tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, airway mucosa, and skin isolated from the body, or body fluids such as tears and nasal secretions, blood, and blood cells. After reacting the antibody of the present invention or an antibody fragment thereof, and further reacting with an anti-immunoglobulin antibody labeled with an enzyme such as peroxidase or biotin, the coloring pigment is measured with an absorptiometer, and the pollen antigen is detected. This is a method for quantifying or detecting
放射性物質標識免疫抗体法 (RIA)は、生体内から分離された鼻粘膜、 口蓋粘膜、 気道粘膜、皮膚といった花粉抗原にさらされる組織、あるいは涙液、鼻汁などの体液、 血液および血液細胞などに、本発明の抗体またはその抗体断片を反応させ、さらに放 射線標識を施した抗ィムノグロブリン抗体などを反応させた後、シンチレ一シヨンカウン ターなどで測定し、花粉抗原を定量または検出する方法である。  Radiolabeled immunoassay (RIA) is applied to tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, respiratory mucosa and skin isolated from the body, or body fluids such as tears and nasal secretions, blood and blood cells, etc. After reacting the antibody of the present invention or an antibody fragment thereof, and further reacting with an anti-immunoglobulin antibody which has been subjected to radiolabeling, measurement is performed using a scintillation counter or the like to quantify or detect pollen antigens. is there.
免疫細胞染色法は、生体内力 分離された鼻粘膜、 口蓋粘膜、気道粘膜、皮膚とレ、 つた花粉抗原にさらされる組織、あるいは血液細胞などに、本発明の抗体またはその 抗体断片を反応させ、さらにフルォレシン'イソチオシァネート (FITC)などの蛍光物 質、ペルォキシダーゼなどの酵素標識を施した抗ィムノグロブリン抗体などを反応させ た後、顕微鏡を用いて観察する方法である。  In the immunocytochemical staining method, the antibody of the present invention or an antibody fragment thereof is reacted with intranasally separated nasal mucosa, palate mucosa, respiratory mucosa, skin and tissue exposed to ivy pollen antigen, blood cells, and the like. In addition, a fluorescent substance such as fluorescin'isothiosinate (FITC), an anti-immunoglobulin antibody labeled with an enzyme such as peroxidase, and the like are reacted, and then observed using a microscope.
ウェスタンブロッテイング法は、生体内から分離された鼻粘膜、 口蓋粘膜、気道粘膜、 皮膚といった花粉抗原にさらされる組織、あるいは涙液、鼻汁などの体液、血液およ び血液細胞などを SDS-ポリアクリルアミドゲル電気泳動(Antibodies- A Laboratory M anual, Cold Spring Harbor Laboratory, 1988)で分画した後、該ゲルを PVDF膜ある いはニトロセルロース膜にブロッテイングし、該膜に本発明の抗体またはその抗体断片 を反応させ、さらに FITCなどの蛍光物質、ペルォキシダーゼなどの酵素標識を施した 抗 IgG抗体などを反応させた後、確認する方法である。 The Western blotting method uses SDS-polysaccharide to isolate tissues exposed to pollen antigens, such as nasal mucosa, palate mucosa, respiratory mucosa, and skin, or body fluids such as tears and nasal secretions, blood, and blood cells. After fractionation by acrylamide gel electrophoresis (Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988), the gel was blotted onto a PVDF membrane or a nitrocellulose membrane, and the antibody of the present invention or its antibody was applied to the membrane. Antibody fragment And then reacting with a fluorescent substance such as FITC, an anti-IgG antibody labeled with an enzyme such as peroxidase, and the like, followed by confirmation.
免疫沈降法とは、生体内から分離された鼻粘膜、 口蓋粘膜、気道粘膜、皮膚といつ た花粉抗原にさらされる組織、あるいは涙液、鼻汁などの体液、血液および血液細胞 などを本発明の抗体またはその抗体断片と反応させた後、プロテイン G-セファロース 等のィムノグロブリンに特異的な結合能を有する担体を加えて抗原抗体複合体を沈降 させるものである。  The immunoprecipitation method refers to the nasal mucosa, palatal mucosa, airway mucosa, tissues exposed to pollen antigens such as skin, or body fluids such as tears and nasal secretions, blood and blood cells, etc. of the present invention. After reacting with an antibody or an antibody fragment thereof, a carrier having a specific binding ability to immunoglobulin such as protein G-sepharose is added to precipitate an antigen-antibody complex.
サンドイッチ ELISA法とは、本発明の抗体またはその抗体断片で、抗原認識部位の 異なる 2種類の抗体を利用する方法である。具体的には、あらかじめ一方の抗体また は抗体断片はプレートに吸着させ、もう一方の抗体または抗体断片は FITCなどの蛍 光物質、ペルォキシダーゼなどの酵素で標識しておく。抗体吸着プレートに、生体内 から分離された鼻粘膜、 口蓋粘膜、気道粘膜、皮膚といった花粉抗原にさらされる組 織、あるいは涙液、鼻汁などの体液、血液および血液細胞などを反応後、標識した抗 体またはその抗体断片を反応させ、標識物質に応じた反応を行う方法である。  The sandwich ELISA method is a method using two kinds of antibodies having different antigen recognition sites in the antibody of the present invention or an antibody fragment thereof. Specifically, one antibody or antibody fragment is adsorbed on a plate in advance, and the other antibody or antibody fragment is labeled with a fluorescent substance such as FITC or an enzyme such as peroxidase in advance. Tissues exposed to pollen antigens such as nasal mucosa, palate mucosa, respiratory mucosa and skin isolated from the body, body fluids such as tears and nasal secretions, blood and blood cells, etc. were reacted on the antibody adsorption plate and labeled. This is a method in which an antibody or an antibody fragment thereof is reacted and a reaction is performed according to the labeling substance.
9.抗体の使用方法  9.How to use antibodies
上記 8に記載された免疫学的方法を用いて、花粉の抽出物から本発明のタンパク 質、該タンパク質の部分ペプチドおよびそれらの誘導体を検出または定量することが できる。  The protein of the present invention, the partial peptide of the protein, and their derivatives can be detected or quantified from the pollen extract using the immunological method described in 8 above.
上記 2または 3に記載したアレルゲン抽出物よりなるアレルギー疾患の診断薬、治療 薬、予防薬および減感作剤は、活性および品質が常に一定に保たれている必要があ る。しかしながら、既存の多くのアレルゲンエキスは、植物性物質力 溶媒などを用い て取得される抽出物であるため、活性、品質を一定に保つことが難しい。  The diagnostic, therapeutic, prophylactic and desensitizing agents for allergic diseases consisting of the allergen extract described in 2 or 3 above must be kept constant in activity and quality. However, since many existing allergen extracts are extracts obtained using a plant-based solvent, it is difficult to maintain constant activity and quality.
本発明の新規花粉抗原は、高いアレルゲン活性を有し、かつ保存安定性に優れて いるので、主要アレルゲンと考えられる。したがって、アレルゲンエキス中での該花粉 抗原の含有量および性状を一定に保つことが、活性および品質が一定に保たれたァ レルゲンエキスとなる。  The novel pollen antigen of the present invention is considered to be a major allergen because it has high allergen activity and excellent storage stability. Therefore, keeping the content and properties of the pollen antigen in the allergen extract constant results in an allergen extract having constant activity and quality.
アレルゲンエキス中における該花粉抗原の含有量を測定する方法としては、該花粉 抗原を特異的に検出、定量できる抗体は、該花粉抗原を調べるために簡便で有用な ツールとなり得る。該花粉抗原の定量方法としては、上記 8記載の免疫学的測定方法 があげられ、好ましくは、サンドイッチ ELISAがあげられる。  As a method for measuring the content of the pollen antigen in the allergen extract, an antibody capable of specifically detecting and quantifying the pollen antigen can be a simple and useful tool for examining the pollen antigen. Examples of the method for quantifying the pollen antigen include the immunological measurement method described in the above item 8, preferably a sandwich ELISA.
アレルゲンエキス中における該花粉抗原の安定性を測定する方法としては、上記 8 記載のウェスタンブロッテイング法などがあげられる。  Examples of a method for measuring the stability of the pollen antigen in an allergen extract include the Western blotting method described in the above item 8 and the like.
さらに、本発明の抗体またはその抗体断片は、花粉タンパク抽出物より該タンパク質 を単離 '精製するために使用することができる。 Further, the antibody of the present invention or the antibody fragment thereof is obtained by extracting the protein from the pollen protein extract. Can be used to isolate and purify.
