WO2006016571A1 - Remedy for endometriosis - Google Patents

Remedy for endometriosis Download PDF

Info

Publication number
WO2006016571A1
WO2006016571A1 PCT/JP2005/014559 JP2005014559W WO2006016571A1 WO 2006016571 A1 WO2006016571 A1 WO 2006016571A1 JP 2005014559 W JP2005014559 W JP 2005014559W WO 2006016571 A1 WO2006016571 A1 WO 2006016571A1
Authority
WO
WIPO (PCT)
Prior art keywords
endometriosis
gene
expression
mitdocaine
oligonucleotide
Prior art date
Application number
PCT/JP2005/014559
Other languages
French (fr)
Japanese (ja)
Inventor
Yutaka Osuga
Yasushi Hirota
Sadatoshi Sakuma
Original Assignee
Cell Signals Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cell Signals Inc. filed Critical Cell Signals Inc.
Publication of WO2006016571A1 publication Critical patent/WO2006016571A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/36Gynecology or obstetrics
    • G01N2800/364Endometriosis, i.e. non-malignant disorder in which functioning endometrial tissue is present outside the uterine cavity

Definitions

  • the present invention relates to a drug for preventing or treating endometriosis containing a compound that suppresses mitdocaine as an active ingredient, and a test method and a test drug for endometriosis using the expression of mitdocaine as an index.
  • Endometriosis is a mysterious disease that impairs the health of women of reproductive age (Mo moeda et al., 2002; Osuga et al., 2002). It is widely believed that the cause of this disease is the colonization, survival and proliferation of the eclamptic endometrial tissue in retrograde menstruation in the abdominal cavity. However, this hypothesis cannot answer the question of why only a small percentage of women have endometriosis, despite the fact that most women have retrograde menstruation (Halme et al., 1984). ).
  • Mitodocaine (hereinafter referred to as MK) is a molecule having pleiotropic activities such as cell proliferation, cell migration, angiogenesis, induction of inflammation and fibrinolysis (Muramatsu, 2002; Horiba et al., 2000). ; Takada et al., 1997). Functions such as promoting cell proliferation, angiogenesis and inflammation have all been suggested to be important in the development of tumor-like lesions such as tumors and endometriosis. In fact, high levels of MK have been reported in many malignancies and have been suggested to be associated with the development of cancer (Garver, Jr. et al.
  • MK is also detected in eclampsia and its levels are comparable to healthy women. Compared with orthotopic endometrium, ectopic endometrium has a significantly lower level of MK compared to orthotopic endometrium. It is known that when it is expressed! / (See Non-Patent Document 1).
  • Non-Patent Document 1 Chung HW et al, Mol Hum Reprod 8, 350-355 (2002)
  • the present invention has been made in view of such a situation, and the purpose thereof is to clarify that MK is involved in the development of eclampsia and to prevent eclampsia that targets MK. It is also intended to provide a drug for treatment and a test method and a test drug for endometriosis.
  • the present inventors have intensively studied to solve the above problems.
  • ESC cultured endometrial stromal cells
  • MK stimulates ESC proliferation by examining the mitogenic activity of MK.
  • MK concentration in PF we revealed that MK levels increased in PF in women with advanced endometriosis.
  • MK concentrations in female PF were significantly lower than those in other groups!
  • MK mRNA is expressed in peritoneal bone marrow derived cells (hereinafter referred to as PBMC), peritoneum and endometrial thread and tissue.
  • PBMC peritoneal bone marrow derived cells
  • MK is involved in the onset and progression of eclampsia. Therefore, by inhibiting MK, endometriosis can be prevented or treated, and endometriosis can be examined using MK expression as an indicator.
  • the present invention provides an invention including the following [1] to [4].
  • a drug for preventing or treating endometriosis comprising as an active ingredient a compound that suppresses expression of the mitdocaine gene.
  • [2] including a step of measuring the expression of the Midokine gene in a sample collected from the subject, Endometriosis testing method.
  • a reagent for endometriosis examination comprising an anti-mitodocaine antibody.
  • a reagent for endometriosis testing comprising an oligonucleotide that hybridizes to the mitodocaine gene region and has a chain length of at least 15 nucleotides.
  • Fig. 1 shows MK-induced proliferation of cultured ESC.
  • the effect of MK on ESC proliferation was examined by measuring BrdU incorporation into DNA by using cell proliferation ELISA. ESCs were treated with different concentrations of MK for 24 hours. The value is 5 averaged SEMs per sample. Data shown are representative of 3 experiments using different ESC preparations. *; p ⁇ 0.05, **; p ⁇ 0.001 (both relative to control).
  • FIG. 3 is a photograph showing the expression of MK mRNA in PBMC, peritoneum and eclampsia intimal tissue.
  • Total RNA isolated from bone marrow-derived cells, peritoneum, and endometrial tissue was reverse transcribed and amplified by PCR using MK primers. GAPDH amplification was used to confirm RNA quality and quantity.
  • Lanes 1-3 Bone marrow-derived cells in PF.
  • Lanes 4-6 Abdominal tissue.
  • Lanes 7-9 endometrial tissue.
  • Lane 10 negative control without cDNA.
  • Lane 11 DNA molecular weight Ma 1 ⁇ force ⁇ ⁇ .
  • the present invention provides a drug for preventing or treating endometriosis comprising a compound that suppresses the expression of the MK gene as an active ingredient.
  • MK gene as a factor associated with endometriosis preferably has the ability to mention the human MK gene shown in SEQ ID NO: 1. It is not limited to this. 'Similar DNA encoding inducible protein is also known as' endometriosis Included in the MK gene as a “joint factor”.
  • the sequence functionally equivalent to the DNA described in SEQ ID NO: 1 includes mutants derived from the same species, MK gene homologues derived from different species, and the like.
  • the previously reported mouse MK gene (Kadomatu, K., et al., Biochem. Biophy. Res. Commun., 151, 1312 (1988)) is functionally related to the DNA described in SEQ ID NO: 1. It is an example of an equivalent sequence, and is included in the MK gene as an eclampsia-related factor of the present invention.
  • SEQ ID NO: 1 Such a sequence functionally equivalent to the DNA described in SEQ ID NO: 1 is, for example, SEQ ID NO:
  • Such DNA includes a degenerate mutant of the DNA represented by SEQ ID NO: 1.
  • the human-derived MK gene described in SEQ ID NO: 1 is prepared from a human cDNA library with a sequence having high homology with the sequence described in SEQ ID NO: 1 or a part thereof. This can be carried out by selecting a target and recovering from the selected clone.
  • the human cDNA library for obtaining the MK gene for example, those constructed based on human kidney, human fetal kidney, brain, connective tissue, or total RNA derived from these cultured cells can be used. In addition to being obtained from nature, it may be synthesized by a DNA synthesizer based on the sequence described in SEQ ID NO: 1.
  • the preparation of a sequence functionally equivalent to the DNA of SEQ ID NO: 1 is performed by cDNA live derived from non-human mammalian cells (eg, human, monkey, mouse, rat, pig, ushi). From a rally, it can be carried out by selecting clones that can be hybridized under stringent conditions with the sequence of SEQ ID NO: 1 or a part thereof.
  • the conditions for hybridization are preferably highly stringent conditions. High stringency conditions are, for example, 65 ° C, 0.133 and 0.1% 303. Under these conditions, it is expected that DNA having high homology can be efficiently obtained as the temperature is increased.
  • factors that influence the stringency of the nodular and hybridization such as temperature and salt concentration, and those skilled in the art can select similar factors by selecting these factors as appropriate. It is possible to realize genies.
  • the preparation of a sequence functionally equivalent to the DNA described in SEQ ID NO: 1 should be performed as described above.
  • the DNA having the sequence ability described in SEQ ID NO: 1 can be appropriately modified by genetic engineering and prepared.
  • the MK polypeptide as an eclampsia-related factor is preferably a force capable of mentioning the MK polypeptide encoded by the DNA described in SEQ ID NO: 1.
  • SEQ ID NO: not limited thereto A polypeptide having an endometriosis-inducing activity comprising an amino acid sequence in which one or more amino acids are substituted, deleted, inserted or added in the amino acid sequence of the MK polypeptide encoded by the DNA of 1. Can be included.
  • a polypeptide functionally equivalent to the above MK usually has high homology in amino acid sequence with MK.
  • High homology usually means at least 50% identity, preferably 75% identity, more preferably 85% identity, more preferably 95% identity at the amino acid level. Refers to sex.
  • the identity of amino acid sequences and base sequences can be determined by BLAST (Proc. Natl. Acad. Sci. USA 90: 5873-5877, 1993).
  • One or more amino acids are mutated in the amino acid sequence of MK, and a polypeptide functionally equivalent to the polypeptide is also included in the present invention.
  • the number of amino acids to be mutated is usually within 30 amino acids, preferably within 15 amino acids, more preferably within 5 amino acids (eg, within 3 amino acids), and even more preferably within 2 amino acids.
  • the These mutated amino acid residues are preferably mutated to another amino acid that preserves the properties of the amino acid side chain.
  • amino acid side chain properties include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G , H, K, S, T), an amino acid having an aliphatic side chain (G, A, V, L, I, P), an amino acid having a hydroxyl group-containing side chain (S, T, ⁇ ), a sulfur atom-containing side Amino acids with chains (C, M), amino acids with carboxylic acid and amide-containing side chains (D, N, E, Q), amino acids with base-containing side chains (R, K, ⁇ ), aromatic-containing side Amino acids having a chain (H, F, Y, W) can be mentioned (both in parentheses represent single letter amino acids).
  • hydrophobic amino acids A, I, L, M, F, P, W, Y, V
  • hydrophilic amino acids R, D, N, C, E, Q, G , H, K, S,
  • One of the compounds that suppresses the expression of the MK gene in the present invention includes nucleotides that can suppress the expression of the MK gene.
  • the first aspect of the nucleotide capable of suppressing the expression of the MK gene is as follows.
  • Antisense oligonucleotides include, for example, antisense oligonucleotides that hybridize to any position in the base sequence of SEQ ID NO: 1.
  • This antisense oligonucleotide is preferably an antisense oligonucleotide against at least 15 or more consecutive nucleotides in the base sequence of SEQ ID NO: 1. More preferably, it is an antisense oligonucleotide in which at least 15 or more consecutive nucleotides contain a translation initiation codon.
  • Antisense oligonucleotides include not only nucleotides corresponding to nucleotides constituting a predetermined region of target MK DNA or mRNA, but all nucleotides complementary to each other, and oligonucleotides and bases represented by SEQ ID NO: 1. As long as it can specifically hybridize to a sequence, one having one or more nucleotide mismatches is also included.
  • an antisense oligonucleotide capable of suppressing MK gene expression for example, based on the primary sequence of MK mRNA, a secondary structure having the minimum energy is predicted by a computer program. . Based on this predicted secondary structure, a loop portion that does not form a base pair can be selected as an antisense oligonucleotide sequence candidate.
  • the secondary structure prediction of human MK mRNA is performed by computer analysis.
  • SEQ ID NO: 3 5′ GAAGCCTCGGTGCTGCAT-3
  • the oligonucleotide of the present invention can be appropriately designed based on the sequence of SEQ ID NO: 1, which is not limited to the sequence specifically shown above. Determination of whether or not the designed antisense oligonucleotide sequence has the activity of suppressing MK expression can be performed as follows. A substance with an antisense oligonucleotide sequence is administered to cells that synthesize and secrete MK, and MK is secreted into the culture medium by immunobiochemistry. Quantify to. Immunoassays for MK include enzyme immunoassay (Muramatsu, H. et al .: J.
  • oligonucleotide t is also used to refer to naturally occurring oligomeric nucleic acid portions such as DNA and RNA deoxyribonucleotide and ribonucleotide structures, and to naturally occurring nucleic acids. Includes both artificial analogs capable of binding. Examples of the artificial analog include a lower alkyl phosphonate modification, a phosphorothioate modification or a phosphoramidate modification, a morpholino modification, a reverse-oriented thymidine modification, such as a methylphosphonate type or an ethylphosphonate type. Therefore, the oligonucleotide designed as described above may be modified by any of these as required.
  • modified products have already been developed, and the already developed modified products may be applied to the therapeutic agent for eclampsia of the present invention.
  • morpholino modified products are disclosed in Japanese Patent Application Laid-Open No. 2003-210170
  • phosphorothioate modified products are disclosed in Japanese Patent Application Laid-Open No. 2002-142778
  • reverse thymidine modified products are disclosed in Japanese Patent Application Laid-Open No. 2003-210171.
  • phosphodiester-linked oligonucleotides are particularly susceptible to nuclease action in serum or inside cells. Therefore, a preferred embodiment of the antisense oligonucleotide of the present invention is a phosphorothioate-linked or methylophosphonate-linked analog that is nuclease resistant (Stein et al., Cancer Research 48: 2659, 1998).
  • oligonucleotide artificial analogs are prepared by methods well known to those skilled in the art, for example, methods using commercially available machines and reagents available from Perkin-Elmer / Applieds Biosystem (Foster City, CA). be able to.
  • the antisense oligonucleotide may not be in the form of an oligonucleotide itself, for example, after the target cell is infected as an expression construct mounted on an expression vector or the like. It can be generated as an RNA molecule.
  • the antisense oligonucleotide of the present invention is applied to a patient so that it can reach the affected area by force applied directly to the affected area of the patient or by administration into the blood vessel.
  • an antisense encapsulating material that enhances durability and membrane permeability can also be used. Examples include liposomes, poly-L-lysine, lipids, cholesterol, lipofectin, or derivatives thereof.
  • the dosage of the antisense oligonucleotide of the present invention can be appropriately adjusted according to the patient's condition, and a preferred amount can be used. For example, it can be administered in the range of 0.1 to 100 mg / kg, preferably 0.1 to 50 mg / kg.
  • the second embodiment of the nucleotide that suppresses the expression of the MK gene includes dsRNA targeting the MK gene.
  • the “dsRNA targeting the MK gene” means a dsRNA that can suppress the expression of the MK gene by RNA interference (RNAi).
  • RNAi RNA interference
  • One strand of dsRNA for suppressing MK gene expression with RNAi has a sequence complementary to at least MK mRNA. As long as it is a site where expression can be suppressed, it may have a complementary relationship in any region of MK mRNA. In addition, this complementarity may have a mismatch of about several bases with a target sequence that does not need to be complete as long as it can induce RNAi and suppress MK gene expression.
  • dsRNA both forms of artificially synthesized dsRNA itself or dsRNA expressed in mammalian cells by an expression vector can be adopted.