次に実施例をあげて本発明を具体的に説明する。しかし、本発明はこれらの実施例 により何ら限定されるものではなレ、。  Next, the present invention will be specifically described with reference to examples. However, the present invention is not limited by these examples.
図面の簡単な説明 BRIEF DESCRIPTION OF THE FIGURES
第 1図は、本発明のタンパク質の分子量を測定することを目的に、ドデシル硫酸ナトリ ゥム一ポリアクリルアミドゲル電気泳動(以下、 SDS-PAGEと記す)(7.5%ポリアクリルァ ミドゲル濃度)後、クマシ一染色によりタンパク質成分を検出した結果を示す図である。 本発明タンパク質 (New)の対照として未精製総タンパク質画分 (CE)、 Cry j 1 および C ry j 2を使用した。 New、 Cry j 1、 Cryj 2は各 1 μ g、 CEは 10 μ gである。  FIG. 1 shows the results obtained by performing sodium dodecyl sulfate-polyacrylamide gel electrophoresis (hereinafter referred to as SDS-PAGE) (7.5% polyacrylamide gel concentration) for the purpose of measuring the molecular weight of the protein of the present invention. It is a figure showing the result of having detected the protein component by staining. As a control for the protein of the present invention (New), unpurified total protein fraction (CE), Cry j 1 and Cry j 2 were used. New, Cry j 1 and Cry j 2 are each 1 μg, and CE is 10 μg.
第 2図は、本発明のタンパク質に対する、スギ花粉症患者 12名、健常者 4名の血清 中の IgEの反応性を酵素免疫測定法により解析した結果を示す図である。 3 / g/mlで 9 6ウェルマイク口プレートに固相化した該タンパク質 (New)に対し、 2倍に希釈したスギ 花粉症患者および健常人血清を反応させた後、抗原に結合した IgEを抗ヒト IgE抗体に より検出した。対照として、 3 g/mlで固相化した Cry j 1 、 Cry j 2、 30 / g/mlで固相 化した未精製総タンパク画分 (CE)について、該タンパク質と同様の試験を行った。ま た、同時に医療用診断試薬(キャップ RAST FEIA)により、スギ花粉抗原に特異的な I gE抗体価の検査を行レ、、比較検討した結果を示した。  FIG. 2 is a diagram showing the results of analysis of the reactivity of IgE in the sera of 12 cedar pollinosis patients and 4 healthy subjects with the protein of the present invention by enzyme immunoassay. After reacting the protein (New) immobilized on a 96-well microphone opening plate at 3 / g / ml with 2-fold diluted cedar pollinosis patients and healthy human sera, IgE bound to the antigen was removed. Detected by anti-human IgE antibody. As a control, the same test was performed on Cry j 1 and Cry j 2 immobilized at 3 g / ml, and unpurified total protein fraction (CE) immobilized at 30 / g / ml. . At the same time, the IgE antibody titer specific to cedar pollen antigen was examined using a medical diagnostic reagent (Cap RAST FEIA), and the results of comparative examination were shown.
第 3図は、キャップ RAST FEIAによるスギ花粉抗原に特異的な IgE抗体価と、本発明 のタンパク質 (New)、未精製総タンパク質画分 (CE)、 Cry j 1および Cry j 2の各抗原に 特異的な IgE抗体価との相関関係を示す散布図である。  Fig. 3 shows the IgE antibody titer specific to the cedar pollen antigen by the cap RAST FEIA and the antigens of the protein (New), unpurified total protein fraction (CE), Cry j 1 and Cry j 2 of the present invention. It is a scatter diagram showing a correlation with a specific IgE antibody titer.
第 4図は、本発明のモノクローナル抗体 KM2715の反応特異性を酵素免疫測定法 により解析した結果を示す図である。 5 /i g/mlで固相化した抗原ペプチド一 THYコン ジュゲート、および対照ペプチド THYコンジュゲートに対する該モノクローナル抗体 の反応性を比較検討した図である。  FIG. 4 is a diagram showing the results of analyzing the reaction specificity of the monoclonal antibody KM2715 of the present invention by an enzyme immunoassay. FIG. 3 is a diagram comparing the reactivity of the monoclonal antibody with the antigen peptide-THY conjugate immobilized at 5 / ig / ml and the control peptide THY conjugate.
第 5図は、本発明のモノクローナル抗体 KM2715による、未精製総タンパク画分(C E)中の当該タンパク質の特異的検出をウェスタンブロッテイングで解析した結果を示 す図である。 CE 2 μ gを SDS- PAGEに処し、ウェスタンブロティングを行った図であ る。 発明を実施するための最良の形態  FIG. 5 shows the results of Western blotting analysis of the specific detection of the protein in the unpurified total protein fraction (CE) by the monoclonal antibody KM2715 of the present invention. FIG. 2 is a diagram showing 2 μg of CE subjected to SDS-PAGE and Western blotting. BEST MODE FOR CARRYING OUT THE INVENTION
1 .花粉抗原の精製  1.Purification of pollen antigen
日本スギの雄花力ら採取した花粉 100gを、 PBS (phosphate- buffered salin;以下、 PBS と記す) 2Lに懸濁させ、 4°Cで 4時間撹拌しながら可溶性成分を溶出し、遠心分離によ り上清を回収した。残渣に対してさらに PBS 2Lをカ卩え、同様の溶出操作を行った後上 清を回収し、初回に得られた上清と合わせて、粗抽出画分とした。ついで、この粗抽出 画分に硫酸アンモニゥムを 80%飽和となるように加え、 4°Cに 2時間静置してタンパク質 成分を塩析させた。遠心分離により回収した塩析物を、 50mM Tris - HC1緩衝液 (pH7. 8)100mlに溶解し、 4°C下で同緩衝液 2Lに対する 4〜 12時間の透析を 2回行った後、予 め 50mM Tris - HC1緩衝液 (ρΗ7·8)で平衡化してお!/、た DEAE -セファロース CL6B (ファ ルマシア社製) 10mlに通塔した。さらに 50mM Tris - HC1緩衝液 (pH7,8)30mlを通液して 非吸着画分 130mlを回収した。 100 g of pollen collected from male cedar of Japanese cedar is suspended in 2 L of PBS (phosphate-buffered salin; hereinafter, referred to as PBS), and soluble components are eluted while stirring at 4 ° C for 4 hours, and centrifuged. The supernatant was recovered. The residue was further washed with 2 L of PBS, the same elution procedure was performed, and the supernatant was recovered and combined with the supernatant obtained the first time to obtain a crude extract fraction. Then, ammonium sulfate was added to the crude extract to 80% saturation, and the mixture was allowed to stand at 4 ° C for 2 hours to salt out protein components. The salted out product collected by centrifugation is dissolved in 100 ml of 50 mM Tris-HC1 buffer (pH 7.8) and dialyzed twice for 4 to 12 hours against 2 L of the same buffer at 4 ° C. After equilibration with 50 mM Tris-HC1 buffer (ρΗ7.8), the mixture was passed through 10 ml of DEAE-Sepharose CL6B (Pharmacia). Further, 30 ml of a 50 mM Tris-HC1 buffer (pH 7, 8) was passed, and 130 ml of the non-adsorbed fraction was collected.
この非吸着画分を、 4°C下で 200mM NaClを含む 10mM酢酸水溶液 (NaC卜酢酸水溶 液: ρΗ3·4)に対する 4〜12時間の透析を 2回行った後、予め NaC卜酢酸水溶液で平衡 化しておいた CM-セファロース CL6B (フアルマシア社製) 10mlに通塔した。 NaCト酢酸 水溶液 20mlで洗浄した後、 NaC卜酢酸水溶液および 1M NaClを含む 10mM酢酸水溶 液 (PH3.4)を用いて、 0〜1Mまでの連続した NaCl濃度勾配下で吸着成分を分離.溶出 させた。分取した各分画について、 280nmでの吸光度を測定するとともに、 SDS - PAGE の後ゲルをクマシ一染色してタンパク質成分を検出したところ、本発明のタンパク質は NaCl濃度 0.45M付近に回収されていた。本溶出画分は該タンパク質以外に Cry j 1、 未同定のタンパク質なども含まれてレ、た。  This non-adsorbed fraction was dialyzed twice at 4 ° C against a 10 mM acetic acid aqueous solution containing 200 mM NaCl (NaC triacetic acid aqueous solution: ρΗ3.4) for 4 to 12 hours, and then subjected to NaC triacetic acid aqueous solution in advance. The column was passed through 10 ml of equilibrated CM-Sepharose CL6B (Pharmacia). After washing with 20 ml of an aqueous solution of NaC-to-acetic acid, the adsorbed components are separated under a continuous NaCl concentration gradient from 0 to 1 M using an aqueous solution of NaC-to-acetic acid and an aqueous solution of 10 mM acetic acid (PH3.4) containing 1 M NaCl. I let it. The absorbance at 280 nm of each fraction was measured, and the gel was stained with SDS-PAGE to detect protein components.The protein of the present invention was recovered at a NaCl concentration of around 0.45 M. Was. This eluted fraction contained Cry j 1, unidentified protein and the like in addition to the protein.
次いで、この粗画分を限外ろ過膜セントリコン 10 (アミコン社製)を用いて遠心濃縮 後、予め PBSで平衡化してぉレ、たスーパーロース 12 (フアルマシア社製)に通塔してゲ ル濾過を行った。分子量 110,000 ± 15,000Daを極大とする画分を該タンパク質を含む 画分として回収した。本画分には該タンパク質の他未同定タンパク質 1種が含まれてい た。ゲル濾過を行う際の分子量マ一カーにはグルタメートデヒドロゲナーゼ (290,000D a)、ラクテ一トデヒドロゲナーゼ (140,000Da;)、エノラーゼ(67,000Da)、アデ二レートキナ —ゼ (32,000Da)、チトクローム c(12,400Da)を使用した。  Next, this crude fraction is concentrated by centrifugation using an ultrafiltration membrane Centricon 10 (manufactured by Amicon), equilibrated with PBS in advance, and passed through a column of Super Loose 12 (manufactured by Pharmacia) to gel. Filtration was performed. The fraction having a maximum molecular weight of 110,000 ± 15,000 Da was collected as a fraction containing the protein. This fraction contained the protein and one unidentified protein. The molecular weight markers for gel filtration include glutamate dehydrogenase (290,000 Da), lactate dehydrogenase (140,000 Da;), enolase (67,000 Da), adenylate kinase (32,000 Da), and cytochrome c. (12,400 Da) was used.
さらに該タンパク質を精製する目的で、バッチ法によるイオン交換クロマトを行った。 すなわち、ゲルろ過で回収した画分を、 200mM NaClを含む 10mM酢酸緩衝液 (pH5. 0)に対して 4〜12時間、 2回透析した後、ミクロ遠心管内で予め同緩衝液で平衡化して おいた CM-セファロース CL6B (フアルマシア社製) 50 μ 1と 30分間反応させた。反応 後、非吸着画分を該タンパクの精製標品として回収した。精製標品は必要に応じて ΡΒ Sでバッファー置換して、以下の試験に用いた。なお、該タンパク質の収量は、 BCAキ ット(ピアス社)でゥシ血清アルブミンに換算して求めたところ、原料に用いたスギ花粉 1 g当り 0.4 μ であった。  Further, for the purpose of purifying the protein, ion exchange chromatography was performed by a batch method. That is, the fraction collected by gel filtration was dialyzed twice against 10 mM acetate buffer (pH 5.0) containing 200 mM NaCl for 4 to 12 hours, and then equilibrated with the same buffer in a microcentrifuge tube in advance. The mixture was reacted with 50 μl of CM-Sepharose CL6B (Pharmacia) placed for 30 minutes. After the reaction, the non-adsorbed fraction was collected as a purified sample of the protein. The purified sample was used in the following tests after buffer replacement with ΡΒS as necessary. The yield of the protein was 0.4 μ / g of cedar pollen used as a raw material, as determined by converting it into serum albumin using a BCA kit (Pierce).
2.タンパク質の理化学的性質 上記 1で精製したタンパク質の理化学的性質を解析した。 2. Physicochemical properties of proteins The physicochemical properties of the protein purified in 1 above were analyzed.
(1 ) 分子量の同定  (1) Identification of molecular weight
Laemmliらの方法 (Nature, 227, 680-685, 1970)に準じて SDS- PAGEを行い、本発明 のタンパク質の分子量を決定した。結果を第 1図に示す。  SDS-PAGE was performed according to the method of Laemmli et al. (Nature, 227, 680-685, 1970) to determine the molecular weight of the protein of the present invention. Fig. 1 shows the results.
2—メルカプトエタノール添カロ、還元条件下で分子量 51 ,000土 5,000Daに相当する位 置に主要なバンドを検出した。また、非還元条件では分子量 48,000± 5,000Daに相当 する位置に主要なバンドを検出した。なお、 SDS-PAGEの分子量マ一カーにはミオシ ン (200,000 Da)、 ]3—ガラクトシダ一ゼ (116,300 Da)、フォスフオリラーゼ b(97,400Da)、 ゥシ血清アルブミン (66,300Da)、アルドラーゼ (42,400Da;)、カルボニックアンヒドラーゼ (30,000Da)、トリプシンインヒビター (20,100Da)、リゾチーム (14,400Da)を用いた。  A major band was detected at a position corresponding to a molecular weight of 51,000 soils and 5,000 Da under reducing conditions of 2-mercaptoethanol-added calo. Under non-reducing conditions, a major band was detected at a position corresponding to a molecular weight of 48,000 ± 5,000 Da. The molecular weight markers of SDS-PAGE include myosin (200,000 Da),] -galactosidase (116,300 Da), phosphorylase b (97,400 Da), and serum albumin (66,300 Da). Aldolase (42,400 Da;), carbonic anhydrase (30,000 Da), trypsin inhibitor (20,100 Da), and lysozyme (14,400 Da) were used.
(2) N末端アミノ酸配列の決定  (2) Determination of N-terminal amino acid sequence
精製タンパク質の N末端アミノ酸配列は、エドマン分解法を利用する自動プロテイン シーケンサー(島津、 PPSQ- 10)を使用し、タンパク質化学の常法により分析した。その 結果、 Xaa— Leu_Phe_Pro— Lys— Glu— Ala— Leu— Pro— Thr (ただし、 Xaa は、 Glu、 Asp、 His, Gly、 Arg、 Trpまたは Cysのいずれかを示す)で表わされる配列 が確認された。  The N-terminal amino acid sequence of the purified protein was analyzed by an ordinary protein chemistry method using an automated protein sequencer (Shimadzu, PPSQ-10) utilizing the Edman degradation method. As a result, a sequence represented by Xaa—Leu_Phe_Pro—Lys—Glu—Ala—Leu—Pro—Thr (where Xaa indicates any of Glu, Asp, His, Gly, Arg, Trp, or Cys) was confirmed. Was.
(3) 紫外吸収スペクトル  (3) UV absorption spectrum
分光光度計を用いて本発明のタンパク質の水溶液中での紫外吸収スペクトルを測 定したところ、波長 280nm付近に吸収極大を示した。  When an ultraviolet absorption spectrum of the protein of the present invention in an aqueous solution was measured using a spectrophotometer, it showed an absorption maximum near a wavelength of 280 nm.
(4) 生物作用: IgEへの結合性試験  (4) Biological effect: IgE binding test
本発明のタンパク質力スギ花粉症患者血清中の IgEに対する結合活性を有すること を、以下に示す IgE- ELISAにより解析した。なお、本実施例でのスギ花粉症患者は、フ アルマシア.アップジョン社製のキャップ RAST FEIAアレルゲン検査診断試薬での検 查により、スギアレルゲンに対して高反応性(+ 4グレード)を示す患者である。  The protein having the binding activity to IgE in the serum of patients with cedar pollinosis of the present invention was analyzed by the following IgE-ELISA. In this example, the cedar pollinosis patients were those who showed high reactivity (+4 grade) to sugya allergen by testing with a cap RAST FEIA allergen test diagnostic reagent manufactured by Pharmacia Upjohn. It is.
精製タンパク質を 3 g/mlとなるように PBSにて調製した後、 96ウェルマイク口プレート に 50 / 1/ゥェルずつ加えて 4°Cで 16時間静置し、プレートに固相化した。プレートの各 ゥエルを PBSで洗浄後、 l% (w/v) BSAを含む PBS (B-PBS)を 100 1/ゥエルずつ加え、 室温で 2時間静置した。各ゥエルを PBSで洗浄後、 B - PBSで 2倍に希釈したスギ花粉 症患者および健常人の血清を 50 μ 1/ゥエルずつ添加し、固相化タンパク質に対して 4°Cで 16時間反応させた。 0. 05% (v/v) Tween20を含む PBS (T- PBS)で洗浄後、 B- PBS で 1,000倍に希釈したビォチン標識ャギ抗ヒ HgE抗体(AmericanQualex、コード; A116 BN)を 50 μ 1/ゥエルずつ添加し、さらに室温で 1時間反応させた。ついで、 T - PBSで洗 浄後、 B-PBSで 4,000倍に希釈したアビジン D結合西洋ヮサビパーォキシダーゼ(VEC TOR社製、コード; A- 2004)を 50 / l/ゥエルずつ添加し、室温で 30分間静置した。 T - P BSで洗浄後、 0.1Mクェン酸緩衝液 (pH4.2)に 0.55mg/ml ABTS[2,2し Azino - bis(3-ethy l-benzothiazolin-6-sulfonic acid)]および 0.03%(v/v)過酸化水素を加えた発色基質液 を 100 ゥエルずつ添加し、室温で発色させた後、 5%SDS水溶液を 100 // 1/ゥエルず つ添加して反応を停止した。発色した基質の吸光度 (波長 415nm)を測定し、抗原タン パク質に対して結合した IgEを検出した。 The purified protein was adjusted to 3 g / ml in PBS, added to a 96-well mic opening plate at 50/1 / well, allowed to stand at 4 ° C for 16 hours, and immobilized on the plate. After washing each well of the plate with PBS, PBS (B-PBS) containing l% (w / v) BSA was added at 100 1 / well, and the plate was allowed to stand at room temperature for 2 hours. After washing each well with PBS, add 50 μl / well of cedar pollinosis patient and healthy human serum diluted 2-fold with B-PBS, and react with the immobilized protein at 4 ° C for 16 hours. I let it. After washing with PBS containing 0.05% (v / v) Tween20 (T-PBS), 50 μl of biotin-labeled goat anti-HgE antibody (AmericanQualex, code: A116 BN) diluted 1,000-fold with B-PBS was used. 1 / well was added thereto, and the reaction was further performed at room temperature for 1 hour. After washing with T-PBS, avidin D-conjugated horseradish peroxidase (VEC) diluted 4,000-fold with B-PBS TOR Co., Code; A-2004) was added at 50 / l / well, and left at room temperature for 30 minutes. After washing with T-PBS, 0.5Mmg / ml ABTS [2,2 Azino-bis (3-ethyl-benzothiazolin-6-sulfonic acid)] and 0.03% in 0.1M citrate buffer (pH4.2) (v / v) The coloring substrate solution to which hydrogen peroxide was added was added in 100 µl each, and the color was developed at room temperature. After that, the reaction was stopped by adding a 5% aqueous SDS solution at 100 // 1/1 / well. The absorbance (wavelength: 415 nm) of the developed substrate was measured to detect IgE bound to the antigen protein.