  • dsRNA When dsRNA is artificially synthesized and introduced into mammalian cells, it is necessary to consider the cytotoxicity of long dsRNA to mammalian cells, even if the dsRNA is expressed from an expression vector. . Therefore, both artificially synthesized dsRNA (hereinafter referred to as “synthetic dsRNA”) and dsRNA generated by expression should have a chain length of, for example, 19 to 30 base pairs, but this is longer than this. Even dsRNA can be used if it does not add cytotoxicity.
  • synthetic dsRNA artificially synthesized dsRNA
  • dsRNA generated by expression should have a chain length of, for example, 19 to 30 base pairs, but this is longer than this. Even dsRNA can be used if it does not add cytotoxicity.
  • a drug for preventing or treating endometriosis which comprises a compound that suppresses the function of MK protein as an active ingredient.
  • the compound that suppresses the function of MK includes an antibody that binds to MK.
  • An already developed anti-MK antibody (Muramatsu H. et al., Dev. Biol. 159: 392-402, 1993) that can suppress the action of MK is applied to the treatment and prevention of eclampsia in the present invention. May be.
  • a monoclonal antibody against human MK has also been developed (Japanese Patent Laid-Open No. 2002-085058), and this may be applied.
  • the antibodies of the present invention are not limited to these already reported antibodies, and anti-MK polyclonal antibodies or monoclonal antibodies prepared by the anti-MK antibody preparation method described in the later-described test method can also be used. .
  • a recombinant antibody or a modified antibody as the MK antibody used as a therapeutic agent.
  • a recombinant antibody for example, as a monoclonal antibody, the antibody gene is cloned by high-pridoma force, incorporated into an appropriate vector, and introduced into a host.
  • Recombinant antibodies produced using genetic recombination techniques can be used (for example, Borrebaeck CAK & Larrick JW, THERAPEUTIC MONOCLONAL ANTI BODIES, Published in the United Kingdom MACMILLAN PUBLISHERS LTD, 1990).
  • a chimeric antibody or a humanized antibody can be used as the modified antibody.
  • a chimeric antibody can be obtained by linking DNA encoding an antibody V region other than a human antibody and DNA encoding a human antibody C region, incorporating this into an expression vector, introducing it into a host, and producing it. (EP 125023, PCT WO96 / 02576) o
  • the antibody used in the present invention may be a modified antibody fragment as long as it binds to MK and inhibits the activity of MK.
  • Fab fragment, F (ab '), Fv or H chain
  • a method for examining endometriosis which comprises the step of measuring the expression of the MK gene in a sample collected from a subject.
  • MK is thought to be involved in the development of endometriosis, as will be described in the examples below. Therefore, in order to investigate the cause of endometriosis in patients and perform appropriate treatment for patients, it is useful to measure the expression of the MK gene as a test for endometriosis.
  • PF PF
  • PBMC peritoneal tissue
  • eclampsia intimal tissue can be used as a sample collected from a subject.
  • the translation product MK protein is measured.
  • a protein sample is prepared from the above-described subject sample.
  • the amount of MK protein is measured from the protein sample. Compare the measured protein amount to a control value based on the expression level of MK in healthy subjects. As a result of this comparison, if the subject's MK protein is significantly elevated compared to the control, it is determined that the subject may or has already developed endometriosis.
  • the above MK protein was measured by SDS polyacrylamide electrophoresis and Western blotting, dot blotting, immunoprecipitation, enzyme-linked immunoassay (ELISA), and immunoassay using antibodies to MK protein.
  • a fluorescence method can be exemplified.
  • the transcription product MK mRNA is measured.
  • a total RNA sample is prepared from the subject sample.
  • Total RNA can be prepared by methods well known to those skilled in the art. For example, it can be carried out by using a total RNA preparation kid Isogen "(Nitsubon Gene Co., Ltd.). Then, the amount of MK mRNA contained in the total RNA sample is measured. The amount of RNA measured is used as a control. Compare.
  • a cDNA sample is prepared from a subject sample.
  • a cDNA sample can be prepared by synthesizing cDNA using reverse transcriptase using the above-mentioned total RNA as a saddle. Next, the amount of MK cDNA contained in the cDNA sample is measured. The amount of cDN A measured is compared to the control.
  • Examples of methods for measuring gene expression based on these mRNAs and cDNAs include methods well known to those skilled in the art, such as Northern blotting, RT-PCR, and DNA array.
  • the subject may develop endometriosis, or It is determined that it has already developed.
  • a test reagent for use in a test method for endometriosis Is provided.
  • a test reagent include a test drug containing an oligonucleotide containing a partial sequence of the MK gene that can be used in the above test method (including a substrate on which an oligonucleotide probe is immobilized)
  • test drugs containing MK antibodies include test drugs containing MK antibodies. These antibodies and oligonucleotides may be labeled according to the test method.
  • test reagent for use in the endometriosis test method a test reagent for eclampsia that contains an anti-MK antibody can be mentioned.
  • the anti-MK antibody as the test agent may be, for example, a polyclonal antibody or a monoclonal antibody as long as it can detect MK.
  • a polyclonal antibody an antibody already developed by the present inventors may be used (Muramatsu H. et al, Dev. Biol. 159: 392-402, 1993).
  • Monoclonal antibodies have also been developed (Japanese Patent Application Laid-Open No. 2002-085 058) and can be suitably used.
  • Preparation of a specific antibody against MK can be performed according to a known method.
  • Monoclonal antibodies can be obtained, for example, using the entire human MK polypeptide encoded by the DNA represented by SEQ ID NO: 1 as a sensitizing antigen, or a partial sequence having a length capable of retaining antigenicity as a sensitizing antigen. You can use it to make it.
  • a hybridoma that produces a monoclonal antibody can basically be prepared as follows according to the method of Kohler and Milstein (Kohler G. & C. Milstein, Nature 256: 49 5-497, 1975). .
  • the above-described MK protein or a part thereof as a sensitizing antigen, it is immunized according to a normal immunization method, and the resulting immune cell is fused with a known parent cell by a normal cell fusion method. It can be prepared by screening monoclonal antibody-producing cells by ordinary screening methods.
  • the mammal to be immunized with the sensitizing antigen is not particularly limited, but it is generally preferable to select the mammal in consideration of compatibility with the parent cell used for cell fusion. Animals such as mice, rats, hamsters and the like are used.
  • a second embodiment is a reagent for endometriosis testing that includes an oligonucleotide that hybridizes to the MK gene region and has a chain length of at least 15 nucleotides.
  • the oligonucleotide specifically hybridizes to DNA (normal DNA or mutant DNA) containing the MK gene region.
  • DNA normal DNA or mutant DNA
  • specifically hybridize are described under normal and abbreviated hybridization conditions, preferably under stringent hybridization conditions (e.g., described in Sambu Norec et al., Molecular Cloning. Cold Spring Harbor Laboratory Press, New York, USA, 2nd edition 1989). In the above condition) does not cause significant cross-hybridization with DNA encoding other proteins.
  • the oligonucleotide need not be completely complementary to the DNA containing the MK gene region, if specific noidisation is possible! /.
  • Oligonucleotides that hybridize to DNA containing the MK gene region and have a chain length of at least 15 nucleotides can be used as probes (including the substrate on which the probe is immobilized) and primers in the test method of the present invention. it can.
  • the oligonucleotide is used as a primer, the length is usually 15 bp to 100 bp, preferably 17 bp to 30 bp.
  • the primer is not particularly limited as long as it can amplify at least a part of the MK gene region.
  • the probe is not particularly limited as long as it specifically hybridizes to DNA containing the MK gene region.
  • the probe may be a synthetic oligonucleotide and usually has a chain length of at least 15 bp.
  • the oligonucleotide of the present invention can be produced by, for example, a commercially available oligonucleotide synthesizer.
  • the probe can be prepared as a double-stranded DNA fragment obtained by restriction enzyme treatment or the like.
  • the oligonucleotide of the present invention is used as a probe, it is preferably used after being appropriately labeled.
  • T4 polynucleotide kinase is used to label the oligonucleotide by phosphorylating the 5 ′ end with 32 P
  • a random hexamer oligonucleotide is used using a DNA polymerase such as a Talenow enzyme.
  • Examples thereof include a method of incorporating a substrate base labeled with an isotope such as 32 P using a nucleotide or the like, a fluorescent dye, or piotin (such as a random prime method).
  • oligonucleotide of SEQ ID NO: 2 or SEQ ID NO: 3 As the oligonucleotide of the present invention
  • the oligonucleotide of the present invention is not limited to the sequence specifically shown above. Number: Designed appropriately based on the sequence of 1 can do.
  • ESC primary cultures were prepared from biopsies by the method described in Ryan, IP et al. With some modifications. Endometrial tissue is separated from the underlying soft tissue force and subdivided into small pieces, and type I collagenase (Sigma, St. Louis, MO) and deoxyribonuclease I (Takara, Tokyo) in DMEM / F12 , Japan) at 37 ° C for 1-2 hours and separated using continuous filtration. Cell debris was removed with a 100 / zm Nylon cell strainer (Becton Dickinson, Lincoln Park, NJ) and some of the epithelial glands were removed with a 70 m nylon cell strainer (Beta Ton Dickinson).
  • stromal cells remaining in the filtrate are collected by centrifugation, resuspended in DMEM / F12 medium, seeded in a 100 mm dish, allowed to adhere for 30 minutes at 37 ° C, and then rinsed with phosphate buffered saline. As a result, non-adherent epithelial cells and blood cells were removed. Cells were cultured in DMEM / F12 medium supplemented with 10% activated carbon treated FBS (HyClone, Logan, Utah) and antibiotics (Sigma).
  • the stromal cell population was subsequently purified by immunocytochemical staining with the following antibodies (Dako); vimentin (stromal cells), cytokeratin (epithelial cells) and CD45 (single cells). Spheres and other white blood cells). The purity of the stromal cells was 98% or higher, judging from the positive cell staining for vimentin and the negative cell staining for cytokeratin and CD45.
  • the age of women in the endometriosis group was 32.7 ⁇ 4.1 years (mean SD), essentially the same as that of women in the non-endometriosis group (31.1 ⁇ 5.6 years). This study was approved by the University of Tokyo Institutional Review Board, and consent was signed by each woman regarding sample collection.
  • PF also collected laparoscopic force-Eureka introduced into the Douglas fossa before any manipulation technique. PF was centrifuged at 400 xg for 10 minutes and the supernatant was stored frozen at -80 ° C until assembly. Peritoneal and endometrial tissues were also collected.
  • PBMC peripheral blood mononuclear cells
  • MK concentrations in PF were donated by Meiji Dairies (Odawara, Japan) or determined using the enzyme-linked immunoassay (EIA) for Cell Signals (Yokohama, Japan).
  • the detection limit of this EIA was 0.1 ng / ml / sample.
  • RNA extraction, MK mRNA reverse transcription-polymerase chain reaction (RT-PCR) MK mRNA reverse transcription-polymerase chain reaction (RT-PCR)
  • GPDH Human glyceraldehyde-3-phosphate dehydrogenase
  • an MK primer (sense, 5′-CCTGCAACTGGAAGAAGGAG-3 ′ (SEQ ID NO: 4); antisense, 5′-AGCAGACAGAAGGCACTGGT-3 ′ (SEQ ID NO: 5)) was selected.
  • PCR conditions for amplification of MK and GA PDH were 10 seconds at 98 ° C, 2 seconds at 60 ° C and 30 seconds at 74 ° C for 20 seconds.
  • PCR products were analyzed by agarose gel electrophoresis using ethidium bromide. Each PCR product was purified using the QIAEX II gel extraction kit (Qiagen) and the same using an ABI PRISM TM 310 gene analyzer (Applied Biosystems, Foster City, Calif.). The sex was confirmed.
  • MK mitogenic activity on cultured ESCs was examined by measuring 5-bromo-2'-deoxyuridine (Br dU) incorporation.
  • MK concentrations in PF in women with endometriosis or healthy women and in women treated with GnRH agonists were measured using the specific enzyme immunoassay.
  • the expression of MK mRNA in PBMC, peritoneum and endometrial tissue was assessed by RT-PCR.
  • Figure 1 shows the effect of MK on DNA synthesis in ESC. BrdU incorporation into DNA was significantly increased by 100 and 1000 ng / ml MK. 1000 ng / ml MK enhanced BrdU incorporation level to 125% of control.
  • MK concentrations in PF (0.93 ng / ml, 0.87 to 1.17; median, IQR) in women with mild eclampsia (stage I) were substantially the same as those in healthy women (0.97 ng / ml, 0.67-1.27). Therefore, we combined data from healthy women and women suffering from mild endometriosis to develop progressive epidural endometriosis (stage 11, III, and IV) (r-ASRM score ⁇ 6) Compared to the data of other women. As shown in Figure 2, MK concentrations in PF (1.21 ng / ml, 0.80-2.27) in women with advanced endometriosis are MK concentrations in healthy women and women with mild endometriosis.
  • MK concentrations in women receiving GnRHagost treatment were significantly lower than those in the other two groups (p 0.001).
  • MK concentrations in PF were compared according to menstrual period, the concentrations in the luteal phase (1.32 ng / ml, 0.72 to 2.21) were significantly higher than those in the follicular phase (0.95 ng / mU 0.68 to 1.24, p ⁇ 0.05) .
  • MK mRNA expression was detected as a clear band at 320 bp in PBMC, peritoneum, and eclampsia tissue.
  • MK stimulates ESC proliferation.
  • MK levels in PF were increased in women with advanced endometriosis compared to women with mild eclampsia or healthy women.
  • orthotopic endometrial cells in women with endometriosis express higher levels of MK compared to healthy women (Chung et al., 2002).
  • MKs derived from other sources other than ectopic endometrium may be involved in disease progression, but MK increased in orthotopic endometrium is very early stage of endometriosis May help in the establishment and survival of reflux endometrial cells.
  • MK stimulates intraperitoneal adhesions in a murine abdominal injury model (Inoh et al., 2004). Given that they tend to form internal adhesions, MK may also be involved in adhesion formation.
  • MK in PF The origin of MK in PF seems to be widely distributed.
  • Our RT-PCR analysis demonstrated that MK is expressed in PBMC, peritoneum and endometrial tissue.
  • MK in the follicular fluid released during ovulation is assumed to constitute part of the MK in PF because the follicular fluid contains a large amount of MK (Ohyama et al., 1994).
  • the high MK concentration in the follicular fluid! May partially explain the finding that the MK in PF is increased in the luteal phase compared to the follicular phase.
  • the hypoestrogenic state induced by GnR Ha treatment may repress MK gene transcription in various cells.
  • anovulation caused by GnRHa treatment may be involved in suppressing the influx of follicular fluid into the peritoneal cavity by suppressing MK levels in PF.
  • MK grows several cells, including fibroblasts, tumor cells, and keratinocytes (Inazumi et al., 1997; Muramatsu and Muramatsu, 1991; Mura matsu et al., 1993).
  • MK receptor capable of transmitting growth factor signals (Muramatsu, 2002).
  • endometriosis may be treated with MK inhibitors.
  • antisense oligonucleotides against MK have been shown to suppress tumor formation (Takei et al., 2001).
  • MK levels in PF have been shown to increase in women with advanced endometriosis compared to women with mild endometriosis or no endometriosis, and MK levels in PF Have been shown to be involved in the development and progression of eclampsia.