(5) 溶剤への溶解性  (5) Solubility in solvents
本発明のタンパク質の各種溶媒に対する溶解性を調べた。水、生理的食塩液およ びリン酸緩衝液には可溶性を示した。  The solubility of the protein of the present invention in various solvents was examined. It was soluble in water, saline and phosphate buffer.
(6) 安定性  (6) Stability
(5)で溶解した水溶液を、 4°C冷蔵庫で保存した。 2ヶ月後、該水溶液を用いて、 (4) の実験を行ったところ、 2ヶ月前と同様の結果が得られた。  The aqueous solution dissolved in (5) was stored in a refrigerator at 4 ° C. Two months later, when the experiment (4) was performed using the aqueous solution, the same result as that obtained two months ago was obtained.
以上のような理化学的性質を有するタンパク質は現在までに知られておらず、新規 物質であると判断された。  The protein having the above physicochemical properties was not known until now, and was determined to be a novel substance.
3.アレルゲン活性の解析および診断  3. Analysis and diagnosis of allergen activity
本発明によるタンパク質の診断および治療における有用性を示すことを目的に、スギ 花粉症患者血清中の特異的 IgEの抗体価を指標とする、以下の試験(IgE- ELISA)を 行った。比較の対照として、既知アレルゲンである Cry j 1 、 Cry j 2および未精製総 タンパク質画分(crude extract : CE)を用いて同様の試験を行うとともに、既存の医療 用体外診断法による試験結果との比較を行った。  In order to demonstrate the utility of the protein according to the present invention in diagnosis and treatment, the following test (IgE-ELISA) was performed using the antibody titer of specific IgE in the serum of cedar pollinosis patients as an index. As a control for comparison, a similar test was conducted using known allergens Cry j 1 and Cry j 2 and crude protein fraction (crude extract: CE). Was compared.
Cry j 1 およびし ry j 2ί 、 Yasuedaら(journal of Allergy and し linical Immunolog y, Vol.71, 77- 86, 1983)および Sakaguchiら(Allergy Vol.45, 309-312, 1990)の方法 に準じて精製した後、最終的に予め PBSで平衡化してぉレ、たスーパ一ロース 12 (ファ ルマシア社製)によるゲルろ過により精製し、以下の試験に使用した。なお、各精製画 分は、 SDS- PAGEおよび N末端アミノ酸配列の解析より目的の単一タンパク質であるこ とを確認した。また、未精製総タンパク質画分には、前述の 1で示した粗抽出画分より 硫酸アンモニゥム 80%飽和により得られる塩析物を PBSに溶解、透析後、 IgE- ELISAに 使用した。 IgE- EL1SAについて具体的に示す。  Cry j 1 and ry j 2ί, according to the methods of Yasueda et al. (Journal of Allergy and shi clinical Immunology, Vol. 71, 77-86, 1983) and Sakaguchi et al. (Allergy Vol. 45, 309-312, 1990). After purification, the mixture was finally equilibrated with PBS in advance, and purified by gel filtration using Super-Rose 12 (manufactured by Pharmacia) and used in the following tests. Each purified fraction was confirmed to be the target single protein by SDS-PAGE and N-terminal amino acid sequence analysis. For the unpurified total protein fraction, a salted-out product obtained by saturation of ammonium sulfate 80% from the crude extract fraction described in 1 above was dissolved in PBS, dialyzed, and used for IgE-ELISA. IgE-EL1SA will be specifically described.
該タンパク質、 Cry j 1 および Cry j 2の精製品は 3 μ g/mlとなるように、 CEは 30 /i g /mlとなるように、 PBSでそれぞれ調製した後、それぞれ 96ウェルマイク口プレートに 50 ゥエルずつ加え、 4°Cで 16時間静置してプレートに固相化した。各ゥエルを PBSで 洗浄した後、 B- PBSを 100 /i 1/ゥエルずつ加え、室温で 2時間静置した。再び、各ゥェ ルを PBSで洗浄後、 B-PBSで 2倍に希釈したスギ花粉症患者の血清を 50 μ 1/ゥエルず つ添加し、 4°Cで 16時間反応させた。反応後、 T- PBSで洗浄した後、 B-PBSで 1,000倍 に希釈したピオチン標識ャギ抗ヒ HgE抗体(AmericanQualex、コード; A116BN)を 50 μ 1/ゥエルずつ添加し、さらに室温で 1時間静置した。ついで、 Τ - PBSで洗浄した後、 Β-Ρ BSで 4,000倍に希釈したアビジン D結合西洋ヮサビパーォキシダーゼ (VECTOR社 製、コード; A- 2004)を 50 μ 1/ゥエルずつ添加し、室温で 30分間静置した。 T- PBSで洗 浄後、 ABTS発色基質液を 100 μ 1/ゥエルずつ添加し室温で反応させ、発色後、 5%SDS 水溶液を 100 ゥエルずつ添加して反応を停止した。発色した基質の吸光度を波長 415匪にて測定し、抗原タンパク質に特異的な IgEの抗体価の指標とした。 The purified proteins Cry j 1 and Cry j 2 were prepared in PBS at 3 μg / ml and CE at 30 / ig / ml in PBS. The solution was added in 50-well portions, and allowed to stand at 4 ° C for 16 hours to immobilize on a plate. After each well was washed with PBS, B-PBS was added at 100 / i1 / well and left at room temperature for 2 hours. After washing each well with PBS again, 50 μl / well of serum of a Japanese cedar pollinosis patient diluted 2-fold with B-PBS was used. And reacted at 4 ° C. for 16 hours. After the reaction, after washing with T-PBS, add a biotin-labeled goat anti-human HgE antibody (AmericanQualex, code: A116BN) diluted 1,000-fold with B-PBS in 50 μl / ゥ l volumes, and further at room temperature for 1 hour. It was left still. Then, after washing with Τ-PBS, avidin D-conjugated horseradish peroxidase (VECTOR, code: A-2004) diluted 4,000-fold with Β-ΡBS was added at 50 μl / ゥ l. It was left at room temperature for 30 minutes. After washing with T-PBS, the ABTS chromogenic substrate solution was added at 100 μl / ゥ l at room temperature and reacted at room temperature. After color development, the reaction was stopped by adding a 5% SDS aqueous solution at 100 ゥ l / well. The absorbance of the developed substrate was measured at a wavelength of 415 wavelengths and used as an index of the antibody titer of IgE specific to the antigen protein.
スギ花粉症の診断に使用される医療用診断薬は、いずれの場合もスギ花粉より抽出 した総タンパク質画分を抗原として使用し、スギ花粉アレルゲンに特異的な血清中 IgE の抗体価を測定するものである。本実施例では、この種の診断法として一般的に使用 されている、フアルマシア'アップジョン社製のキャップ RAST FEIA (スギアレルゲン結 合キャップ;コード No.CT17、酵素標識抗体試薬、レファレンス試薬により構成)を対照 例に用いた。キャップ RAST FEIAはフアルマシア'アップジョン社推奨の方法により 行われた。すなわち、予め総スギ花粉アレルゲン (未精製総タンパク画分)を固相化し たキャップに対して検体血清を反応させ、洗浄後、酵素標識抗 IgE抗体を反応させた。 さらに洗浄後、基質液を加えて生成した蛍光物質を蛍光光度計により検出し、アレル ゲンに特異的に結合した IgEを定量した。  In any case, the diagnostic reagent used for the diagnosis of cedar pollinosis uses the total protein fraction extracted from cedar pollen as an antigen and measures the antibody titer of serum IgE specific to cedar pollen allergen Things. In this example, a cap RAST FEIA manufactured by Pharmacia's Upjohn Co., Ltd. (a spear allergen binding cap; code No.CT17, an enzyme-labeled antibody reagent, and a reference reagent), which is generally used as this kind of diagnostic method, is used. ) Was used as a control. Cap RAST FEIA was performed according to the method recommended by Pharmacia's Upjohn. That is, the sample serum was reacted with the cap on which the total cedar pollen allergen (unpurified total protein fraction) had been previously immobilized, washed, and then reacted with an enzyme-labeled anti-IgE antibody. After further washing, the fluorescent substance generated by adding the substrate solution was detected by a fluorometer, and IgE specifically bound to the allergen was quantified.
IgE- ELISAの結果を第 2図に示す。いずれの患者血清においても、該タンパク質に 対する特異 IgE抗体が検出され、該タンパク質力スギ花粉症におけるアレルゲンである ことが示された。一方、健常者では該タンパク質に対する特異 IgE抗体は検出されなか つた。  Fig. 2 shows the results of IgE-ELISA. In all patient sera, a specific IgE antibody against the protein was detected, indicating that the protein was an allergen in cedar pollinosis. On the other hand, no specific IgE antibody against the protein was detected in healthy subjects.
スギ花粉症患者 12例において、該タンパク質に対する特異 IgE抗体価(吸光度)を C ry j 2特異 IgE抗体価と比較すると、ほぼ全例で該タンパク質に対する特異 IgE抗体価 のほうが高値を示した。このことは、該タンパク質は Cry j 2より強いアレルゲンであるこ とを意味する。また、該タンパクと Cry j 1の場合を比較すると、患者 12例中 9例で該タ ンパクに対する特異 IgE抗体価が高値を示した。さらに患者検体数を増やし、該タンパ ク質と Cry j 1のアレルゲン活性について詳細な解析を行った。その結果を第 1表に示 した。 第 1 表 検体 特異 IgE抗体価, OD 比 In 12 cases of cedar pollinosis patients, when the specific IgE antibody titer (absorbance) against the protein was compared with the Cryj2-specific IgE antibody titer, almost all the cases showed higher specific IgE antibody titers against the protein. This means that the protein is a stronger allergen than Cry j2. Further, when comparing the case of the protein with that of Cry j 1, 9 out of 12 patients showed a high value of the specific IgE antibody against the protein in 9 cases. Further, the number of patient samples was further increased, and the protein and Cry j 1 allergen activity were analyzed in detail. The results are shown in Table 1. Table 1 Sample-specific IgE antibody titer and OD ratio
R AS T^ 患者 御 ew* 抗 Cry j 1 (iiNew/ 抗 R AS T ^ Patients ew * Anti-Cry j 1 (iiNew /
Ciy j 1) Ciy j 1)
2+ 9705 0.06 0.04 1.7 2+ 9705 0.06 0.04 1.7
9706 0.15 0.16 0.9 9706 0.15 0.16 0.9
9707 0.49 0.32 1.59707 0.49 0.32 1.5
9810 0.07 0.06 1.39810 0.07 0.06 1.3
9813 0.14 0.08 1.79813 0.14 0.08 1.7
9814 0.65 0.39 1.79814 0.65 0.39 1.7
9815 0.24 0.26 0.99815 0.24 0.26 0.9
9816 0.38 0.61 0.69816 0.38 0.61 0.6
9817 0.30 , 0.24 1.29817 0.30, 0.24 1.2
3+ 9708 0.40 0.31 1.3 3+ 9708 0.40 0.31 1.3
9709 1.30 1.20 1.1 9709 1.30 1.20 1.1
9710 0.63 0.43 1.59710 0.63 0.43 1.5
9711 0.80 0.83 1.09711 0.80 0.83 1.0
9818 0.27 0.37 0.79818 0.27 0.37 0.7
9819 0.47 0.39 1.29819 0.47 0.39 1.2
9820 1.00 0.73 1.49820 1.00 0.73 1.4
9821 0.38 0.46 0.89821 0.38 0.46 0.8
9822 0.51 0.45 1.19822 0.51 0.45 1.1
4+ 9713 0.86 0.95 0.9 4+ 9713 0.86 0.95 0.9
9714 1.64 1.27 1.3 9714 1.64 1.27 1.3
9715 1.63 1.40 1.29715 1.63 1.40 1.2
9716 2.06 1.88 1.19716 2.06 1.88 1.1
9825 0.80 0.76 1.19825 0.80 0.76 1.1
9826 0.58 0.75 0.89826 0.58 0.75 0.8
* 抗 Newとは、 本発明のタンパク質に る抗体を示す。 該タンパク質に対する特異 IgE抗体価を Cry j 1に対する特異 IgE抗体価で除した比 (該タンパク ZCry j 1)で比較すると、患者 24例中、 9例で 1 · 3以上を示した。また、 1. 1以上の場合を含めると 16例であった。一方、 0· 9以下は 4例のみであった。すな わち、該タンパクは 3種の精製標品のうちで最も強いアレルゲン活性を示し、診断、治 療における抗原としての有用性が示された。さらに、キャップ RAST FEIAによるスギ 花粉.アレルゲン特異的 IgE抗体価と、 IgE- ELISAによる各精製タンパク質および未精製 総タンパク質画分(CE)に対する特異的 IgE抗体価の相関にっレ、て検討した。スギ花 粉症患者 12名および健常人 4名について解析した 2変数散布図を第 3図に示した。キ ヤップ RAST FEIAによるスギアレルゲン特異 IgE抗体価と、 IgE- ELISAによる本発明の タンパク質に特異的な IgE抗体価は高い相関性を示した (相関係数 r=0.932)。従って、 該タンパクは総スギ花アレルゲンの IgE結合活性を極めて良く反映している。 Cry j 1* Anti-New indicates an antibody against the protein of the present invention. A comparison of the ratio obtained by dividing the specific IgE antibody titer against the protein by the specific IgE antibody titer against Cry j 1 (the protein ZCry j 1) showed that 9 out of 24 patients showed 1.3 or more. In addition, there were 16 cases including cases of 1.1 or more. On the other hand, there were only 4 cases with a value of 0.9 or less. That is, the protein showed the strongest allergen activity among the three purified preparations, indicating its usefulness as an antigen in diagnosis and therapy. In addition, the correlation between the IgE antibody titer specific to cedar pollen and allergen by RAST FEIA and the IgE antibody titer specific to each purified protein and unpurified total protein fraction (CE) by IgE-ELISA was examined. Fig. 3 shows a two-variable scatter plot of 12 cedar pollinosis patients and 4 healthy subjects. There was a high correlation between the titer of IgE antibody specific to Sgia allergen by RAST FEIA and the titer of IgE antibody specific to the protein of the present invention by IgE-ELISA (correlation coefficient r = 0.932). Therefore, the protein reflects the IgE binding activity of total cedar flower allergen very well. Cry j 1
、 Cry j 2に特異的な lgE抗体価とキャップ RAST FEIAの結果との相関係数はそれ ぞれ 0.938、 0.401であった。 , The correlation coefficient between the results of specific l g E antibody titers and the cap RAST FEIA to Cry j 2 are respectively was 0.938, was 0.401.
キャップ RAST FEIAと IgE-ELISAでは、抗原の固相化法、検出方法および該検 出に用いる酵素/基質は異なるが、キャップ RAST FEIAによる特異的 IgE抗体価と、 Ig E-EL1SAでの未精製総タンパク質特異的 IgE抗体価との相関係数は 0.921を示してい る。すなわち、今回行った特異 IgE抗体価の評価は測定系の違いにより影響されてい なレ、。  Cap RAST FEIA and IgE-ELISA use different antigen immobilization methods, detection methods, and enzymes / substrates for detection, but specific IgE antibody titers with cap RAST FEIA and unpurified IgE-EL1SA The correlation coefficient with the total protein-specific IgE antibody titer is 0.921. In other words, the evaluation of the specific IgE antibody titer performed this time was not affected by differences in the measurement system.
以上のことから、該タンパク質に特異的な IgEを定性的および定量的に分析すること は、スギ花粉症の診断として有用であり、該タンパク質はスギ花粉症を診断するための 抗原として使用すること力 Sできる。  From the above, qualitative and quantitative analysis of IgE specific to the protein is useful as a diagnosis of cedar pollinosis, and the protein should be used as an antigen for diagnosing cedar pollinosis. Power S can.
さらに、該タンパク質のスギ花粉総タンパク質に占める割合が極めて低いにも関わら ず、スギ花粉症患者血清中の該タンパク質に対する特異 IgE抗体価は、 Cry j 1、 Cry j 2に対する特異 IgE抗体価と同等以上に高かった。これほどまでに高いアレルゲン 活性を示す単一のタンパク質は未だ知られていなレ、。従って、該タンパク質は極めて 高い抗原性を有し、スギ花粉症の発症において極めて重要な原因物質となっていると いえる。  Furthermore, the specific IgE antibody titer against the protein in the serum of cedar pollinosis patients is equivalent to the specific IgE antibody titer against Cry j1 and Cry j2, even though the ratio of the protein to the total protein of cedar pollen is extremely low. It was more expensive. A single protein exhibiting such high allergen activity is not yet known. Therefore, it can be said that the protein has extremely high antigenicity and is a very important causative substance in the onset of cedar pollinosis.
4.花粉抗原に対する抗体の作製  4. Production of antibodies against pollen antigen
(1)免疫原の調製 (1) Preparation of immunogen
上述 2 (2)で得られた配列番号 1に示した N末端アミノ酸配列の C末端側の Thrに、キ ャリアタンパク質との結合のために Cysを付加したペプチド(配列番号 2)についてぺプ チド合成を行った。ペプチド合成には、他種品目同時固相法自動ペプチド合成装置 P SSM-8 (島津製作所社製)を用レ、た。免疫原性を高める目的で、合成ペプチドと KLH (Keyhole Lympet Hemocyanin ;キーホーノレリンペットへモシァニン、カノレビオケム社 製)とのコンジュゲートを作製し、免疫原とした。すなわち、 KLHを PBSに溶解して 10mg /mlに調整し、 KLH溶液の 1/10容量の 25mg/ml MBS[N-(maleimidobenzoyloxy)succini mide、ナカライテスク社製]を滴下して 30分間撹拌反応させた。あらかじめ PBSで平衡 化したゲルろ過カラムであるセフアデックス G-25カラム(フアルマシア社製)でフリーの MBSを除き、 KLH-MBSコンジュゲート 2.5mgを得た。さらに、 0.1Mリン酸ナトリウムバッ ファ一(PH7.0)に溶解したペプチド lmgと混合し、室温で 3時間、攪拌反応した。反応 後、 PBS- 0.5MN&C1で透析したものを抗原ペプチド一 KLHコンジュゲートとして得、これ を免疫原として用いた。 A peptide (SEQ ID NO: 2) with Cys added to the C-terminal Thr of the N-terminal amino acid sequence shown in SEQ ID NO: 1 obtained in 2 (2) above for binding to a carrier protein (SEQ ID NO: 2) Tide synthesis was performed. For peptide synthesis, an automated peptide synthesizer P SSM-8 (manufactured by Shimadzu Corporation) was used for simultaneous solid-phase method synthesis of other types of items. Synthetic peptides and KLH to enhance immunogenicity (Keyhole Lympet Hemocyanin; conjugate with Keyhole Lympet Hemocyanin; manufactured by Canolebiochem Co., Ltd.) was used as an immunogen. That is, KLH is dissolved in PBS to adjust to 10 mg / ml, 25 mg / ml MBS [N- (maleimidobenzoyloxy) succini mide, manufactured by Nacalai Tesque, Inc.] of 1/10 volume of the KLH solution is added dropwise and stirred for 30 minutes. I let it. Free MBS was removed using a Sephadex G-25 column (Pharmacia), a gel filtration column equilibrated with PBS in advance, to obtain 2.5 mg of a KLH-MBS conjugate. Furthermore, it was mixed with 1 mg of peptide dissolved in 0.1 M sodium phosphate buffer (PH 7.0), and reacted by stirring at room temperature for 3 hours. After the reaction, what was dialyzed against PBS-0.5 MN & C1 was obtained as an antigen peptide-KLH conjugate, which was used as an immunogen.