Abstract

By studying the mitogen activity of MK on cultured endometrial stromal cells, it is clarified that MK stimulates the proliferation of ESC. By evaluating the MK concentration in the peritoneal fluid (PF), it is clarified that the MK level is elevated in a female with the progress of endometriosis. It is further found out that the MK concentration in the PF of a female under the therapy with GnRH agonist is significantly lower than the concentration in another group. It is indicated that MK mRNA is expressed in cells originating in peritoneal bone marrow, the peritoneal membrane and an intrauterine tissue.

Description

子宮内膜症治療剤  Treatment for endometriosis
技術分野  Technical field
[0001] 本発明は、ミツドカインを抑制する化合物を有効成分として含有する子宮内膜症を 予防または治療するための薬剤並びにミツドカインの発現を指標とする子宮内膜症 の検査方法および検査薬に関する。  TECHNICAL FIELD [0001] The present invention relates to a drug for preventing or treating endometriosis containing a compound that suppresses mitdocaine as an active ingredient, and a test method and a test drug for endometriosis using the expression of mitdocaine as an index.
背景技術  Background art
[0002] 子宫内膜症は、生殖可能年齢の女性の健康を悪ィ匕させる不可解な疾患である (Mo moedaら、 2002 ; Osugaら、 2002)。この疾患の原因は、逆行性月経における子宫内膜 組織片が腹腔において定着、生存および増殖することにあると広く考えられている。 しかし、この仮説は、女性のほとんどに逆行性月経が認められるにも関わらず、なぜ ごく一部の女性のみが子宫内膜症に罹患するの力との疑問に答えられない(Halmeら 、 1984)。子宮内膜症を患う女性で認められる様々な異常の中で、疾患の発症に腹 腔環境の変化が極めて重要であることを、多くの証拠が示唆している(Haradaら、 200 l ; Lebovicら、 2001)。また、 TNF、 HGF、 SCF、 IFN- γのような成長因子もしくはサイト 力インの濃度が、子宮内膜症の女性の腹腔液 (以下 PFと称す)では、健常な女性と 比較して、変化することが示されている(Kogaら、 2000 ; Osugaら、 1999 ; Osugaら、 200 0 ;Yoshinoら、 2003) oこれらの物質は、細胞の生存、細胞増殖、および血管新生に お!、て様々な役割を有し、子宮内膜症の進行を刺激することが示唆されて 、る。  [0002] Endometriosis is a mysterious disease that impairs the health of women of reproductive age (Mo moeda et al., 2002; Osuga et al., 2002). It is widely believed that the cause of this disease is the colonization, survival and proliferation of the eclamptic endometrial tissue in retrograde menstruation in the abdominal cavity. However, this hypothesis cannot answer the question of why only a small percentage of women have endometriosis, despite the fact that most women have retrograde menstruation (Halme et al., 1984). ). Among the various abnormalities observed in women with endometriosis, much evidence suggests that changes in the abdominal environment are crucial for the onset of the disease (Harada et al., 200 l; Lebovic Et al., 2001). In addition, the concentrations of growth factors such as TNF, HGF, SCF, and IFN-γ, or cytodynamic force-in, change in the peritoneal fluid of endometriosis women (hereinafter referred to as PF) compared to healthy women. (Koga et al., 2000; Osuga et al., 1999; Osuga et al., 2000; Yoshino et al., 2003) o These substances are involved in cell survival, cell proliferation, and angiogenesis! It has been suggested to have various roles and stimulate the progression of endometriosis.
[0003] ミツドカイン (以下 MKと称す)は、細胞増殖、細胞遊走、血管新生、炎症の誘導およ び線維素溶解のような多面的活性を有する分子である(Muramatsu、 2002 ; Horibaら、 2000 ;Takadaら、 1997)。細胞増殖の促進、血管新生および炎症などの機能は全て、 腫瘍および子宮内膜症のような腫瘍様病変の発症において重要であることが示唆さ れている。実際に、多くの悪性腫瘍において高レベルの MKが報告されており、癌の 発症に関係することが示唆されている(Garver, Jr.ら 1994 ;Tsutsuiら、 1993 ;Nakagawa ら、 1995 ; O'Brienら、 1996 ; Koideら、 1999 ;Aridomeら、 1995 ; Katoら、 2000 ; Konishiら 、 1999)。 MKはまた、子宫内膜症においても検出され、そのレベルは健常な女性と比 較して子宮内膜症の女性における正所子宮内膜細胞では高い (非特許文献 1参照) 一方、正所子宮内膜と比較して、異所子宮内膜が顕著に低レベルの MKを発現す ると!/、うことが知られて 、る (非特許文献 1参照)。 [0003] Mitodocaine (hereinafter referred to as MK) is a molecule having pleiotropic activities such as cell proliferation, cell migration, angiogenesis, induction of inflammation and fibrinolysis (Muramatsu, 2002; Horiba et al., 2000). ; Takada et al., 1997). Functions such as promoting cell proliferation, angiogenesis and inflammation have all been suggested to be important in the development of tumor-like lesions such as tumors and endometriosis. In fact, high levels of MK have been reported in many malignancies and have been suggested to be associated with the development of cancer (Garver, Jr. et al. 1994; Tsutsui et al., 1993; Nakagawa et al., 1995; O ' Brien et al., 1996; Koide et al., 1999; Aridome et al., 1995; Kato et al., 2000; Konishi et al., 1999). MK is also detected in eclampsia and its levels are comparable to healthy women. Compared with orthotopic endometrium, ectopic endometrium has a significantly lower level of MK compared to orthotopic endometrium. It is known that when it is expressed! / (See Non-Patent Document 1).
非特許文献 1: Chung HW et al, Mol Hum Reprod 8, 350-355(2002)  Non-Patent Document 1: Chung HW et al, Mol Hum Reprod 8, 350-355 (2002)
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0004] 本発明はこのような状況に鑑みてなされたものであり、その目的は、 MKが子宫内膜 症の発症に関与することを明らかにし、 MKをターゲットとした子宫内膜症を予防また は治療するための薬剤、並びに子宮内膜症の検査方法および検査薬を提供するこ とにある。 [0004] The present invention has been made in view of such a situation, and the purpose thereof is to clarify that MK is involved in the development of eclampsia and to prevent eclampsia that targets MK. It is also intended to provide a drug for treatment and a test method and a test drug for endometriosis.
課題を解決するための手段  Means for solving the problem
[0005] 本発明者らは、上記の課題を解決するために鋭意研究を行った。本発明者らは、 まず、子宫内膜細胞力 子宫内膜細胞由来の MKのみならず、他の起源力 の MKに よって刺激されると考え、培養された子宮内膜間質細胞 (以下 ESCと称す)に及ぼす MKのマイトゲン活性を調べることにより、 MKが ESCの増殖を刺激することを明らかに した。次に、 PFにおける MK濃度を評価することにより、進行子宫内膜症の女性の PF において MKレベルが増加していることを明らかにした。さらに、 GnRHァゴニストによ る治療を受けて 、る女性の PFにおける MK濃度は、他の群の濃度より有意に低!、こと が分力つた。また、 MK mRNAが、腹腔骨髄由来細胞 (以下 PBMCと称す)、腹膜およ び子宮内膜糸且織において発現していることが判明した。 [0005] The present inventors have intensively studied to solve the above problems. First, the present inventors consider that cultured endometrial stromal cells (hereinafter referred to as ESC) are thought to be stimulated not only by MK derived from eclamptic endometrial cells but also by MKs of other origins. It was revealed that MK stimulates ESC proliferation by examining the mitogenic activity of MK. Next, by evaluating the MK concentration in PF, we revealed that MK levels increased in PF in women with advanced endometriosis. In addition, after treatment with GnRH agonists, it was found that MK concentrations in female PF were significantly lower than those in other groups! It was also found that MK mRNA is expressed in peritoneal bone marrow derived cells (hereinafter referred to as PBMC), peritoneum and endometrial thread and tissue.
以上の知見は、 MKが子宫内膜症の発症や進行に関与することを示唆している。従 つて、 MKの阻害により、子宮内膜症を予防または治療でき、また MKの発現を指標と して子宮内膜症を検査できると考えられる。  These findings suggest that MK is involved in the onset and progression of eclampsia. Therefore, by inhibiting MK, endometriosis can be prevented or treated, and endometriosis can be examined using MK expression as an indicator.
[0006] すなわち本発明は、以下の〔1〕〜〔4〕を含む発明を提供するものである。  That is, the present invention provides an invention including the following [1] to [4].
〔1〕ミツドカイン遺伝子の発現を抑制する化合物を有効成分とする、子宮内膜症を予 防または治療するための薬剤。  [1] A drug for preventing or treating endometriosis, comprising as an active ingredient a compound that suppresses expression of the mitdocaine gene.
〔2〕被検者より採取した試料中のミツドカイン遺伝子の発現を測定する工程を含む、 子宮内膜症の検査方法。 [2] including a step of measuring the expression of the Midokine gene in a sample collected from the subject, Endometriosis testing method.
〔3〕抗ミツドカイン抗体を含む子宮内膜症検査用試薬。  [3] A reagent for endometriosis examination comprising an anti-mitodocaine antibody.
〔4〕ミツドカイン遺伝子領域にハイブリダィズし、少なくとも 15ヌクレオチドの鎖長を有 するオリゴヌクレオチドを含む、子宮内膜症検査用試薬。  [4] A reagent for endometriosis testing, comprising an oligonucleotide that hybridizes to the mitodocaine gene region and has a chain length of at least 15 nucleotides.
図面の簡単な説明  Brief Description of Drawings
[0007] [図 1]培養 ESCの MK誘発増殖を示す図である。 ESCの増殖に及ぼす MKの作用を、 細胞増殖 ELISAを用いることによって、 BrdUの DNAへの取り込みを測定することによ つて調べた。 ESCを異なる濃度の MKによって 24時間処置した。値は 1試料あたり 5個 ずつの平均値士 SEMである。示したデータは、異なる ESC調製物を用いた 3実験の代 表である。 *;p〈0.05、 **;p〈0.001 (いずれも対照に対して)。  [0007] Fig. 1 shows MK-induced proliferation of cultured ESC. The effect of MK on ESC proliferation was examined by measuring BrdU incorporation into DNA by using cell proliferation ELISA. ESCs were treated with different concentrations of MK for 24 hours. The value is 5 averaged SEMs per sample. Data shown are representative of 3 experiments using different ESC preparations. *; p <0.05, **; p <0.001 (both relative to control).
[図 2]健常な女性、軽度の子宫内膜症を患う女性 (r-ASRMスコア、 0〜5、 n=58)、進 行子宫内膜症を患う女性 (r- ASRMスコア、≥6、 n=69)、および GnRHァゴ-ストによ つて治療した女性 (GnRHa、 n= 12)の PFにおける MK濃度を示す図である。四角の枠 は、第一四分位 (25%)から第三四分位 (75%)までの距離を表し、四角の中の水平 線は中央値を表す。バーは下限での 10パーセンタイルおよび上限での 90パーセンタ ィルを表す。 [Figure 2] Healthy women, women with mild eclampsia (r-ASRM score, 0-5, n = 58), women with progressive eclampsia ( r -ASRM score, ≥6, n = 69), and MK concentration in PF of women treated with GnRH agonist (GnRHa, n = 12). The square frame represents the distance from the first quartile (25%) to the third quartile (75%), and the horizontal line in the square represents the median. The bar represents the 10th percentile at the lower limit and the 90th percentile at the upper limit.
[図 3]PBMC、腹膜および子宫内膜組織における MK mRNAの発現を示す写真である 。骨髄由来細胞、腹膜、および子宮内膜組織から単離した総 RNAを逆転写して、 MK のプライマーを用いて PCRによって増幅した。 GAPDHの増幅を用いて RNAの質およ び量を確認した。レーン 1〜3 : PF中の骨髄由来細胞。レーン 4〜6:腹腔組織。レーン 7〜9:子宮内膜組織。レーン 10 : cDNAを含まない陰性対照。レーン 11 : DNA分子量 マ1 ~~力' ~~。 FIG. 3 is a photograph showing the expression of MK mRNA in PBMC, peritoneum and eclampsia intimal tissue. Total RNA isolated from bone marrow-derived cells, peritoneum, and endometrial tissue was reverse transcribed and amplified by PCR using MK primers. GAPDH amplification was used to confirm RNA quality and quantity. Lanes 1-3: Bone marrow-derived cells in PF. Lanes 4-6: Abdominal tissue. Lanes 7-9: endometrial tissue. Lane 10: negative control without cDNA. Lane 11: DNA molecular weight Ma 1 ~~ force` ~~.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0008] 本発明は、 MK遺伝子の発現を抑制する化合物を有効成分とする、子宮内膜症を 予防または治療するための薬剤を提供するものである。 [0008] The present invention provides a drug for preventing or treating endometriosis comprising a compound that suppresses the expression of the MK gene as an active ingredient.
子宫内膜症関連因子としての MK遺伝子は、好適には、配列番号: 1に示すヒト MK 遺伝子を挙げることができる力 これに限定されるものではなぐ MK遺伝子と同等に 子宮内膜症を発生'誘導し得るタンパク質をコードした類似の DNAも「子宮内膜症関 連因子」としての MK遺伝子に含まれる。配列番号: 1に記載の DNAと機能的に同等 な配列は、同種由来の変異体や異種由来の MK遺伝子ホモログなどが含まれる。例 えば、既に報告されているマウス MK遺伝子(Kadomatu, K., et al., Biochem. Biophy. Res. Commun., 151, 1312(1988))は配列番号: 1に記載の DNAと機能的に同等な配 列の一例であり、本発明の子宫内膜症関連因子としての MK遺伝子に含まれる。 MK gene as a factor associated with endometriosis preferably has the ability to mention the human MK gene shown in SEQ ID NO: 1. It is not limited to this. 'Similar DNA encoding inducible protein is also known as' endometriosis Included in the MK gene as a “joint factor”. The sequence functionally equivalent to the DNA described in SEQ ID NO: 1 includes mutants derived from the same species, MK gene homologues derived from different species, and the like. For example, the previously reported mouse MK gene (Kadomatu, K., et al., Biochem. Biophy. Res. Commun., 151, 1312 (1988)) is functionally related to the DNA described in SEQ ID NO: 1. It is an example of an equivalent sequence, and is included in the MK gene as an eclampsia-related factor of the present invention.
[0009] このような配列番号: 1に記載の DNAと機能的に同等の配列は、例えば、配列番号  [0009] Such a sequence functionally equivalent to the DNA described in SEQ ID NO: 1 is, for example, SEQ ID NO:
: 1に記載の塩基配列を含む DNAとストリンジェントな条件下でハイブリダィズし、かつ 子宫内膜症誘導活性を有するポリペプチドをコードした DNAを挙げることができる。こ のような DNAには、配列番号: 1に記載の DNAの縮重変異体も含まれる。  : A DNA that hybridizes with a DNA comprising the nucleotide sequence of 1 under stringent conditions and encodes a polypeptide having an activity of inducing eclampsia. Such DNA includes a degenerate mutant of the DNA represented by SEQ ID NO: 1.
[0010] 上記配列番号: 1に記載のヒト由来の MK遺伝子の調製は、ヒト cDNAライブラリーより 配列番号: 1に記載された配列またはその一部と相同性の高 、配列を保持するクロ ーンを選択し、選択されたクローンより回収することにより実施することができる。 MK 遺伝子を得るためのヒト cDNAライブラリ一は、たとえば、ヒトの腎臓、ヒトの胎児の腎臓 、脳、結合組織又はこれらの培養細胞由来トータル RNAに基づき構築されたものを 用いることができる。また、天然から取得する以外に、配列番号: 1に記載の配列に基 づヽて DNA合成器により合成してもよ 、。  [0010] The human-derived MK gene described in SEQ ID NO: 1 is prepared from a human cDNA library with a sequence having high homology with the sequence described in SEQ ID NO: 1 or a part thereof. This can be carried out by selecting a target and recovering from the selected clone. As the human cDNA library for obtaining the MK gene, for example, those constructed based on human kidney, human fetal kidney, brain, connective tissue, or total RNA derived from these cultured cells can be used. In addition to being obtained from nature, it may be synthesized by a DNA synthesizer based on the sequence described in SEQ ID NO: 1.
[0011] また、配列番号: 1に記載の DNAと機能的に同等の配列の調製は、ヒト以外の哺乳 動物細胞(例えば、ヒト、サル、マウス、ラット、ブタ、ゥシ)由来の cDNAライブラリーより 配列番号: 1記載の配列またはその一部とストリンジェントな条件下でノ、イブリダィズ するクローンを選択することにより実施することができる。ハイブリダィゼーシヨンの条 件は、当業者であれば適宜選択することができる。例えば、ハイブリダィゼーシヨンの 条件として、好ましくは、高ストリンジェントな条件が挙げられる。高ストリンジェントな条 件とは、例えば 65°C、 0.1 33じ及び0.1%303の条件でぁる。これらの条件において 、温度を上げる程に高い相同性を有する DNAが効率的に得られると期待される。伹 し、ノ、イブリダィゼーシヨンのストリンジエンシーに影響する要素としては温度や塩濃 度など複数の要素が考えられ、当業者であればこれら要素を適宜選択することで同 様のストリンジエンシーを実現することが可能である。  [0011] In addition, the preparation of a sequence functionally equivalent to the DNA of SEQ ID NO: 1 is performed by cDNA live derived from non-human mammalian cells (eg, human, monkey, mouse, rat, pig, ushi). From a rally, it can be carried out by selecting clones that can be hybridized under stringent conditions with the sequence of SEQ ID NO: 1 or a part thereof. Those skilled in the art can appropriately select the conditions for hybridization. For example, the conditions for hybridization are preferably highly stringent conditions. High stringency conditions are, for example, 65 ° C, 0.133 and 0.1% 303. Under these conditions, it is expected that DNA having high homology can be efficiently obtained as the temperature is increased. However, there are a number of factors that influence the stringency of the nodular and hybridization, such as temperature and salt concentration, and those skilled in the art can select similar factors by selecting these factors as appropriate. It is possible to realize genies.
また、配列番号: 1に記載の DNAと機能的に同等の配列の調製は、上述のように天 然に存在するものから選抜する方法の他に、配列番号: 1に記載の配列力 なる DNA を適宜、遺伝子工学的に改変して調製することもできる。 In addition, the preparation of a sequence functionally equivalent to the DNA described in SEQ ID NO: 1 should be performed as described above. In addition to the method of selecting from the existing ones, the DNA having the sequence ability described in SEQ ID NO: 1 can be appropriately modified by genetic engineering and prepared.
[0012] 子宫内膜症関連因子としての MKポリペプチドは、好ましくは、配列番号: 1に記載 の DNAがコードする MKポリペプチドを挙げることができる力 これに限定されるもので はなぐ配列番号: 1に記載の DNAがコードする MKポリペプチドのアミノ酸配列にお いて 1若しくは複数のアミノ酸が置換、欠失、挿入または付加されたアミノ酸配列から なり、子宮内膜症誘導活性を有するポリペプチドを含めることができる。  [0012] The MK polypeptide as an eclampsia-related factor is preferably a force capable of mentioning the MK polypeptide encoded by the DNA described in SEQ ID NO: 1. SEQ ID NO: not limited thereto : A polypeptide having an endometriosis-inducing activity comprising an amino acid sequence in which one or more amino acids are substituted, deleted, inserted or added in the amino acid sequence of the MK polypeptide encoded by the DNA of 1. Can be included.
[0013] 上記 MKと機能的に同等なポリペプチドは、通常、 MKとアミノ酸配列において高い 相同性を有する。高い相同性とは、アミノ酸レベルにおいて、通常、少なくとも 50%以 上の同一性、好ましくは 75%以上の同一性、さらに好ましくは 85%以上の同一性、さ らに好ましくは 95%以上の同一性を指す。なお、アミノ酸配列や塩基配列の同一性 は、 BLAST(Proc. Natl. Acad. Sci. USA 90:5873-5877, 1993)などによって決定するこ とがでさる。  [0013] A polypeptide functionally equivalent to the above MK usually has high homology in amino acid sequence with MK. High homology usually means at least 50% identity, preferably 75% identity, more preferably 85% identity, more preferably 95% identity at the amino acid level. Refers to sex. The identity of amino acid sequences and base sequences can be determined by BLAST (Proc. Natl. Acad. Sci. USA 90: 5873-5877, 1993).
[0014] MKのアミノ酸配列において 1もしくは複数のアミノ酸が変異したアミノ酸配列力もなり 、該ポリペプチドと機能的に同等なポリペプチドもまた本発明に含まれる。変異するァ ミノ酸数は、通常、 30アミノ酸以内であり、好ましくは 15アミノ酸以内であり、より好まし くは 5アミノ酸以内(例えば、 3アミノ酸以内)であり、さらに好ましくは 2アミノ酸以内であ る。これら変異するアミノ酸残基においては、アミノ酸側鎖の性質が保存されている別 のアミノ酸に変異されることが望ましい。例えば、アミノ酸側鎖の性質としては、疎水 性アミノ酸 (A、 I、 L、 M、 F、 P、 W、 Y、 V)、親水性アミノ酸(R、 D、 N、 C、 E、 Q、 G、 H、 K 、 S、 T)、脂肪族側鎖を有するアミノ酸 (G、 A、 V、 L、 I、 P)、水酸基含有側鎖を有する アミノ酸 (S、 T、 Υ)、硫黄原子含有側鎖を有するアミノ酸 (C、 M)、カルボン酸及びアミ ド含有側鎖を有するアミノ酸 (D、 N、 E、 Q)、塩基含有側鎖を有するアミノ酸 (R、 K、 Η )、芳香族含有側鎖を有するアミノ酸 (H、 F、 Y、 W)を挙げることができる(括弧内はい ずれもアミノ酸の一文字表記を表す)。  [0014] One or more amino acids are mutated in the amino acid sequence of MK, and a polypeptide functionally equivalent to the polypeptide is also included in the present invention. The number of amino acids to be mutated is usually within 30 amino acids, preferably within 15 amino acids, more preferably within 5 amino acids (eg, within 3 amino acids), and even more preferably within 2 amino acids. The These mutated amino acid residues are preferably mutated to another amino acid that preserves the properties of the amino acid side chain. For example, amino acid side chain properties include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G , H, K, S, T), an amino acid having an aliphatic side chain (G, A, V, L, I, P), an amino acid having a hydroxyl group-containing side chain (S, T, Υ), a sulfur atom-containing side Amino acids with chains (C, M), amino acids with carboxylic acid and amide-containing side chains (D, N, E, Q), amino acids with base-containing side chains (R, K, Η), aromatic-containing side Amino acids having a chain (H, F, Y, W) can be mentioned (both in parentheses represent single letter amino acids).
[0015] 本発明における MK遺伝子の発現を抑制する化合物の一つとしては、 MK遺伝子の 発現を抑制し得るヌクレオチドを挙げることができる。  [0015] One of the compounds that suppresses the expression of the MK gene in the present invention includes nucleotides that can suppress the expression of the MK gene.
MK遺伝子の発現を抑制し得るヌクレオチドの第一の態様としては、 MK遺伝子のァ ンチセンスオリゴヌクレオチドが挙げられる。 The first aspect of the nucleotide capable of suppressing the expression of the MK gene is as follows. An antisense oligonucleotide.
アンチセンスオリゴヌクレオチドとしては、例えば、配列番号: 1の塩基配列中のいず れかの箇所にハイブリダィズするアンチセンスオリゴヌクレオチドが含まれる。このアン チセンスオリゴヌクレオチドは、好ましくは配列番号: 1の塩基配列中の連続する少な くとも 15個以上のヌクレオチドに対するアンチセンスオリゴヌクレオチドである。さらに 好ましくは、連続する少なくとも 15個以上のヌクレオチドが翻訳開始コドンを含むアン チセンスオリゴヌクレオチドである。  Antisense oligonucleotides include, for example, antisense oligonucleotides that hybridize to any position in the base sequence of SEQ ID NO: 1. This antisense oligonucleotide is preferably an antisense oligonucleotide against at least 15 or more consecutive nucleotides in the base sequence of SEQ ID NO: 1. More preferably, it is an antisense oligonucleotide in which at least 15 or more consecutive nucleotides contain a translation initiation codon.
[0016] アンチセンスオリゴヌクレオチドは、標的となる MK DNA又は mRNAの所定の領域を 構成するヌクレオチドに対応するヌクレオチドが全て相補配列であるもののみならず 、オリゴヌクレオチドと配列番号: 1に示される塩基配列に特異的にハイブリダィズでき る限り、 1又は複数個のヌクレオチドのミスマッチが存在しているものも含まれる。  [0016] Antisense oligonucleotides include not only nucleotides corresponding to nucleotides constituting a predetermined region of target MK DNA or mRNA, but all nucleotides complementary to each other, and oligonucleotides and bases represented by SEQ ID NO: 1. As long as it can specifically hybridize to a sequence, one having one or more nucleotide mismatches is also included.
[0017] MK遺伝子発現を抑制し得るアンチセンスオリゴヌクレオチドの分子設計にっ 、て は、例えば、 MK mRNAの一次配列を基に、最小のエネルギーを持つ二次構造をコ ンピュータープログラムで予測する。この予測される二次構造に基づいて、塩基対を 作らないループ部分をアンチセンスオリゴヌクレオチド配列候補として選択することが できる。一例を示せば、ヒト MKの mRNA(GenBank Accession No. M69148 :Tsutsui J. et al. Biochem. Biophys. Res. Commun., 176:792-797, 1991)の二次構造予測をコン ピュータ解析により行うと、(ヒト MK mRNA配列の塩基番号 15番目から 32番目に基づ く)(配列番号: 2)、および 5'末端近傍でループを作らない配列に対応するアンチセ ンス DNA、(ヒト MK mRNA配列の塩基番号 1番目から 18番目に基づく)(配列番号: 3) を設計することができる。  [0017] According to the molecular design of an antisense oligonucleotide capable of suppressing MK gene expression, for example, based on the primary sequence of MK mRNA, a secondary structure having the minimum energy is predicted by a computer program. . Based on this predicted secondary structure, a loop portion that does not form a base pair can be selected as an antisense oligonucleotide sequence candidate. For example, the secondary structure prediction of human MK mRNA (GenBank Accession No. M69148: Tsutsui J. et al. Biochem. Biophys. Res. Commun., 176: 792-797, 1991) is performed by computer analysis. And (based on nucleotide numbers 15 to 32 of human MK mRNA sequence) (SEQ ID NO: 2), and an antisense DNA corresponding to a sequence that does not form a loop near the 5 'end (human MK mRNA sequence (SEQ ID NO: 3) can be designed.
配列番号 : 2 5' - AGGGTGAGGAGGAGGAAG - 3 '  SEQ ID NO: 2 5 '-AGGGTGAGGAGGAGGAAG-3'
配列番号:3 5 '— GAAGCCTCGGTGCTGCAT - 3,  SEQ ID NO: 3 5′—GAAGCCTCGGTGCTGCAT-3,
[0018] しかし、本発明のオリゴヌクレオチドは上記具体的に示された配列に限定されるもの ではなぐ配列番号: 1の配列に基づいて、適宜設計することができる。そして、設計 されたアンチセンスオリゴヌクレオチド配列が MKの発現抑制活性を有する力否かの 判定は、次の通り行うことができる。アンチセンスオリゴヌクレオチド配列を持つ物質を 、 MKを合成分泌している細胞に投与し、培養液中に分泌される MKを免疫生化学的 に定量する。 MKの免疫生化学的定量法としては酵素免疫法 (Muramatsu, H. et al. : J. Biochem., 119 : 1171-1175, 1996)あるいは SDSポリアクリルアミドゲル電気泳動後 のウェスタンブロッテイング(Muramatsu, H. et al. : Dev. Biol, 159 ; 392-402, 1993) そしてデンシトメ一ターによる定量がある。この定量により MK遺伝子産物の産生を低 下させ得るアンチセンスオリゴヌクレオチドは MKの遺伝子発現を抑制し得るものとし て選択することができる。 [0018] However, the oligonucleotide of the present invention can be appropriately designed based on the sequence of SEQ ID NO: 1, which is not limited to the sequence specifically shown above. Determination of whether or not the designed antisense oligonucleotide sequence has the activity of suppressing MK expression can be performed as follows. A substance with an antisense oligonucleotide sequence is administered to cells that synthesize and secrete MK, and MK is secreted into the culture medium by immunobiochemistry. Quantify to. Immunoassays for MK include enzyme immunoassay (Muramatsu, H. et al .: J. Biochem., 119: 1171-1175, 1996) or Western blotting after SDS polyacrylamide gel electrophoresis (Muramatsu, H. et al .: Dev. Biol, 159; 392-402, 1993) and quantification by densitometer. An antisense oligonucleotide that can reduce the production of the MK gene product by this quantification can be selected as one that can suppress the MK gene expression.
[0019] また上記「オリゴヌクレオチド」 t 、う用語は、 DNAおよび RNAのデォキシリボヌクレ ォチドおよびリボヌクレオチド構造のような天然に存在するオリゴマーの核酸部分、な らびに天然に存在する核酸に結合する能力のある人工アナログの両方を包含する。 人工アナログとしては、例えばメチルホスホネート型又はェチルホスホネート型のよう な低級アルキルホスホネート修飾体、ホスホロチォエート修飾体又はホスホロアミデ ート修飾体、モルホリノ修飾体、逆向きチミジン修飾体等が挙げられる。したがって、 上記設計されたオリゴヌクレオチドは必要に応じて、これらいずれかにより修飾されて もよい。こうした修飾体は既に開発されており、既に開発されている修飾体を本発明 の子宫内膜症治療薬に応用してもよい。例えば、モルホリノ修飾体は特開 2003-2101 70に、ホスホロチォエート修飾体は特開 2002-142778に、逆向きチミジン修飾体は特 開 2003-210171に開示されて!ヽる。  [0019] The term "oligonucleotide" t is also used to refer to naturally occurring oligomeric nucleic acid portions such as DNA and RNA deoxyribonucleotide and ribonucleotide structures, and to naturally occurring nucleic acids. Includes both artificial analogs capable of binding. Examples of the artificial analog include a lower alkyl phosphonate modification, a phosphorothioate modification or a phosphoramidate modification, a morpholino modification, a reverse-oriented thymidine modification, such as a methylphosphonate type or an ethylphosphonate type. Therefore, the oligonucleotide designed as described above may be modified by any of these as required. Such modified products have already been developed, and the already developed modified products may be applied to the therapeutic agent for eclampsia of the present invention. For example, morpholino modified products are disclosed in Japanese Patent Application Laid-Open No. 2003-210170, phosphorothioate modified products are disclosed in Japanese Patent Application Laid-Open No. 2002-142778, and reverse thymidine modified products are disclosed in Japanese Patent Application Laid-Open No. 2003-210171.
[0020] これら修飾体のうちホスホジエステル結合したオリゴヌクレオチドは、血清でまたは 細胞内部でヌクレアーゼの作用を特に受けやすい。そのため本発明のアンチセンス オリゴヌクレオチドの好適な態様としては、ヌクレアーゼ抵抗性であるホスホロチォェ ート結合あるいはメチロホスホネート結合したアナログなどである(Stein et al., Cancer Research 48: 2659, 1998)。  [0020] Of these modifications, phosphodiester-linked oligonucleotides are particularly susceptible to nuclease action in serum or inside cells. Therefore, a preferred embodiment of the antisense oligonucleotide of the present invention is a phosphorothioate-linked or methylophosphonate-linked analog that is nuclease resistant (Stein et al., Cancer Research 48: 2659, 1998).
なお、このようなオリゴヌクレオチドの人工アナログは、当業者らに周知の方法、例 えば、市販の機械および Perkin- Elmer/Applieds Biosystem (Foster City, CA)より入 手できる試薬を用いる方法によって調製することができる。  Such oligonucleotide artificial analogs are prepared by methods well known to those skilled in the art, for example, methods using commercially available machines and reagents available from Perkin-Elmer / Applieds Biosystem (Foster City, CA). be able to.