(2)動物の免疫と抗体産生細胞の調製  (2) Animal immunization and preparation of antibody-producing cells
上述の 4 (1)に示した方法により調製した抗原ペプチド— KLHコンジユゲー M00 / g を水酸ィ匕ァノレミニゥムアジュノ ント( Antibodies ― A Laboratory Manual, Cold Sprin g Harbor Laboratory, P99, 1988) 2mgおよび百日咳ワクチン(千葉県血清研究所製) 1 X 109 細胞とともに 5週令雌マウス(Balb/c)に投与し、 2週間後より 100 // gの抗原ぺ プチドー KLHコンジュゲートを 1週間に 1回、計 4回投与した。該マウスの眼底静脈叢よ り採血し、その血清抗体価を 4 (3)に述べる酵素免疫測定法で調べた。最終免疫 3日 後、血清抗体価が充分高かったマウスより脾臓を摘出した。 Above 4 (1) antigen was prepared by the method shown in the peptide - KLH Konjiyuge M00 / g to Mizusani匕§ Honoré mini © Moore Ho cement (Antibodies - A Laboratory Manual, Cold Sprin g Harbor Laboratory, P 99, 1988 ) 2 mg and pertussis vaccine (manufactured by Chiba Prefectural Serum Research Institute) were administered to 5-week-old female mice (Balb / c) together with 1 × 10 9 cells, and after 2 weeks, 100 // g of antigen ぺ peptide KLH conjugate was administered. It was administered once a week for a total of four doses. Blood was collected from the fundus venous plexus of the mouse, and its serum antibody titer was examined by the enzyme immunoassay described in 4 (3). Three days after the final immunization, spleens were removed from mice with sufficiently high serum antibody titers.
脾臓を MEM (Minimum Essential Medium)培地(日水製薬社製)中で細断し、ピン セットでほぐし、遠心分離( OO rpm、 5分間)した。得られた沈殿画分にトリス—塩化 アンモニゥム緩衝液 (PH7.6)を添加し、:!〜 2分間処理することにより赤血球を除去し た。得られた沈殿画分(細胞画分)を MEM培地で 3回洗浄し、細胞融合に用いた。 また、脾臓摘出時に全血液を採取し、室温で 2時間静置後得られる血清はペプチド特 異的抗血清であり、該抗血清は上述の 1で得られた本発明のタンパク質に特異的な抗 血清であった。 The spleen was shredded in MEM (Minimum Essential Medium) medium (manufactured by Nissui Pharmaceutical Co., Ltd.), loosened with forceps, and centrifuged (OO rpm, 5 minutes). ! Resulting tris precipitate fraction - was added Anmoniumu buffer chloride solution (P H7.6),: erythrocytes were removed by treating 1-2 minutes. The resulting precipitate fraction (cell fraction) was washed three times with MEM medium and used for cell fusion. In addition, whole blood is collected at the time of spleen extraction, and the serum obtained after standing at room temperature for 2 hours is a peptide-specific antiserum, and the antiserum is specific for the protein of the present invention obtained in the above 1). It was an antiserum.
(3)酵素免疫測定法  (3) Enzyme immunoassay
配列番号 1に示したアミノ酸配列を有するペプチドに特異的に反応する抗体のみを 検出する目的で、合成ペプチドと THY (Thyroglobulin ;サイログロブリン、シグマ社)の コンジュゲートを作製し、アツセィ用の抗原として用いた。すなわち、 THYを PBSに溶解 して lOmg/mlに調整し、 THY溶液の 1/10容量の 25mg/ml SMCC [4-(N-maleimidometh yl)- cyclohexane- 1- carboxylic acid N-hydroxysuccinimiae ester、シグマ社 ¾¾¾r滴 して 30分間撹拌反応させた cあらかじめ PBSで平衡化したゲルろ過カラムであるセファ デックス G- 25カラム(フアルマシア社製)でフリーの SMCCを除き、 THY-SMCCコンジュ ゲート 2.5mgを得た。さらに、 0.1Mリン酸ナトリウムバッファ一(PH7.0)に溶解した該ぺプ チド lmgと混合し、室温で 3時間、攪拌反応した。反応後、 PBS- 0.5MNaClで透析したも のを抗原ペプチド一 THYコンジュゲートとして得、これを抗原として用いた。対照抗原 としては、配列番号 3に示すアミノ酸配列を有する対照ペプチドを前述と同様の方法 で THYと架橋したコンジュゲートを用いた。 A conjugate of a synthetic peptide and THY (Thyroglobulin; thyroglobulin, Sigma) was prepared and used as an antigen for Atsei to detect only antibodies that specifically react with the peptide having the amino acid sequence shown in SEQ ID NO: 1. Was. That is, THY is dissolved in PBS to adjust to lOmg / ml, and 1/10 volume of 25 mg / ml SMCC [4- (N-maleimidomethyl) -cyclohexane-1-carboxylic acid N-hydroxysuccinimiae ester, Sigma C. The reaction was stirred for 30 minutes after dropping. C ) Sephadex G-25 column (Pharmacia), a gel filtration column pre-equilibrated with PBS, was used to remove free SMCC to obtain 2.5 mg of THY-SMCC conjugate. Was. Further, the capsule dissolved in 0.1 M sodium phosphate buffer (PH7.0) After mixing with 1 mg of tide, the mixture was stirred and reacted at room temperature for 3 hours. After the reaction, a solution dialyzed against PBS-0.5M NaCl was obtained as an antigen peptide-THY conjugate, which was used as an antigen. As a control antigen, a conjugate obtained by crosslinking a control peptide having the amino acid sequence shown in SEQ ID NO: 3 with THY in the same manner as described above was used.
ゥエルの EIA用プレート (グライナ一社製)に、 5 μ g/mlで調製した上述のコンジュゲ ートを 50 /Z 1/ゥエルで分注し、 4°Cでー晚放置して吸着させた。 PBSで洗浄後、残って いる活性基をブロックするために、 l%(w/v)BSAを含む PBS(B- PBS)を 100 μ 1/ゥエルで 加え、室温 1時間反応させた。 B-PBSを捨て、被免疫マウス抗血清、ハイプリドーマ培 養上清もしくは精製モノクローナル抗体を 50 μ 1/ゥエルで分注し 2時間反応させた。 0.0 5%(v/v)Tween20を含む PBS(T- PBS)で洗浄後、ペルォキシダーゼ標識ゥサギ抗マウス ィムノグロブリン (ダコ社製)を 50 μ 1/ゥエルで加えて室温、 1時間反応させた。 T- PBSで 洗浄後、 ABTS発色基質液を用いて発色させ波長 415nmの吸光度(Emax、和光純薬 工業社製)を測定した。  The above conjugate prepared at 5 μg / ml was dispensed at 50 / Z 1 / well onto an EIA plate (Grainer Co., Ltd.) and left at 4 ° C for adsorption. . After washing with PBS, PBS containing l% (w / v) BSA (B-PBS) was added at 100 μl / well to block the remaining active groups, and the mixture was reacted at room temperature for 1 hour. The B-PBS was discarded, and the antiserum of the immunized mouse, the supernatant of the hybridoma culture or the purified monoclonal antibody was dispensed at 50 μl / well and allowed to react for 2 hours. After washing with 0.05% (v / v) Tween 20 in PBS (T-PBS), peroxidase-labeled ゥ sagi anti-mouse immunoglobulin (manufactured by Dako) was added at 50 μl / ゥ and reacted at room temperature for 1 hour. . After washing with T-PBS, color was developed using an ABTS chromogenic substrate solution, and the absorbance at a wavelength of 415 nm (Emax, manufactured by Wako Pure Chemical Industries, Ltd.) was measured.
(4)マウス骨髄腫細胞の調製  (4) Preparation of mouse myeloma cells
8-ァザグァニン耐性マウス骨髄腫細胞株 P3X63Ag8U.l (P3- U1: ATCCより購入)を 正常培地(10%ゥシ胎児血清添加 RPMI培地)で培養し、細胞融合時に 2 X 107個以上 の細胞を確保し、親株として細胞融合に供した。 8-azaguanine-resistant mouse myeloma cell line P3X63Ag8U.l (P3-U1: purchased from ATCC) was cultured in normal medium (RPI medium supplemented with 10% fetal calf serum), and 2 x 10 7 or more cells were fused at the time of cell fusion. And used for cell fusion as a parent strain.
(5)ハイプリドーマの作製  (5) Preparation of hybridoma
4 (2)で得られたマウス脾細胞と 4 (4)で得られた骨髄腫細胞とを 10 : 1になるよう混 合し、遠心分離(l,200rpm、 5分間)した。得られた沈澱画分の細胞群を良くほぐした 後、攪拌しながら、 37°Cで、ポリエチレングリコール一 1000 (PEG- 1000) 2g、 MEM培地 2mlおよびジメチルスルホキシド 0.7mlの混液を 108個のマウス脾細胞あたり 0.5ml加え た。さらに、該懸濁液に 1〜2分間毎に MEM培地 lmlを数回加えた後、 MEM培地を 加えて全量が 50mlになるようにした。 The mouse spleen cells obtained in 4 (2) and the myeloma cells obtained in 4 (4) were mixed at a ratio of 10: 1, and centrifuged (1, 200 rpm, 5 minutes). Was loosened well resulting precipitated fraction of cell groups, while stirring, at 37 ° C, polyethylene glycol one 1000 (PEG- 1000) 2g, mixture 10 8 of MEM medium 2ml and dimethyl sulfoxide 0.7ml 0.5 ml was added per mouse splenocytes. Further, lml of MEM medium was added several times to the suspension every 1 to 2 minutes, and then MEM medium was added so that the total volume became 50ml.
該懸濁液を遠心分離(1 ,200rpm、 5分間)して得られた沈澱画分の細胞をタッピング により緩やかにほぐした後、緩やかにピペッティングをしつつ、 100 mlの HAT培地〔1 0%ゥシ胎児血清添加 RPMI培地に HAT Media Supplement (ベーリンガーマンノヽィム 社製)を加えた培地〕中に懸濁した。該懸濁液を 96穴培養用プレートに 200 mlZゥェ ルずつ分注し、 5%C02インキュベータ一中で 37°Cで 10〜 14日間培養した。 After centrifuging the suspension (1,200 rpm, 5 minutes), the cells in the precipitate fraction obtained are gently loosened by tapping, and 100 ml of HAT medium [10. % Hat fetal serum-supplemented RPMI medium supplemented with HAT Media Supplement (Boehringer Mannheim). The suspension was dispensed into a 96-well culture plate at a volume of 200 ml / well and cultured at 37 ° C. for 10 to 14 days in a 5% CO 2 incubator.
培養後、培養上清を 4 (3)に記載した酵素免疫測定法で調べ、抗原ペプチドに反応 して対照ペプチドに反応しないゥエルを選んだ。さらに、そのゥエルに含まれる細胞か ら限界希釈法によるクロ一ニングを 2回繰り返し、本発明のタンパク質の部分配列を認 識するモノクローナル抗体産生ハイブリドーマ KM2715を取得した。該モノクローナル 抗体 KM2715の抗原ペプチド及び対照ペプチドに対する反応性を酵素免疫測定法に より解析した結果を第 4図に示した。 After the culture, the culture supernatant was examined by the enzyme immunoassay described in 4 (3), and cells that reacted with the antigen peptide but did not react with the control peptide were selected. Further, the cells contained in the well were repeatedly cloned twice by the limiting dilution method to obtain a monoclonal antibody-producing hybridoma KM2715 recognizing a partial sequence of the protein of the present invention. The monoclonal The results of analyzing the reactivity of the antibody KM2715 with the antigen peptide and the control peptide by an enzyme immunoassay are shown in FIG.
第 4図に示すように、 該モノクローナル抗体 KM2715は本発明のタンパク質の部分 配列からなる抗原ペプチドに特異的な反応性を示した。  As shown in FIG. 4, the monoclonal antibody KM2715 showed specific reactivity to an antigen peptide consisting of a partial sequence of the protein of the present invention.
(6)モノクローナル抗体の精製 (6) Purification of monoclonal antibody
モノクローナル抗体はマウス腹水より精製した。プリスタン処理した 8週令ヌード雌マ ウス(BALB/c)に 4 (5)で得られたノ、イブリドーマ株を 5〜20 X 106細胞/匹それぞれ腹 腔內注射した。 10〜21日後、ノ、イブリドーマが腹水癌化したマウスから腹水を採取(1 〜8 mlZ匹)した。該腹水を遠心分離(l,200 X g、 5分間)して固形分を除去した後、 カプリノレ酸沈殿法 [Antibodies - A Laboratory Manual, Cold Spring Harbor Labor atory, 1988〕によりモノクローナル抗体を精製した。該モノクローナル抗体のサブクラス はサブクラスタイピングキットを用いた ELISA法により IgGlと決定された。 Monoclonal antibodies were purified from mouse ascites. An 8-week-old nude female mouse (BALB / c) treated with pristane was intraperitoneally injected with 5 to 20 × 10 6 cells / animal of the hybridoma and the hybridoma strain obtained in 4 (5). 10 to 21 days later, ascites was collected from mice in which the ascitic carcinoma of the hybridoma was changed (1 to 8 mlZ mice). After the ascites was centrifuged (1, 200 × g, 5 minutes) to remove solids, the monoclonal antibody was purified by the caprinoleic acid precipitation method [Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988]. The subclass of the monoclonal antibody was determined to be IgGl by ELISA using a subcluster typing kit.
5.モノクローナル抗体 ΚΜ271δによる花粉アレルゲンエキス中の本発明のタンパクの 検出(ウェスタンブロッテイング) 5. Detection of the protein of the present invention in pollen allergen extract by monoclonal antibody ΚΜ271δ (Western blotting)
3で使用した未精製総タンパク質画分(CE)を抗原としてウェスタンブロッテイングを 行レ、、モノクローナル抗体 KM2715の花粉アレルゲンエキス中の本発明のタンパク質 の検出、定量系への応用を検討した。  Western blotting was performed using the unpurified total protein fraction (CE) used in step 3 as an antigen, and the detection of the protein of the present invention in the pollen allergen extract of the monoclonal antibody KM2715 and its application to a quantitative system were examined.
CE2 μ g/レーンを SDS- PAGE (5- 20%グラジェントゲル、アト一社製) [Antibodies - A Laboratory Manual, Cold Spring Harbor Laboratory, 1988〕にて分画した後、 PV DF膜 (ミリポア社製)にブロッテイングした。該膜を 1 % BSA- PBSでブロッキング後、モ ノクローナル抗体 KM2715の培養上清を添加し、室温で 2時間放置した。該膜を T - P BSでよく洗浄した後、第 2抗体として 1000倍希釈したペルォキシダーゼ標識ゥサギ抗 マウスィムノグロブリン抗体 (ダコ社製)を添加し、室温で 1時間放置した。  After fractionating 2 μg / CE of lane by SDS-PAGE (5-20% gradient gel, manufactured by Atto) [Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory, 1988], PV DF membrane (Millipore) Was made. After blocking the membrane with 1% BSA-PBS, the culture supernatant of the monoclonal antibody KM2715 was added and left at room temperature for 2 hours. After thoroughly washing the membrane with T-PBS, a 1000-fold diluted peroxidase-labeled rabbit herb anti-mouse immunoglobulin antibody (manufactured by Dako) was added as a second antibody, and left at room temperature for 1 hour.
該膜を T- PBSでよく洗浄した後、 ECL kit (アマシャムフアルマシアバイオテク社製)を 用いて検出した。  After thoroughly washing the membrane with T-PBS, detection was carried out using an ECL kit (Amersham Pharmacia Biotech).
第 5図に示すように、該モノクローナル抗体 KM2715は、 CE中の当該タンパク質の 分子量に相当する 51kDa付近のバンドに特異的に反応した。一方、対照抗体として 用いた IgGlタイプで、 G- CSF改変体に対するモノクローナル抗体である KM511 (特開 平 8—165300)ではこのような反応は認められなかった。このことは、該モノクローナ ル抗体は花粉アレルゲンエキス中の本発明のタンパク質を特異的に検出、定量するこ とが可能であることを示し、その有用性が確認された。  As shown in FIG. 5, the monoclonal antibody KM2715 specifically reacted with a band around 51 kDa corresponding to the molecular weight of the protein in CE. On the other hand, no such reaction was observed with KM511 (Japanese Unexamined Patent Publication No. 8-165300), which is a monoclonal antibody against the G-CSF variant of the IgGl type used as a control antibody. This indicates that the monoclonal antibody can specifically detect and quantify the protein of the present invention in pollen allergen extract, confirming its usefulness.
現在、減感作用アレルゲンエキスは素抽出物を利用している場合が多ぐ安定した 治療効果を維持するためにアレルゲンエキスの標準化が期待されている [「アレルゲン 免疫療法:アレルギ一疾患の治療ワクチン」に対する WHO見解, アレルギー, 69 8 (1998)]。 3に示したように、該タンパク質は極めて高い抗原性を有し、スギ花粉症の 発症において極めて重要な原因物質となっているといえる。したがって、該モノクロ一 ナル抗体を用いたアレルゲンエキスの定量、アレルゲンエキスの標準化およびその方 法は、効率的な減感作療法を進めるうえで極めて有用であると考えられる。 Currently, desensitizing allergen extracts are often stable using elementary extracts Standardization of allergen extracts is expected to maintain therapeutic effects [WHO's view on "allergen immunotherapy: a vaccine for the treatment of allergic diseases", Allergy, 696 (1998)]. As shown in 3, the protein has extremely high antigenicity and can be said to be a very important causative substance in the onset of cedar pollinosis. Therefore, quantification of the allergen extract using the monoclonal antibody, standardization of the allergen extract, and a method thereof are considered to be extremely useful in promoting efficient desensitization therapy.
産業上の利用可能性 Industrial applicability
本発明のタンパク質は、花粉症患者血清中の IgEと結合する性質を有することから、 免疫化学的診断法における抗原としての有用性はもとより、アレルギー疾患を惹起す る性質を有することから、アレルギー疾患を診断もしくは治療、予防するための減感作 剤としても有用である。  Since the protein of the present invention has the property of binding to IgE in the serum of hay fever patients, it is useful not only as an antigen in immunochemical diagnostic methods, but also has the property of inducing allergic disease, and It is also useful as a desensitizing agent for diagnosing, treating, or preventing.