[0021] 本発明の他の態様において、このアンチセンスオリゴヌクレオチドは、オリゴヌクレオ チドそのものの形態でなくともよぐ例えば、発現ベクターなどに搭載させた発現構築 物として標的細胞を感染させた後にアンチセンス RNA分子として生成させてもょ 、。 本発明のアンチセンスオリゴヌクレオチドは患者の患部に直接適用する力、又は血 管内に投与するなどして結果的に患部に到達し得るように患者に適用する。さらには 、持続性、膜透過性を高めるアンチセンス封入素材を用いることもできる。例えば、リ ポソーム、ポリ- L-リジン、リピッド、コレステロール、リポフエクチン又はこれらの誘導体 が挙げられる。 [0021] In another embodiment of the present invention, the antisense oligonucleotide may not be in the form of an oligonucleotide itself, for example, after the target cell is infected as an expression construct mounted on an expression vector or the like. It can be generated as an RNA molecule. The antisense oligonucleotide of the present invention is applied to a patient so that it can reach the affected area by force applied directly to the affected area of the patient or by administration into the blood vessel. Furthermore, an antisense encapsulating material that enhances durability and membrane permeability can also be used. Examples include liposomes, poly-L-lysine, lipids, cholesterol, lipofectin, or derivatives thereof.
本発明のアンチセンスオリゴヌクレオチドの投与量は、患者の状態に応じて適宜調 整し、好ましい量を用いることができる。例えば、 0.1〜100mg/kg、好ましくは 0.1〜50 mg/kgの範囲で投与することができる。  The dosage of the antisense oligonucleotide of the present invention can be appropriately adjusted according to the patient's condition, and a preferred amount can be used. For example, it can be administered in the range of 0.1 to 100 mg / kg, preferably 0.1 to 50 mg / kg.
[0022] MK遺伝子の発現を抑制するヌクレオチドの第二の態様としては、 MK遺伝子を標的 とした dsRNAが挙げられる。  [0022] The second embodiment of the nucleotide that suppresses the expression of the MK gene includes dsRNA targeting the MK gene.
「MK遺伝子を標的とした dsRNA」とは、 MK遺伝子の発現を RNA干渉(RNAi)により 抑制し得る dsRNAを意味する。 MK遺伝子発現を RNAiで抑制するための dsRNAは、 一方の鎖は少なくとも MK mRNAと相補する配列を備える。発現抑制し得る部位であ れば、 MK mRNAのいずれの領域において相補関係を有してもよい。また、この相補 性は、 RNAiを誘導し MK遺伝子発現を抑制し得る限り、完全である必要はなぐ標的 配列との間で数塩基程度のミスマッチを有しても良い。  The “dsRNA targeting the MK gene” means a dsRNA that can suppress the expression of the MK gene by RNA interference (RNAi). One strand of dsRNA for suppressing MK gene expression with RNAi has a sequence complementary to at least MK mRNA. As long as it is a site where expression can be suppressed, it may have a complementary relationship in any region of MK mRNA. In addition, this complementarity may have a mismatch of about several bases with a target sequence that does not need to be complete as long as it can induce RNAi and suppress MK gene expression.
[0023] dsRNAは、人工的に合成された dsRNAそのもの、あるいは発現ベクターにより哺乳 動物細胞内で発現させた dsRNAの双方の形態を採用することができる。  [0023] As the dsRNA, both forms of artificially synthesized dsRNA itself or dsRNA expressed in mammalian cells by an expression vector can be adopted.
dsRNAを人工的に合成し、哺乳動物細胞に導入する場合、発現ベクターから dsRN Aを発現させる場合の 、ずれの場合にも、長鎖 dsRNAによる哺乳動物細胞への細胞 毒性を考慮する必要がある。そのため、人工的に合成した dsRNA (以下「合成 dsRNA 」という)、発現により生成される dsRNAのいずれも、鎖長は例えば 19〜30塩基対とす ることがょ 、が、これよりも長 ヽ dsRNAであっても細胞毒性を与えな 、ものであれば用 いることがでさる。  When dsRNA is artificially synthesized and introduced into mammalian cells, it is necessary to consider the cytotoxicity of long dsRNA to mammalian cells, even if the dsRNA is expressed from an expression vector. . Therefore, both artificially synthesized dsRNA (hereinafter referred to as “synthetic dsRNA”) and dsRNA generated by expression should have a chain length of, for example, 19 to 30 base pairs, but this is longer than this. Even dsRNA can be used if it does not add cytotoxicity.
上記 MK mRNAを標的とした dsRNAは、生体内で MK mRNAと対合し、生体内の酵 素により MK mRNAの切断を誘導する。そのため、 MKの mRNAからタンパク質への翻 訳を阻害し、結果として MKの発現を抑制し、子宮内膜症発症を抑制することが可能 となる。 [0024] 本発明の第二の側面として、 MKタンパク質の機能を抑制する化合物を有効成分と する、子宮内膜症を予防または治療するための薬剤が提供される。 The above dsRNA targeting MK mRNA is paired with MK mRNA in vivo, and induces cleavage of MK mRNA by an in vivo enzyme. Therefore, translation of MK from mRNA to protein is inhibited, and as a result, expression of MK can be suppressed and the onset of endometriosis can be suppressed. [0024] As a second aspect of the present invention, there is provided a drug for preventing or treating endometriosis, which comprises a compound that suppresses the function of MK protein as an active ingredient.
MKの機能を抑制する化合物としては、 MKに結合する抗体が挙げられる。 MKの作用を抑え得る既に開発されている抗 MK抗体(Muramatsu H. et al., Dev. Bi ol. 159: 392-402, 1993)を本発明の子宫内膜症治療 ·予防用に応用してもよい。また 、ヒト MKに対するモノクローナル抗体も開発されており(特開 2002-085058)、これを 応用してもよい。しかし、本発明の抗体はこれら既に報告されている抗体に限定され るものではなぐ後述の検査方法において説明する抗 MK抗体の作製方法により作製 された抗 MKポリクローナル抗体またはモノクローナル抗体を用いることもできる。  The compound that suppresses the function of MK includes an antibody that binds to MK. An already developed anti-MK antibody (Muramatsu H. et al., Dev. Biol. 159: 392-402, 1993) that can suppress the action of MK is applied to the treatment and prevention of eclampsia in the present invention. May be. A monoclonal antibody against human MK has also been developed (Japanese Patent Laid-Open No. 2002-085058), and this may be applied. However, the antibodies of the present invention are not limited to these already reported antibodies, and anti-MK polyclonal antibodies or monoclonal antibodies prepared by the anti-MK antibody preparation method described in the later-described test method can also be used. .
[0025] また、治療剤に使用する MK抗体は組み換え型抗体や改変抗体を用いることも有効 である。組み換え型抗体としては、例えば、モノクローナル抗体として、抗体遺伝子を ハイプリドーマ力 クロー-ングし、適当なベクターに組み込んでこれを宿主に導入し  [0025] It is also effective to use a recombinant antibody or a modified antibody as the MK antibody used as a therapeutic agent. As a recombinant antibody, for example, as a monoclonal antibody, the antibody gene is cloned by high-pridoma force, incorporated into an appropriate vector, and introduced into a host.
、遺伝子組み換え技術を用いて産生させた組み換え型抗体を用いることができる(例 えば、 Borrebaeck C. A. K. & Larrick J. W., THERAPEUTIC MONOCLONAL ANTI BODIES, Published in the United Kingdom MACMILLAN PUBLISHERS LTD, 1990 )。改変抗体としては、例えばキメラ抗体、ヒト型化抗体が使用できる。キメラ抗体は、 ヒト抗体以外の抗体 V領域をコードする DNAとヒト抗体 C領域をコードする DNAとを連 結し、これを発現ベクターに組み込んで宿主に導入し、産生させることにより得ること ができる(EP 125023, PCT WO96/02576) o Recombinant antibodies produced using genetic recombination techniques can be used (for example, Borrebaeck CAK & Larrick JW, THERAPEUTIC MONOCLONAL ANTI BODIES, Published in the United Kingdom MACMILLAN PUBLISHERS LTD, 1990). As the modified antibody, for example, a chimeric antibody or a humanized antibody can be used. A chimeric antibody can be obtained by linking DNA encoding an antibody V region other than a human antibody and DNA encoding a human antibody C region, incorporating this into an expression vector, introducing it into a host, and producing it. (EP 125023, PCT WO96 / 02576) o
本発明で使用される抗体は、 MKに結合し MKの活性を阻害するかぎり、抗体の断 片ゃ修飾物であってもよい。例えば、抗体の断片として、 Fab、 F(ab')、 Fvまたは H鎖  The antibody used in the present invention may be a modified antibody fragment as long as it binds to MK and inhibits the activity of MK. For example, as an antibody fragment, Fab, F (ab '), Fv or H chain
2  2
と L鎖の Fvを適当なリンカ一で連結させたシングルチヱイン Fv(scFv)が挙げられる。  And single chain Fv (scFv) in which Fv of L chain is linked with an appropriate linker.
[0026] 本発明の第三の側面として、被検者より採取した試料中の MK遺伝子の発現を測定 する工程を含む子宮内膜症の検査方法が提供される。 [0026] As a third aspect of the present invention, there is provided a method for examining endometriosis, which comprises the step of measuring the expression of the MK gene in a sample collected from a subject.
MKは後述の実施例でも説明するように、子宮内膜症の発生に関与すると考えられ る。従って、患者における子宮内膜症の発生原因を調査し、患者に対して適切な処 理を行うためには、子宫内膜症の検査として MK遺伝子の発現を測定することが有用 となる。 被検者より採取される試料としては、例えば、 PF、 PBMC、腹膜組織または子宫内 膜組織などを用いることができる。 MK is thought to be involved in the development of endometriosis, as will be described in the examples below. Therefore, in order to investigate the cause of endometriosis in patients and perform appropriate treatment for patients, it is useful to measure the expression of the MK gene as a test for endometriosis. As a sample collected from a subject, for example, PF, PBMC, peritoneal tissue or eclampsia intimal tissue can be used.
[0027] MK遺伝子発現の測定における第一の態様として、翻訳産物である MKタンパク質 を測定する。先ず、上述した被検者の試料よりタンパク質試料を調製する。次いで、 該タンパク質試料カゝら MKタンパク質量を測定する。測定されたタンパク質量を健常 者などの MK発現量に基づいた対照値などと比較する。この比較の結果、被検者の M Kタンパク質が対照に比べて有意に上昇している場合には、被験者が子宮内膜症を 発症する可能性がある、または、すでに発症していると判定される。  [0027] As a first embodiment in the measurement of MK gene expression, the translation product MK protein is measured. First, a protein sample is prepared from the above-described subject sample. Next, the amount of MK protein is measured from the protein sample. Compare the measured protein amount to a control value based on the expression level of MK in healthy subjects. As a result of this comparison, if the subject's MK protein is significantly elevated compared to the control, it is determined that the subject may or has already developed endometriosis. The
上記 MKタンパク質の測定は、 SDSポリアクリルアミド電気泳動法、並びに MKタンパ ク質に対する抗体を用いた、ウェスタンブロッテイング法、ドットブロッテイング法、免疫 沈降法、酵素結合免疫測定法 (ELISA)、および免疫蛍光法を例示することができる。  The above MK protein was measured by SDS polyacrylamide electrophoresis and Western blotting, dot blotting, immunoprecipitation, enzyme-linked immunoassay (ELISA), and immunoassay using antibodies to MK protein. A fluorescence method can be exemplified.
[0028] また、 MK遺伝子の発現を測定するための別の方法としては、転写産物である MK mRNAを測定する。先ず、上記被検者試料からトータル RNA試料を調製する。トータ ル RNAの調製は、当業者に周知の方法で行うことができる。例えば、トータル RNA調 製用のキッド Isogen " (二ツボンジーン社)を用いて実施することができる。次いで、該 トータル RNA試料に含まれる MK mRNA量を測定する。測定された RNAの量を対照と 比較する。  [0028] As another method for measuring the expression of the MK gene, the transcription product MK mRNA is measured. First, a total RNA sample is prepared from the subject sample. Total RNA can be prepared by methods well known to those skilled in the art. For example, it can be carried out by using a total RNA preparation kid Isogen "(Nitsubon Gene Co., Ltd.). Then, the amount of MK mRNA contained in the total RNA sample is measured. The amount of RNA measured is used as a control. Compare.
[0029] 別の態様としては、被検者試料から cDNA試料を調製する。 cDNA試料の調製は、 上述したトータル RNAを铸型として、逆転写酵素を用いて cDNAの合成を行うことで実 施し得る。次いで、該 cDNA試料に含まれる MK cDNA量を測定する。測定された cDN Aの量を対照と比較する。  [0029] In another embodiment, a cDNA sample is prepared from a subject sample. A cDNA sample can be prepared by synthesizing cDNA using reverse transcriptase using the above-mentioned total RNA as a saddle. Next, the amount of MK cDNA contained in the cDNA sample is measured. The amount of cDN A measured is compared to the control.
これら mRNAや cDNAを基に遺伝子発現を測定する方法としては、当業者に周知の 方法、例えばノーザンブロッテイング法、 RT-PCR法、 DNAアレイ法等を挙げることが できる。  Examples of methods for measuring gene expression based on these mRNAs and cDNAs include methods well known to those skilled in the art, such as Northern blotting, RT-PCR, and DNA array.
mRNAまたは cDNAの比較の結果、被検者の MKタンパク mRNAまたは cDNAのが対 照に比べて有意に上昇している場合には、被験者が子宮内膜症を発症する可能性 がある、または、すでに発症していると判定される。  If the mRNA or cDNA comparison results in a significant increase in the subject's MK protein mRNA or cDNA compared to the control, the subject may develop endometriosis, or It is determined that it has already developed.
[0030] 本発明の第四の側面として、子宮内膜症の検査方法に用いるための検査用試薬 が提供される。このような検査用試薬としては、上記検査方法に使用し得る MK遺伝 子の一部配列を含むオリゴヌクレオチドを主成分とする検査薬 (オリゴヌクレオチドプ ローブが固定された基板を含む)や、抗 MK抗体を含む検査薬が挙げられる。これら 抗体やオリゴヌクレオチドは、検査方法に応じて、標識が付加されていてもよい。 [0030] As a fourth aspect of the present invention, a test reagent for use in a test method for endometriosis Is provided. Examples of such a test reagent include a test drug containing an oligonucleotide containing a partial sequence of the MK gene that can be used in the above test method (including a substrate on which an oligonucleotide probe is immobilized) Examples include test drugs containing MK antibodies. These antibodies and oligonucleotides may be labeled according to the test method.
[0031] 子宮内膜症の検査方法に用いるための検査用試薬における第一の態様として、抗 MK抗体を含む子宫内膜症検査用試薬が挙げられる。上記検査薬としての抗 MK抗 体は、例えば、ポリクローナル抗体であっても、モノクローナル抗体であってもよぐ M Kを検出し得る抗体であればよい。例えば、ポリクローナル抗体であれば、既に本願 発明者らにより開発されている抗体を用いてもよい(Muramatsu H. et al, Dev. Biol. 159: 392-402, 1993)。モノクローナル抗体もまた既に開発されており(特開 2002-085 058)、これを好適に利用することができる。  [0031] As a first aspect of the test reagent for use in the endometriosis test method, a test reagent for eclampsia that contains an anti-MK antibody can be mentioned. The anti-MK antibody as the test agent may be, for example, a polyclonal antibody or a monoclonal antibody as long as it can detect MK. For example, in the case of a polyclonal antibody, an antibody already developed by the present inventors may be used (Muramatsu H. et al, Dev. Biol. 159: 392-402, 1993). Monoclonal antibodies have also been developed (Japanese Patent Application Laid-Open No. 2002-085 058) and can be suitably used.
[0032] MKに対する特異抗体の作製は、公知の方法に準じて実施することができる。モノク ローナル抗体は、例えば、配列番号: 1に記載の DNAがコードするヒト MKポリべプチ ド全体を感作抗原として、または抗原性を保持し得る長さを有する一部配列を感作 抗原として用いて作製してもよ ヽ。モノクローナル抗体を産生するハイプリドーマは、 基本的には、ケーラーとミルスタインの方法(Kohler G. & C. Milstein, Nature 256: 49 5-497, 1975)に準じて、以下のようにして作製できる。すなわち、上述した MKタンパ ク質またはその一部を感作抗原として使用して、これを通常の免疫方法に従って免 疫し、得られる免疫細胞を通常の細胞融合法によって公知の親細胞と融合させ、通 常のスクリーニング法により、モノクローナル抗体産生細胞をスクリーニングすることに よって作製できる。  [0032] Preparation of a specific antibody against MK can be performed according to a known method. Monoclonal antibodies can be obtained, for example, using the entire human MK polypeptide encoded by the DNA represented by SEQ ID NO: 1 as a sensitizing antigen, or a partial sequence having a length capable of retaining antigenicity as a sensitizing antigen. You can use it to make it. A hybridoma that produces a monoclonal antibody can basically be prepared as follows according to the method of Kohler and Milstein (Kohler G. & C. Milstein, Nature 256: 49 5-497, 1975). . That is, using the above-described MK protein or a part thereof as a sensitizing antigen, it is immunized according to a normal immunization method, and the resulting immune cell is fused with a known parent cell by a normal cell fusion method. It can be prepared by screening monoclonal antibody-producing cells by ordinary screening methods.
感作抗原で免疫される哺乳動物としては、特に限定されるものではないが、細胞融 合に使用する親細胞との適合性を考慮して選択するのが好ましぐ一般には、齧歯 類の動物、例えば、マウス、ラット、ハムスター等が使用される。  The mammal to be immunized with the sensitizing antigen is not particularly limited, but it is generally preferable to select the mammal in consideration of compatibility with the parent cell used for cell fusion. Animals such as mice, rats, hamsters and the like are used.
[0033] 第二の態様としては、 MK遺伝子領域にハイブリダィズし、少なくとも 15ヌクレオチド の鎖長を有するオリゴヌクレオチドを含む子宮内膜症検査用試薬である。 [0033] A second embodiment is a reagent for endometriosis testing that includes an oligonucleotide that hybridizes to the MK gene region and has a chain length of at least 15 nucleotides.
該オリゴヌクレオチドは、 MK遺伝子領域を含む DNA (正常型 DNAまたは変異型 DN A)に特異的にノ、イブリダィズするものである。ここで「特異的にハイブリダィズする」と は、通常のノ、イブリダィゼーシヨン条件下、好ましくはストリンジェントなハイブリダィゼ ーシヨン条件下 (例えば、サムブノレックら, Molecular Cloning.Cold Spring Harbour Lab oratory Press, New York,USA,第 2版 1989に記載の条件)において、他のタンパク質を コードする DNAとクロスハイブリダィゼーシヨンを有意に生じな 、ことを意味する。特異 的なノヽイブリダィズが可能であれば、該オリゴヌクレオチドは、 MK遺伝子領域を含む DNAに対し、完全に相補的である必要はな!/、。 The oligonucleotide specifically hybridizes to DNA (normal DNA or mutant DNA) containing the MK gene region. Where “specifically hybridize” Are described under normal and abbreviated hybridization conditions, preferably under stringent hybridization conditions (e.g., described in Sambu Norec et al., Molecular Cloning. Cold Spring Harbor Laboratory Press, New York, USA, 2nd edition 1989). In the above condition) does not cause significant cross-hybridization with DNA encoding other proteins. The oligonucleotide need not be completely complementary to the DNA containing the MK gene region, if specific noidisation is possible! /.
[0034] MK遺伝子領域を含む DNAにハイブリダィズし、少なくとも 15ヌクレオチドの鎖長を 有するオリゴヌクレオチドは、上記本発明の検査方法におけるプローブ (該プローブ が固定された基板を含む)やプライマーとして用いることができる。該オリゴヌクレオチ ドをプライマーとして用いる場合、その長さは、通常 15bp〜100bpであり、好ましくは 17 bp〜30bpである。プライマーは MK遺伝子領域の少なくとも一部を増幅しうるものであ れば、特に制限されない。  [0034] Oligonucleotides that hybridize to DNA containing the MK gene region and have a chain length of at least 15 nucleotides can be used as probes (including the substrate on which the probe is immobilized) and primers in the test method of the present invention. it can. When the oligonucleotide is used as a primer, the length is usually 15 bp to 100 bp, preferably 17 bp to 30 bp. The primer is not particularly limited as long as it can amplify at least a part of the MK gene region.
[0035] また、上記オリゴヌクレオチドをプローブとして使用する場合、該プローブは、 MK遺 伝子領域を含む DNAに特異的にハイブリダィズするものであれば、特に制限されな い。該プローブは、合成オリゴヌクレオチドであってもよぐ通常少なくとも 15bp以上の 鎖長を有する。  [0035] When the oligonucleotide is used as a probe, the probe is not particularly limited as long as it specifically hybridizes to DNA containing the MK gene region. The probe may be a synthetic oligonucleotide and usually has a chain length of at least 15 bp.
本発明のオリゴヌクレオチドは、例えば市販のオリゴヌクレオチド合成機により作製 することができる。プローブは、制限酵素処理等によって取得される二本鎖 DNA断片 として作製することちできる。  The oligonucleotide of the present invention can be produced by, for example, a commercially available oligonucleotide synthesizer. The probe can be prepared as a double-stranded DNA fragment obtained by restriction enzyme treatment or the like.
[0036] 本発明のオリゴヌクレオチドをプローブとして用いる場合は、適宜標識して用いるこ とが好ましい。標識する方法としては、 T4ポリヌクレオチドキナーゼを用いて、オリゴヌ クレオチドの 5'端を32 Pでリン酸ィ匕することにより標識する方法、およびタレノウ酵素等 の DNAポリメラーゼを用い、ランダムへキサマーオリゴヌクレオチド等をプライマーとし て32 P等のアイソトープ、蛍光色素、またはピオチン等によって標識された基質塩基を 取り込ませる方法 (ランダムプライム法等)を例示することができる。 [0036] When the oligonucleotide of the present invention is used as a probe, it is preferably used after being appropriately labeled. As a labeling method, T4 polynucleotide kinase is used to label the oligonucleotide by phosphorylating the 5 ′ end with 32 P, and a random hexamer oligonucleotide is used using a DNA polymerase such as a Talenow enzyme. Examples thereof include a method of incorporating a substrate base labeled with an isotope such as 32 P using a nucleotide or the like, a fluorescent dye, or piotin (such as a random prime method).
本発明のオリゴヌクレオチドとして、配列番号: 2または配列番号: 3に記載のオリゴ ヌクレオチドを利用することができる力 本発明のオリゴヌクレオチドは上記具体的に 示された配列に限定されるものではなぐ配列番号: 1の配列に基づいて、適宜設計 することができる。 The ability to utilize the oligonucleotide of SEQ ID NO: 2 or SEQ ID NO: 3 as the oligonucleotide of the present invention The oligonucleotide of the present invention is not limited to the sequence specifically shown above. Number: Designed appropriately based on the sequence of 1 can do.
なお本明細書において引用された全ての先行技術文献は、参照として本明細書に 組み入れられる。  All prior art documents cited in the present specification are incorporated herein by reference.
実施例  Example
[0037] 以下、本発明を実施例により、さらに具体的に説明するが本発明はこれら実施例に 制限されるものではない。  Hereinafter, the present invention will be described more specifically with reference to examples, but the present invention is not limited to these examples.
1. ESCの単離、精製および培養  1. ESC isolation, purification and culture
ESCの初代培養は、 Ryan, IP et al.に記載されている方法に、多少の改変を加えた 方法により、生検から調製した。子宮内膜組織をその下の柔組織力 切り離し、小片 に細分化し、 DMEM/F12において I型コラゲナーゼ(シグマ(Sigma)、セントルイス、ミ ズーリ州)およびデォキシリボヌクレアーゼ I (タカラ (Takara)、東京、 日本)と共に 37°C で 1〜2時間インキュベートして、連続濾過を用いて分離した。細胞片を 100 /z mのナ イロン製細胞濾過器(ベタトンディッキンソン(Becton Dickinson)、リンカーンパーク、 ニュージャージー州)によって除去して、上皮腺のいくつかを 70 mのナイロン製細 胞濾過器 (ベタトンディッキンソン)によって除去した。濾液に残っている間質細胞を 遠心によって回収し、 DMEM/F12培地に再浮遊させ、 100 mm皿に播種して 37°Cで 3 0分間接着させた後、リン酸緩衝生理食塩液ですすぐことにより非接着上皮細胞およ び血球を除去した。細胞を、 10%活性炭処理した FBS (ノヽイクローン(HyClone)、ロー ガン、ユタ州)および抗生物質 (シグマ)を添カ卩した DMEM/F12培地で培養した。細 胞がコンフルェントに達した後、 0.25%トリプシンによって細胞を解離して、遠心によ つて回収し、ファルコン 96マルチウエルプレート(ベタトンディッキンソン)に 1 X 104個/ ゥエルで播種して、加湿された 5%CO /95%空気の環境で 37°Cで維持した。 24時間 ESC primary cultures were prepared from biopsies by the method described in Ryan, IP et al. With some modifications. Endometrial tissue is separated from the underlying soft tissue force and subdivided into small pieces, and type I collagenase (Sigma, St. Louis, MO) and deoxyribonuclease I (Takara, Tokyo) in DMEM / F12 , Japan) at 37 ° C for 1-2 hours and separated using continuous filtration. Cell debris was removed with a 100 / zm Nylon cell strainer (Becton Dickinson, Lincoln Park, NJ) and some of the epithelial glands were removed with a 70 m nylon cell strainer (Beta Ton Dickinson). The stromal cells remaining in the filtrate are collected by centrifugation, resuspended in DMEM / F12 medium, seeded in a 100 mm dish, allowed to adhere for 30 minutes at 37 ° C, and then rinsed with phosphate buffered saline. As a result, non-adherent epithelial cells and blood cells were removed. Cells were cultured in DMEM / F12 medium supplemented with 10% activated carbon treated FBS (HyClone, Logan, Utah) and antibiotics (Sigma). After the cells reach confluence, dissociate the cells with 0.25% trypsin, collect them by centrifugation, seed them in a Falcon 96 multiwell plate (Bettaton Dickinson) at 1 x 10 4 cells / well, and humidify. Maintained at 37 ° C in a 5% CO 2/95% air environment. 24hours
2  2
後、間質細胞集団が精製されたことを、以下の抗体 (ダコ(Dako) )による免疫細胞化 学染色によって確認した;ビメンチン(間質細胞)、サイトケラチン (上皮細胞)および C D45 (単球および他の白血球)。間質細胞の純度は、ビメンチンに関する細胞染色が 陽性であったことと、サイトケラチンおよび CD45に関する細胞染色が陰性であったこ とから判断して、 98%以上であった。  The stromal cell population was subsequently purified by immunocytochemical staining with the following antibodies (Dako); vimentin (stromal cells), cytokeratin (epithelial cells) and CD45 (single cells). Spheres and other white blood cells). The purity of the stromal cells was 98% or higher, judging from the positive cell staining for vimentin and the negative cell staining for cytokeratin and CD45.
[0038] 2. PF試料の採取 本試験の被験者は、疼痛および Zまたは不妊のために腹腔鏡検査を受けた年齢 2 0〜48歳の子宫内膜症を患う女性 (n= 106)および健常な女性 (n=33)の、全体で 13 9人である。子宮内膜症は腹腔鏡および組織学の双方によって診断した。疾患の程 度は、改訂アメリカ生殖医学会 (r-ASRM)分類システム (アメリカ生殖医学会、 1997) に従って採点した。非子宮内膜症群において、女性 14人が卵胞期で、 19人が黄体 期であった。子宮内膜症群には、腹腔鏡検査まで 3ヶ月を超える期間 GnRHァゴニス トによる治療を受けた女性 12人を含む (GnRHa群)。 GnRHa群を除く女性は全て、月 経周期が規則的であった;そのうち、女性 49人が卵胞期であり、 45人が黄体期であつ た。 GnRHa治療を受けていない子宫内膜症群では、子宫内膜症の状態は、軽度の 子宫内膜症 (r- ASRMスコア、 l〜5 ;n=25)、および進行子宫内膜症 (r- ASRMスコア ≥6、 n=69)であると分類され、軽度の子宮内膜症は疾患の開始段階であると仮定し た。子宮内膜症群の女性の年齢は 32.7±4.1歳(平均値士 SD)であり、非子宮内膜症 群の女性の年齢と本質的に同じであった (31.1 ±5.6歳)。本研究は、東京大学学内 倫理委員会の承認を受け、試料の採取に関しては各女性カゝら署名された同意書を 得た。 [0038] 2. Collection of PF sample The subjects in this study included women (n = 106) and healthy women (n = 33) with eclampsia who were laparoscopic for pain and Z or infertility, aged 20-48 years. There are 13 9 people in total. Endometriosis was diagnosed by both laparoscope and histology. The degree of disease was scored according to the revised American Society for Reproductive Medicine (r-ASRM) classification system (American Society for Reproductive Medicine, 1997). In the non-endometriosis group, 14 women were in the follicular phase and 19 were in the luteal phase. The endometriosis group includes 12 women treated with GnRH agonist for more than 3 months before laparoscopy (GnRHa group). All women except the GnRHa group had a regular menstrual cycle; of these, 49 were in the follicular phase and 45 were in the luteal phase. In the group of eclampsia that has not received GnRHa treatment, the status of eclampsia is mild eclampsia (r-ASRM score, l-5; n = 25), and advanced eclampsia (r -ASRM score ≥6, n = 69), assuming that mild endometriosis is the onset stage of the disease. The age of women in the endometriosis group was 32.7 ± 4.1 years (mean SD), essentially the same as that of women in the non-endometriosis group (31.1 ± 5.6 years). This study was approved by the University of Tokyo Institutional Review Board, and consent was signed by each woman regarding sample collection.
[0039] PFは、何らかの操作技法の前にダグラス窩に導入した腹腔鏡力-ユーレカも採取し た。 PFを 400 X gで 10分間遠心して、上清をアツセィするまで- 80°Cで凍結保存した。 腹膜および子宮内膜組織も同様に採取した。  [0039] PF also collected laparoscopic force-Eureka introduced into the Douglas fossa before any manipulation technique. PF was centrifuged at 400 xg for 10 minutes and the supernatant was stored frozen at -80 ° C until assembly. Peritoneal and endometrial tissues were also collected.
PBMCは、周知の方法で採取した (Yoshinoら、 2003)。簡単に説明すると、採取した PFを 200 X gで 5分間遠心して、上清を除去した。細胞沈降物をリン酸緩衝生理食塩 液(PBS)に再浮遊させて、フイコール-パック(アマシャムバイオサイェンシズ (Amersh am Biosciences)、アップサラ、スウェーデン)に重層して、 150 X gで 30分間遠心した。 PBMCを界面から回収した。  PBMC were collected by well-known methods (Yoshino et al., 2003). Briefly, the collected PF was centrifuged at 200 × g for 5 minutes to remove the supernatant. Cell pellets are resuspended in phosphate buffered saline (PBS) and overlaid on Ficoll-Pak (Amersh am Biosciences, Upsala, Sweden) at 150 X g for 30 minutes Centrifuged. PBMC were collected from the interface.
[0040] 3.細胞増殖アツセィ  [0040] 3. Cell proliferation