Claims

請求の範囲 The scope of the claims
1. 下記の理化学的性質を有するタンパク質。  1. A protein with the following physicochemical properties:
(1)分子量: 51, 000± 5, OOODa (還元条件下 SDS—ポリアクリルアミドゲル電気泳 動)  (1) Molecular weight: 51,000 ± 5, OOODa (SDS-polyacrylamide gel electrophoresis under reducing conditions)
(2 ) N末端ァミノ酸配列: N末端に Xaa— Leu— Phe— Pro— Lys— Glu— Al a— Leu — Pro— Thrで表わされるアミノ酸配列を有する(ただし、 Xaaは、 Glu, Asp, His, Gl y、 Arg、 Trpまたは Cysのいずれかを示す)。  (2) N-terminal amino acid sequence: N-terminal has an amino acid sequence represented by Xaa—Leu—Phe—Pro—Lys—Glu—Ala—Leu—Pro—Thr (where Xaa is Glu, Asp, His , Gly, Arg, Trp or Cys).
(3)紫外吸収スペクトル:溶媒が、水、生理的食塩液またはリン酸緩衝液のとき、 280η m付近に極大吸収を有する。  (3) Ultraviolet absorption spectrum: when the solvent is water, physiological saline or phosphate buffer, it has a maximum absorption around 280ηm.
(4)溶剤への溶解性:水、生理的食塩液およびリン酸緩衝液に可溶である。  (4) Solubility in solvent: soluble in water, physiological saline and phosphate buffer.
(5)安定性:水溶液 (pH7. 3)中、 4°Cで 2か月間放置しても実質的に失活しない。  (5) Stability: Does not substantially deactivate even when left in an aqueous solution (pH 7.3) at 4 ° C for 2 months.
2. 花粉症患者の血清中 IgEとの結合活性を有し、かつアレルゲン活性を有する請 求項 1記載のタンパク質。  2. The protein according to claim 1, which has a binding activity to IgE in serum of hay fever patients and has allergen activity.
3. タンパク質力スギ花粉に由来する請求項 1または 2記載のタンパク質。  3. The protein according to claim 1, which is derived from cedar pollen.
4. 請求項 1〜3のいずれか 1項に記載のタンパク質の部分ペプチド。  4. A partial peptide of the protein according to any one of claims 1 to 3.
5. 請求項 1〜4のいずれ力 1項に記載のタンパク質または該タンパク質の部分ぺプ チドの誘導体。  5. The protein according to any one of claims 1 to 4, or a derivative of a partial peptide of the protein.
6. 花粉より溶媒で抽出した抽出物を、塩析、透析、濾過、濃縮、遠心分離、各種ク 口マトグラフィー、電気泳動から選ばれる精製手段と、アレルゲン活性を指標とする検 出手段とを組み合わせることによりタンパク質を採取することを特徴とする請求項 1〜3 のレ、ずれか 1項に記載のタンパク質を製造する方法。  6. The extract extracted from the pollen with the solvent is purified by salting-out, dialysis, filtration, concentration, centrifugation, various chromatographies, and electrophoresis. The method for producing a protein according to any one of claims 1 to 3, wherein the protein is collected by combining the proteins.
7. 請求項 1〜3のいずれ力 1項に記載のタンパク質を酵素もしくは化学分解または 合成することにより、請求項 4または 5に記載の部分ペプチドあるレ、は該部分ペプチド 誘導体を製造する方法。  7. A method for producing the partial peptide derivative according to claim 4 or 5, wherein the protein according to claim 1 is enzymatically or chemically decomposed or synthesized to synthesize the partial peptide derivative.
8. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ペプチド またはそれらの誘導体を有効成分として含有するアレルギー疾患診断薬。  8. An allergic disease diagnostic agent comprising the protein according to any one of claims 1 to 5, a partial peptide of the protein, or a derivative thereof as an active ingredient.
9. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ペプチド またはそれらの誘導体を有効成分として含有するアレルギー疾患治療薬。  9. A therapeutic agent for an allergic disease, comprising the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof as an active ingredient.
10. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体を有効成分として含有するアレルギ一疾患予防薬。  10. An allergic disease preventive agent comprising the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof as an active ingredient.
11. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体を有効成分として含有するアレルギー疾患減感作剤。  11. An allergic disease desensitizing agent comprising, as an active ingredient, the protein according to any one of claims 1 to 5, a partial peptide of the protein, or a derivative thereof.
12. 請求項 1〜5のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体に特異的に結合する IgEの定量方法。 12. The protein according to any one of claims 1 to 5, a partial peptide of the protein. Method for quantifying IgE that specifically binds to amides or their derivatives.
13. 請求項 1〜5のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体に特異的に結合する IgEの検出方法。  13. A method for detecting IgE that specifically binds to the protein according to any one of claims 1 to 5, a partial peptide of the protein, or a derivative thereof.
14. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体を用いる、アレルギー疾患の診断方法。  14. A method for diagnosing an allergic disease, using the protein according to any one of claims 1 to 5, a partial peptide of the protein, or a derivative thereof.
15. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体を経口あるいは非経口で反復投与することを特徴とする減感 作療法。  15. A desensitization therapy, wherein the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof is repeatedly orally or parenterally administered.
16. 請求項 1〜5のいずれ力 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分と して含有するアレルギー疾患診断薬。  16. A diagnostic agent for an allergic disease comprising the protein according to claim 1, a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof as an active ingredient.
17. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分と して含有するアレルギー疾患治療薬。  17. A therapeutic agent for an allergic disease, comprising as an active ingredient the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof.
18. 請求項:!〜 5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分と して含有するアレルギー疾患予防薬。  18. Claim: A prophylactic agent for an allergic disease, comprising as an active ingredient the protein according to any one of! To 5, a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof.
19. 請求項:!〜 5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を有効成分と して含有するアレルギー疾患減感作剤。  19. Claim :! 6. A desensitizing agent for an allergic disease, comprising as an active ingredient the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof, and a known allergen or a derivative thereof.
20. 請求項:!〜 5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を含む組成物 を用いる、アレルギー疾患の診断方法。  20. Claim: A method for diagnosing an allergic disease, comprising using the protein according to any one of! To 5, a partial peptide of the protein or a derivative thereof, and a composition containing a known allergen or a derivative thereof.
21. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体、および既知アレルゲンまたはそれらの誘導体を含む組成物 を経口あるレ、は非経口で反復投与することを特徴とする減感作療法。  21. A composition comprising the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof, and a composition comprising a known allergen or a derivative thereof, which is orally repeated parenterally. Desensitization therapy characterized by being administered.
22. 請求項 1〜5のいずれか 1項に記載のタンパク質、該タンパク質の部分ぺプチ ドまたはそれらの誘導体を認識する抗体またはその抗体断片。  22. An antibody or an antibody fragment thereof which recognizes the protein according to any one of claims 1 to 5, a partial peptide of the protein or a derivative thereof.
23. 請求項 1〜5のいずれか 1項に記載のタンパク質の N末端アミノ酸配列に特異的 に反応する、請求項 22に記載の抗体。  23. The antibody according to claim 22, which specifically reacts with the N-terminal amino acid sequence of the protein according to any one of claims 1 to 5.
24. 抗体またはその抗体断片が、配列番号 1記載のアミノ酸配列を認識する請求 項 22記載の抗体またはその抗体断片。  24. The antibody or antibody fragment thereof according to claim 22, wherein the antibody or antibody fragment thereof recognizes the amino acid sequence of SEQ ID NO: 1.
25. 抗体が、モノクローナル抗体である請求項 22〜24のいずれか 1項に記載の抗 体。 25. The antibody according to any one of claims 22 to 24, wherein the antibody is a monoclonal antibody. body.
26. モノクローナル抗体が、 KM2715である請求項 25記載の抗体。  26. The antibody according to claim 25, wherein the monoclonal antibody is KM2715.
27. 請求項 22〜26のいずれ力、 1項に記載の抗体または抗体断片を有効成分とし て含有するアレルギー疾患診断薬。  27. A diagnostic agent for an allergic disease, comprising the antibody or antibody fragment according to any one of claims 22 to 26 as an active ingredient.
28. 請求項 22〜26のレ、ずれ力、 1項に記載の抗体または抗体断片を有効成分とし て含有するアレルギー疾患治療薬。  28. A remedy for allergic diseases, comprising the antibody or antibody fragment according to claim 22 as an active ingredient.
29. 請求項 22〜26のいずれか 1項に記載の抗体または抗体断片を有効成分とし て含有するアレルギー疾患予防薬。  29. A prophylactic agent for an allergic disease, comprising the antibody or antibody fragment according to any one of claims 22 to 26 as an active ingredient.
30. 請求項 22〜26のいずれか 1項に記載の抗体または抗体断片を用いて、花粉 抗原を定量する方法。  30. A method for quantifying pollen antigen using the antibody or antibody fragment according to any one of claims 22 to 26.
31. 請求項 22〜26のいずれか 1項に記載の抗体または抗体断片を用いて、花粉 抗原中の該タンパク質を定量する方法。  31. A method for quantifying the protein in pollen antigens using the antibody or antibody fragment according to any one of claims 22 to 26.
32. 請求項 22〜26のいずれ力 1項に記載の抗体または抗体断片を用いる、花粉ァ レルギ一疾患の診断方法。  32. A method for diagnosing a pollen allergic disease using the antibody or antibody fragment according to any one of claims 22 to 26.
PCT/JP2000/003031 1999-05-11 2000-05-11 Novel pollen antigen WO2000068262A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU44316/00A AU4431600A (en) 1999-05-11 2000-05-11 Novel pollen antigen

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP11/129363 1999-05-11
JP12936399 1999-05-11

Publications (1)

Publication Number Publication Date
WO2000068262A1 true WO2000068262A1 (en) 2000-11-16

Family

ID=15007745

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2000/003031 WO2000068262A1 (en) 1999-05-11 2000-05-11 Novel pollen antigen

Country Status (2)

Country Link
AU (1) AU4431600A (en)
WO (1) WO2000068262A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0308147A1 (en) * 1987-09-12 1989-03-22 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Hyposensitization agent
EP0595855A1 (en) * 1991-07-12 1994-05-11 Immulogic Pharmaceutical Corporation Allergenic proteins and peptides from japanese cedar pollen
JPH10259198A (en) * 1996-12-24 1998-09-29 Sankyo Co Ltd Connected t-cell epitope and its use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0308147A1 (en) * 1987-09-12 1989-03-22 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Hyposensitization agent
EP0595855A1 (en) * 1991-07-12 1994-05-11 Immulogic Pharmaceutical Corporation Allergenic proteins and peptides from japanese cedar pollen
JPH10259198A (en) * 1996-12-24 1998-09-29 Sankyo Co Ltd Connected t-cell epitope and its use

Also Published As

Publication number Publication date
AU4431600A (en) 2000-11-21

Similar Documents

Publication Publication Date Title
AU2006319358B2 (en) Anti-Abeta globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
EP0683234B2 (en) Antibody against beta-amyloid or their derivative and use thereof
EP2542571B1 (en) Oligomer-specific amyloid beta epitope and antibodies
HU215245B (en) Method for producing human interleukin-4 polypeptides, antibodies produced against them and pharmaceutical preparations containing said antibodies
US20220372117A1 (en) Anti-allergen antibodies
JPH05199895A (en) Anti-human ige monoclonal antibody
KR101365375B1 (en) IDENTIFICATION OF NOVEL IgE EPITOPES
JP3990730B2 (en) Antigenic protein from Malassezia
WO2023206609A1 (en) Antibody against p-tau 217 and use thereof
WO2022179535A1 (en) Anti-sars-cov-2 nucleocapsid protein monoclonal antibody, and preparation method therefor and use thereof
JP2022172170A5 (en)
WO2000068262A1 (en) Novel pollen antigen
KR20070085689A (en) Immunoglobulin fractions
KR101019865B1 (en) RECOMBINANT ALLERGEN WITH REDUCED IgE BINDING, BUT UNDIMINISHED T-CELL ANTIGENICITY
JP7128191B2 (en) Therapeutic anti-IgE antibodies and methods and compositions thereof
US20030180225A1 (en) Walnut and ryegrass allergens
JPH0767689A (en) Anti-ld78 polypeptide monoclonal antibody
KR102406593B1 (en) Antibody specific to Fagales plant pollen or antigen-binding fragment thereof, composition or kit including the same, and detection method using the same
JP3522877B2 (en) Anti-tyrosinase monoclonal antibody F (ab ') 2 fragment
JP3323258B2 (en) Anti-human ADF monoclonal antibody and method for measuring human ADF using the same
JPH08231599A (en) Anti-tyrosinase monoclonal antibody fab fragment
JPH1017599A (en) Antifibroblast growth factor 5 monoclonal antibody
AU2012201856A1 (en) Anti-Abeta globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
AU2002325089A1 (en) Recombinant allergen with reduced IgE binding but undiminished T-cell antigenicity
AU2002325089A2 (en) Recombinant allergen with reduced IgE binding but undiminished T-cell antigenicity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM DZ 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

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

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 616236

Kind code of ref document: A

Format of ref document f/p: F