細胞増殖アツセィは、本発明者らが先に報告した方法で実施した (Tangら、 2002)。 バイオトラック細胞増殖 ELISAシステム(アマシャムバイオサイェンシズ)を用いて製造 元の説明書に従って、 DNAへの 5-ブロモ -2'-デォキシゥリジン(BrdU)取り込みを測 定することによって、 ESCの増殖に及ぼす MKの作用を調べた。簡単に説明すると、 E SCをファルコン 96マルチウエルプレートに 1 X 104個/ゥエルの密度で、異なる濃度(0、 10、 100および 1000 ng/ml)の MKと共に培養培地 100 1〖こおいて播種した。 24時間 後、 BrdU溶液 100 1をカ卩えて 37°Cでさらに 2時間インキュベートした。 培養培地を除 去した後、細胞を固定して、固定液 200 1/ゥエルをカ卩えて DNAを変性させた。ペル ォキシダーゼ標識抗 BrdU抗体は、新たに合成された細胞 DNAに取り込まれた BrdU に結合した。その後の基質反応によって免疫複合体を検出し、得られた色をディジス キャンマイクロプレートリーダー(ASYSノヽィテク GmbH、オイゲンドルフ、オーストリア) において 450 nmで読み取った。 Cell proliferation assay was performed as previously reported by the inventors (Tang et al., 2002). Effect on ESC proliferation by measuring 5-bromo-2'-deoxyuridine (BrdU) incorporation into DNA using Biotrack cell proliferation ELISA system (Amersham Biosciences) according to manufacturer's instructions The effect of MK was examined. In short, E SCs were seeded in Falcon 96 multiwell plates at a density of 1 × 10 4 cells / well in 100 1 culture medium with different concentrations of MK (0, 10, 100 and 1000 ng / ml). After 24 hours, BrdU solution 1001 was added and incubated at 37 ° C for an additional 2 hours. After removing the culture medium, the cells were fixed, and DNA was denatured by adding a fixing solution 2001 / well. Peroxidase-labeled anti-BrdU antibody bound to BrdU incorporated into newly synthesized cellular DNA. Subsequent substrate reactions detected immune complexes and the resulting color was read at 450 nm in a digscan microplate reader (ASYS Neutech GmbH, Eugendorf, Austria).
[0041] 4. MKの測定  [0041] 4. MK measurement
PFにおける MK濃度は、明治乳業 (小田原、 日本)から寄贈され、またはセルシグナ ルズ (Cell Signals,横浜、 日本)所用の酵素結合ィムノアッセィ (EIA)を用いて決定し た。この EIAの検出限界は、 0.1 ng/ml/試料であった。  MK concentrations in PF were donated by Meiji Dairies (Odawara, Japan) or determined using the enzyme-linked immunoassay (EIA) for Cell Signals (Yokohama, Japan). The detection limit of this EIA was 0.1 ng / ml / sample.
[0042] 5. RNA抽出、 MK mRNAの逆転写-ポリメラーゼ連鎖反応(RT-PCR)  [0042] 5. RNA extraction, MK mRNA reverse transcription-polymerase chain reaction (RT-PCR)
RNイージーミニキット(キアゲン(QIAGEN)、ヒルデン、ドイツ)を用いて、総 RNAを P BMC、腹膜、および子宮内膜組織力も抽出した。 RT- PCRは、レバー 'トラ'ダッシュ( 東洋紡、東京、 日本)を用いて行った。総 RNA 1 gを全量 20 1中で逆転写して、ヒ ト MK配列に基づくオリゴヌクレオチドプライマーを用いて cDNAを増幅した。ヒトグリセ ルアルデヒド- 3-リン酸デヒドロゲナーゼ (GAPDH)プライマー(東洋紡、東京、 日本) を用いて、 RNAの質および量を確認した。 320 bp断片を増幅するために、 MKプライ マー(センス、 5 ' -CCTGCAACTGGAAGAAGGAG-3 ' (配列番号: 4);アンチセンス 、 5 ' -AGCAGACAGAAGGCACTGGT-3 ' (配列番号: 5) )を選択した。 MKおよび GA PDHの増幅に関する PCR条件は、 98°Cで 10秒、 60°Cで 2秒および 74°Cで 20秒を 30サ イタルであった。 PCR産物を、ェチジゥムブロマイドを用いるァガロースゲル電気泳動 によって分析した。それぞれの PCR産物を、 QIAEX IIゲル抽出キット(キアゲン)によ つて精製し、 ABI PRISM (商標) 310遺伝子アナライザ (アプライドバイォシステムズ (A pplied Biosystems)、フォスターシティ、カリフォルニア州)を用いてその同一性を確認 した。  Total RNA was also extracted from PBMC, peritoneum, and endometrial tissue using the RN Easy Mini Kit (QIAGEN, Hilden, Germany). RT-PCR was performed using a lever 'tiger' dash (Toyobo, Tokyo, Japan). 1 g of total RNA was reverse transcribed in a total volume of 201 and cDNA was amplified using oligonucleotide primers based on the human MK sequence. Human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) primer (Toyobo, Tokyo, Japan) was used to confirm RNA quality and quantity. To amplify the 320 bp fragment, an MK primer (sense, 5′-CCTGCAACTGGAAGAAGGAG-3 ′ (SEQ ID NO: 4); antisense, 5′-AGCAGACAGAAGGCACTGGT-3 ′ (SEQ ID NO: 5)) was selected. PCR conditions for amplification of MK and GA PDH were 10 seconds at 98 ° C, 2 seconds at 60 ° C and 30 seconds at 74 ° C for 20 seconds. PCR products were analyzed by agarose gel electrophoresis using ethidium bromide. Each PCR product was purified using the QIAEX II gel extraction kit (Qiagen) and the same using an ABI PRISM ™ 310 gene analyzer (Applied Biosystems, Foster City, Calif.). The sex was confirmed.
[0043] 6.統計分析 ANOVAを用いて培養細胞の BrdU取り込みを比較した。 PF中の MKのデータは、中 央値と四分位数間範囲(IQR)として記述した。マン-ホイト-一検定を用いて PF中の MK濃度を比較した。 pく 0.05は、統計学的に有意であると考えられる。 [0043] 6. Statistical analysis ANOVA was used to compare BrdU incorporation of cultured cells. MK data in the PF is described as the median and interquartile range (IQR). The MK concentration in PF was compared using the Mann-Whout-One test. A value of 0.05 is considered statistically significant.
[0044] 〔実施例 1〕 [Example 1]
培養された ESCに及ぼす MKのマイトゲン活性を、 5-ブロモ -2'-デォキシゥリジン(Br dU)取り込みを測定することによって調べた。子宮内膜症を患う女性または健常な女 性の PFにおける MK濃度と、 GnRHァゴニストにより治療中の女性の場合の濃度を、特 異的酵素ィムノアツセィを用いて測定した。 PBMC、腹膜および子宮内膜組織におけ る MK mRNAの発現を、 RT-PCRによって評価した。  MK mitogenic activity on cultured ESCs was examined by measuring 5-bromo-2'-deoxyuridine (Br dU) incorporation. MK concentrations in PF in women with endometriosis or healthy women and in women treated with GnRH agonists were measured using the specific enzyme immunoassay. The expression of MK mRNA in PBMC, peritoneum and endometrial tissue was assessed by RT-PCR.
ESCにおける DNA合成に及ぼす MKの作用を図 1に示す。 DNAへの BrdU取り込み は、 100および 1000 ng/mlの MKによって有意に増加した。 1000 ng/mlの MKは BrdU 取り込みレベルを対照の 125%まで増強した。  Figure 1 shows the effect of MK on DNA synthesis in ESC. BrdU incorporation into DNA was significantly increased by 100 and 1000 ng / ml MK. 1000 ng / ml MK enhanced BrdU incorporation level to 125% of control.
[0045] 調べた PF試料は全て、アツセィ限度を超える検出可能な濃度の MKを含んでいた。 [0045] All PF samples examined contained detectable concentrations of MK above the Atsey limit.
軽度の子宫内膜症(I期)を患う女性の PF中の MK濃度(0.93 ng/ml, 0.87〜1.17;中 央値、 IQR)は、健常な女性の MK濃度と実質的に同じであった (0.97 ng/ml, 0.67〜1 .27)。従って、本発明者らは、健常な女性と軽度の子宮内膜症を患う女性のデータと を合わせて、進行子宫内膜症(11、 III、および IV期)(r-ASRMスコア≥6)の女性のデ ータと比較した。図 2に示すように、進行子宫内膜症の女性の PFにおける MK濃度(1. 21 ng/ml, 0.80-2.27)は、健常な女性および軽度の子宫内膜症である女性の MK濃 度(0.96 ng/ml、 0.67〜1.26、 p〈0.05)より有意に高かった。 GnRHァゴ-スト治療を受 けている女性の MK濃度は、他の二群の濃度より有意に低力つた (pく 0.001)。 PF中の MK濃度を月経期に従って比較すると、黄体期の濃度(1.32 ng/ml, 0.72〜2.21)は、 卵胞期の濃度(0.95 ng/mU 0.68〜1.24、 p〈0.05)より有意に高かった。  MK concentrations in PF (0.93 ng / ml, 0.87 to 1.17; median, IQR) in women with mild eclampsia (stage I) were substantially the same as those in healthy women (0.97 ng / ml, 0.67-1.27). Therefore, we combined data from healthy women and women suffering from mild endometriosis to develop progressive epidural endometriosis (stage 11, III, and IV) (r-ASRM score ≥6) Compared to the data of other women. As shown in Figure 2, MK concentrations in PF (1.21 ng / ml, 0.80-2.27) in women with advanced endometriosis are MK concentrations in healthy women and women with mild endometriosis. (0.96 ng / ml, 0.67-1.26, p <0.05). MK concentrations in women receiving GnRHagost treatment were significantly lower than those in the other two groups (p 0.001). When MK concentrations in PF were compared according to menstrual period, the concentrations in the luteal phase (1.32 ng / ml, 0.72 to 2.21) were significantly higher than those in the follicular phase (0.95 ng / mU 0.68 to 1.24, p <0.05) .
図 3に示すように、 MK mRNAの発現は、 PBMC、腹膜、および子宫内膜組織におい て 320 bpで明確なバンドとして検出された。  As shown in FIG. 3, MK mRNA expression was detected as a clear band at 320 bp in PBMC, peritoneum, and eclampsia tissue.
[0046] 本発明において、本発明者らは、 MKが ESCの増殖を刺激することを示した。さらに 、 PF中の MKレベルは、軽度の子宫内膜症を患う女性または健常な女性と比較して、 進行子宮内膜症の女性では増加した。 興味深いことに、子宮内膜症の女性における正所子宮内膜細胞は、健常な女性と 比較して高いレベルの MKを発現する(Chungら、 2002) oしかし、同時に、異所子宫 内膜病変は、正所子宮内膜と比較して MKの有意に低いレベルを発現することも証 明された(Chungら、 2002) o本発明者らの知見は、 MKが疾患の開始および進行の双 方を促進することを考慮すれば、このジレンマを解決するために何らかの役に立つか も知れない。特に、異所子宫内膜以外の他の起源に由来する MKは、疾患の進行に 関与する可能性があるが、正所子宮内膜において増カロした MKは非常に初期段階の 子宮内膜症における逆流子宮内膜細胞の定着および生存に役立つ可能性がある。 [0046] In the present invention, the inventors have shown that MK stimulates ESC proliferation. In addition, MK levels in PF were increased in women with advanced endometriosis compared to women with mild eclampsia or healthy women. Interestingly, orthotopic endometrial cells in women with endometriosis express higher levels of MK compared to healthy women (Chung et al., 2002). Has also been demonstrated to express significantly lower levels of MK compared to the orthotopic endometrium (Chung et al., 2002). It may be useful to solve this dilemma if we consider promoting it. In particular, MKs derived from other sources other than ectopic endometrium may be involved in disease progression, but MK increased in orthotopic endometrium is very early stage of endometriosis May help in the establishment and survival of reflux endometrial cells.
[0047] 最近の報告から、マウスの腹腔損傷モデルにぉ 、て MKが腹腔内癒着を刺激する ことが示されている(Inohら、 2004) o子宫内膜症は疾患の進行に伴って腹腔内癒着 を形成しやすいことを考慮すると、 MKはまた癒着の形成にも関与する可能性がある。 [0047] A recent report shows that MK stimulates intraperitoneal adhesions in a murine abdominal injury model (Inoh et al., 2004). Given that they tend to form internal adhesions, MK may also be involved in adhesion formation.
PFにおける MK起源は広く分布しているように思われる。本発明者らの RT-PCR分 析は、 MKが PBMC、腹膜および子宮内膜組織において発現されることを証明した。さ らに、排卵時に放出される卵胞液中の MKは、卵胞液が大量の MKを含むことから、 P Fにおける MKの一部を構成すると仮定される(Ohyamaら、 1994)。卵胞液中の MK濃 度が高!、ことは、 PF中の MKが卵胞期と比較して黄体期では増加して 、ると 、う知見 を部分的に説明する可能性がある。  The origin of MK in PF seems to be widely distributed. Our RT-PCR analysis demonstrated that MK is expressed in PBMC, peritoneum and endometrial tissue. Furthermore, the MK in the follicular fluid released during ovulation is assumed to constitute part of the MK in PF because the follicular fluid contains a large amount of MK (Ohyama et al., 1994). The high MK concentration in the follicular fluid! May partially explain the finding that the MK in PF is increased in the luteal phase compared to the follicular phase.
[0048] 本研究における顕著な知見は、 GnRHa治療を受けている女性の MKレベルは、進 行子宮内膜症および軽度の子宮内膜症を患う女性または健常な女性と比較して有 意に低いということである。エストラジオールは、子宫内膜上皮細胞において MK mR NA発現を誘導することが報告された(Zhangら、 1995) 0 MK遺伝子(Ueharaら、 1992) のプロモーター領域に存在する 、くつかの ERE半パリンドロームモチーフ(Katoら、 19 92)が、エストラジオールの作用を引き起こすことが示唆された (Zhangら、 1995)。 GnR Ha治療によって誘導される低エストロゲン状態は、様々な細胞において MKの遺伝子 転写を抑制する可能性がある。さらに、 GnRHa治療によって引き起こされた無排卵は 、 PFにおける MKレベルを抑制して、腹腔への卵胞液の流入を妨害するように関与し うる。 [0048] Notable findings in this study indicate that MK levels in women receiving GnRHa treatment are significantly higher compared to women with advanced endometriosis and mild endometriosis or healthy women That is low. Estradiol has been reported to induce MK mRNA expression in eclamptic intimal epithelial cells (Zhang et al., 1995). 0 Several ERE half palindromes present in the promoter region of the MK gene (Uehara et al., 1992). It was suggested that the motif (Kato et al., 19 92) causes the action of estradiol (Zhang et al., 1995). The hypoestrogenic state induced by GnR Ha treatment may repress MK gene transcription in various cells. In addition, anovulation caused by GnRHa treatment may be involved in suppressing the influx of follicular fluid into the peritoneal cavity by suppressing MK levels in PF.
[0049] MKは、線維芽細胞、腫瘍細胞、およびケラチノサイトを含む 、くつかの細胞を増殖 させることが示されている (Inazumiら、 1997 ;Muramatsu and Muramatsu、 1991 ;Mura matsuら、 1993)。し力し、 MKの作用機序は、増殖因子シグナルを伝達することができ る明確に定義された MK受容体がないことから、あまり明確ではない(Muramatsu、 200 2)。このように、 MKが子宫内膜細胞を刺激するメカニズムを解明することは現時点で は困難であるように思われる。とは言え、 MKが疾患の発症を刺激することを考慮すれ ば、 MK阻害剤により、子宮内膜症を治療することができると考えられる。マウス直腸 癌細胞では、 MKに対するアンチセンスオリゴヌクレオチドは、腫瘍の形成を抑制する ことが示されている(Takeiら、 2001)。 [0049] MK grows several cells, including fibroblasts, tumor cells, and keratinocytes (Inazumi et al., 1997; Muramatsu and Muramatsu, 1991; Mura matsu et al., 1993). However, the mechanism of action of MK is less clear because there is no clearly defined MK receptor capable of transmitting growth factor signals (Muramatsu, 2002). Thus, elucidating the mechanism by which MK stimulates eclampsia intimal cells appears to be difficult at this time. However, given that MK stimulates the onset of the disease, endometriosis may be treated with MK inhibitors. In mouse rectal cancer cells, antisense oligonucleotides against MK have been shown to suppress tumor formation (Takei et al., 2001).
産業上の利用可能性 Industrial applicability
本発明では、子宫内膜細胞が、子宫内膜細胞由来の MKのみならず、他の起源か らの MKによって刺激されることを初めて明らかにした。さらに、 PF中の MKレベルは、 軽度の子宮内膜症を有するまたは子宮内膜症を有しない女性と比較して、進行子宮 内膜症の女性では増加することを証明し、 PF中の MKが子宫内膜症の発症や進行に 関与することを示した。  In the present invention, it has been clarified for the first time that eclampsia endometrial cells are stimulated not only by MK derived from eclampsia endometrial cells but also by MKs from other sources. Furthermore, MK levels in PF have been shown to increase in women with advanced endometriosis compared to women with mild endometriosis or no endometriosis, and MK levels in PF Have been shown to be involved in the development and progression of eclampsia.
以上の知見に基づき、 MKをターゲットとした子宮内膜症を予防または治療するた めの薬剤、並びに MKの発現を指標とした子宮内膜症の検査方法および検査薬を提 供することができる。  Based on the above findings, it is possible to provide a drug for preventing or treating endometriosis targeting MK, and an endometriosis test method and test drug using MK expression as an index.

Claims

請求の範囲 The scope of the claims
[1] ミツドカイン遺伝子の発現を抑制する化合物を有効成分とする、子宮内膜症を予防ま たは治療するための薬剤。  [1] A drug for preventing or treating endometriosis, comprising as an active ingredient a compound that suppresses expression of a mitdocaine gene.
[2] ミツドカイン遺伝子の発現を抑制する化合物がオリゴヌクレオチドである、請求項 1に 記載の薬剤。  [2] The drug according to claim 1, wherein the compound that suppresses expression of the mitdocaine gene is an oligonucleotide.
[3] ミツドカイン遺伝子の発現を抑制し得るオリゴヌクレオチドが以下の(a)または (b)であ る、請求項 2に記載の薬剤。  [3] The drug according to claim 2, wherein the oligonucleotide capable of suppressing the expression of the mitodocaine gene is the following (a) or (b):
(a)ミツドカイン遺伝子のアンチセンスオリゴヌクレオチド  (a) An antisense oligonucleotide of the mitdocaine gene
(b)ミツドカイン遺伝子を標的とする dsRNA  (b) dsRNA targeting mitodocaine gene
[4] ミツドカインタンパク質の機能を抑制する化合物を有効成分とする、子宮内膜症を予 防または治療するための薬剤。  [4] A drug for preventing or treating endometriosis, comprising as an active ingredient a compound that inhibits the function of mitodocaine protein.
[5] ミツドカインタンパク質の機能を抑制する化合物がミツドカインに結合する抗体である[5] A compound that suppresses the function of mitdocaine protein is an antibody that binds to mitdocaine
、請求項 4に記載の薬剤。 The drug according to claim 4.
[6] 被検者より採取した試料中のミツドカイン遺伝子の発現を測定する工程を含む、子宮 内膜症の検査方法。 [6] A method for examining endometriosis, comprising a step of measuring the expression of a mitdocaine gene in a sample collected from a subject.
[7] 被検者より採取した試料が腹腔液、腹腔骨髄由来細胞、腹膜組織または子宮内膜 組織である、請求項 6に記載の検査方法。  7. The examination method according to claim 6, wherein the sample collected from the subject is peritoneal fluid, peritoneal bone marrow derived cells, peritoneal tissue or endometrial tissue.
[8] ミツドカイン遺伝子発現の測定がミツドカインタンパク質またはミツドカインタンパク質を コードした mRNAを基に測定される、請求項 6または 7に記載の検査方法。 [8] The test method according to claim 6 or 7, wherein the measurement of mitdocaine gene expression is performed based on mitodocaine protein or mRNA encoding mitdocaine protein.
[9] 抗ミツドカイン抗体を含む子宮内膜症検査用試薬。 [9] A reagent for endometriosis testing comprising an anti-mitodocaine antibody.
[10] ミツドカイン遺伝子領域にハイブリダィズし、少なくとも 15ヌクレオチドの鎖長を有する オリゴヌクレオチドを含む、子宮内膜症検査用試薬。  [10] A reagent for endometriosis testing, comprising an oligonucleotide that hybridizes to a mitodocaine gene region and has a chain length of at least 15 nucleotides.
PCT/JP2005/014559 2004-08-13 2005-08-09 Remedy for endometriosis WO2006016571A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2004236085A JP2007297282A (en) 2004-08-13 2004-08-13 Influence caused by midkine when endometriosis appear
JP2004-236085 2004-08-13

Publications (1)

Publication Number Publication Date
WO2006016571A1 true WO2006016571A1 (en) 2006-02-16

Family

ID=35839340

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2005/014559 WO2006016571A1 (en) 2004-08-13 2005-08-09 Remedy for endometriosis

Country Status (2)

Country Link
JP (1) JP2007297282A (en)
WO (1) WO2006016571A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008059877A1 (en) 2006-11-14 2008-05-22 Ribomic Inc. Aptamer against midkine and use thereof
WO2008059616A1 (en) 2006-11-14 2008-05-22 Medical Therapies Limited Antibody recognizing c-domain of midkine
WO2023002039A1 (en) * 2021-07-23 2023-01-26 Fundació Hospital Universitari Vall D'hebron - Institut De Recerca Biomarkers for endometrial cancer

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2881467B1 (en) 2012-07-30 2018-10-31 National University Corporation Nagoya University Monoclonal antibody against human midkine
CN114144675A (en) * 2019-07-22 2022-03-04 豪夫迈·罗氏有限公司 S100A9 as a blood biomarker for non-invasive diagnosis of endometriosis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002085058A (en) * 2000-09-07 2002-03-26 Meiji Milk Prod Co Ltd Monoclonal antibody against human mk

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002085058A (en) * 2000-09-07 2002-03-26 Meiji Milk Prod Co Ltd Monoclonal antibody against human mk

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Pleiotrophin (PTN) and midkine(MK) mRNA expression in eutopic and ectopic endometrium in advanced stage endometriosis.", MOLECULAR HUMAN REPRODUCTION., vol. 8, no. 4, 2002, pages 350 - 355, XP002991729 *
DATABASE CA [online] HIROTA H. ET AL: "Possible implication of midkine in the development of endometriosis.", XP002991731, accession no. stn Database accession no. (143:42475) *
HUMAN REPRODUCTION., vol. 20, no. 4M, 2005, pages 1084 - 1089 *
KENJI KADOMATSU ET AL: "Midkine and pleiotrophin in neural development and cancer.", CANCER LETTERS., vol. 204, February 2004 (2004-02-01), pages 127 - 143, XP002991730 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008059877A1 (en) 2006-11-14 2008-05-22 Ribomic Inc. Aptamer against midkine and use thereof
WO2008059616A1 (en) 2006-11-14 2008-05-22 Medical Therapies Limited Antibody recognizing c-domain of midkine
US8080649B2 (en) 2006-11-14 2011-12-20 Ribomic Inc. Aptamer against midkine and use thereof
EP2803674A2 (en) 2006-11-14 2014-11-19 Medical Therapies Limited Antibody recognizing C-domain of midkine
WO2023002039A1 (en) * 2021-07-23 2023-01-26 Fundació Hospital Universitari Vall D'hebron - Institut De Recerca Biomarkers for endometrial cancer

Also Published As

Publication number Publication date
JP2007297282A (en) 2007-11-15

Similar Documents

Publication Publication Date Title
JP6404208B2 (en) Methods of prognosis, diagnosis and treatment of idiopathic pulmonary fibrosis
AU2011311452B2 (en) Method for the diagnosis, prognosis and treatment of breast cancer metastasis
US20220308054A1 (en) Methods of determining prognosis of sepsis and treating same
US7504211B2 (en) Methods of using EPHA2 for predicting activity of compounds that interact with and/or modulate protein tyrosine kinases and/or protein tyrosine kinase pathways in breast cells
TWI359026B (en) Pharmaceutical composition for the osteoclast rela
KR102373754B1 (en) Therapeutic agents for the treatment of diseases associated with undesired cell proliferation
JP2009535642A (en) Method for diagnosing and predicting non-alcoholic steatohepatitis (NASH)
US8741868B2 (en) Pharmaceutical composition including an HIF-2 alpha inhibitor as an active ingredient for preventing or treating arthritis
WO2006016571A1 (en) Remedy for endometriosis
WO2006063462A1 (en) Polynucleotides and polypeptide sequences involved in the process of bone remodeling
US20090274619A1 (en) Tumor vasculature markers and methods of use therof
KR20210111176A (en) Composition for diagnosing or treating drug resistant cancer
KR101416147B1 (en) Use of ADCY3 for the Diagnosis and Treatment of Gastric Cancer
EP2491396A2 (en) Assessment of solid tumor burden
EP3936149A1 (en) Therapeutic and prophylactic agent for glioma, brain tumor malignancy marker, brain tumor prognostic marker, method for determining malignancy and prognosis of brain tumor and antibody inhibiting tumor proliferation
KR102615797B1 (en) Method for predicting the efficacy of treatment effects in glioblastoma by T cells
KR102436301B1 (en) A composition for diagnosing or treating fibrosis
KR102330101B1 (en) The use of MLC1 for the diagnosis, prevention and treatment of epilepsy and associated cognitive dysfunction
CN109628588B (en) Osteoarthritis disorders screening gene PRXL2A and ACTR8 and application thereof
KR102015527B1 (en) Genetic mutation of desmoglein 3 and use thereof
KR20130047874A (en) Use of fibrinogen as a bone loss diagnostic marker
EP3690445A1 (en) Pharmaceutical composition for preventing or treating neurodegenerative disease comprising nckap1 protein or gene encoding same
WO2004009033A2 (en) Methods of modulating proliferative conditions
KR20200112069A (en) Use of Jmjd2b inhibitors for preventing or treating of bone diseases
JP2008263851A (en) Treatment target of heatstroke

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 BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP