WO2019026918A1 - Biomarkers from skin - Google Patents

Biomarkers from skin Download PDF

Info

Publication number
WO2019026918A1
WO2019026918A1 PCT/JP2018/028702 JP2018028702W WO2019026918A1 WO 2019026918 A1 WO2019026918 A1 WO 2019026918A1 JP 2018028702 W JP2018028702 W JP 2018028702W WO 2019026918 A1 WO2019026918 A1 WO 2019026918A1
Authority
WO
WIPO (PCT)
Prior art keywords
measurement value
subject
skin
group
col5a11
Prior art date
Application number
PCT/JP2018/028702
Other languages
French (fr)
Japanese (ja)
Inventor
匠徳 佐藤
Original Assignee
株式会社国際電気通信基礎技術研究所
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 株式会社国際電気通信基礎技術研究所 filed Critical 株式会社国際電気通信基礎技術研究所
Priority to JP2019534538A priority Critical patent/JP7445288B2/en
Publication of WO2019026918A1 publication Critical patent/WO2019026918A1/en

Links

Images

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • 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
    • 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

Definitions

  • the present invention relates to predicting renal disease using a skin-derived sample, predicting metabolic abnormality of phosphorus, predicting activation of FGF23 in a subject, and active ingredients for suppressing functional expression of FGF23.
  • the present invention relates to screening of candidate substances, screening of candidate active ingredients for activating the function of FGF23, and predicting the involvement of FGF23 in diseases.
  • Diseases may or may not be reversibly treatable (i.e., irreversible). It is essential to maintain good health that during the reversible condition, the abnormality is detected and treated promptly or prevented from becoming even such condition. Also, even in reversible conditions, early detection of the disease leads directly to milder treatment regimens, shorter treatment periods, and better prognosis health.
  • abnormalities in one organ or tissue lead to diseases in other organs (generally called complications). In such diseases, it is essential to prevent the disease of one organ or tissue from being caused by another organ or tissue as early as possible.
  • ESKD end-stage renal failure patients
  • CKD Chronic kidney disease
  • FGF23 is a hormone that lowers the level of phosphorus in blood, and is known to lower the level of phosphorus in blood by suppressing the reabsorption of phosphorus in the renal proximal tubule and the absorption of phosphorus from the intestinal tract. There is. It is also known that elevated levels of FGF23 cause bone-mineral metabolism disorder (CKD-MBD) associated with chronic kidney disease.
  • CKD-MBD bone-mineral metabolism disorder
  • Lysaght MJ J Am Soc Nephrol. 2002 Jan; 13 Suppl 1; S 37-40.
  • An object of the present invention is to predict kidney disease using a skin-derived sample.
  • An object of the present invention is to predict metabolic disorders of phosphorus using a skin-derived sample.
  • the present invention makes it one task to predict the activation of FGF23 in a subject.
  • One object of the present invention is to screen candidate substances of the active ingredient for suppressing the functional expression of FGF23.
  • One object of the present invention is to screen candidate substances for active ingredients for activating the function of FGF23.
  • the present invention makes it one task to predict the involvement of FGF23 in diseases.
  • Item 1 Apparatus for predicting renal disease in a subject having the following means: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of Prediction means for predicting the renal disease based on the measurement values acquired by the acquisition means.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7
  • the device according to item 1 which is at least one selected from the group consisting of: Col1a1 and Defb8.
  • the prediction means compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is outside the range of the reference value.
  • Item 5 A program that, when executed on a computer, causes the computer to perform the following processing to predict renal disease in a subject: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of The prediction process which predicts the said renal disease based on the measured value acquired by the said acquisition process.
  • Item 6 A program that, when executed on a computer, causes the computer to perform the following processing to predict renal disease in a subject: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of The prediction process which predicts the said renal disease based on the measured value acquired by the said acquisition process.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 6.
  • Item 7 In the prediction process, the measured value is compared with a predetermined reference value, and if the measured value is out of the range of the reference value, the subject is determined to have a renal disease.
  • a method of predicting renal disease in a subject comprising the following steps: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of A prediction step of predicting the renal disease based on the measurement value acquired in the acquisition step.
  • Item 10 From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject. Obtaining a measurement value of at least one mRNA selected from the group consisting of A prediction step of predicting the renal disease based on the measurement value acquired in the acquisition step.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 10.
  • the prediction step compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is out of the range of the reference value. The method described in. Item 12.
  • Item 13 Apparatus for predicting abnormal phosphorus metabolism in a subject, having the following means: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of Prediction means for predicting the phosphorus metabolism abnormality based on the measurement value acquired by the acquisition means.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 14.
  • the device according to item 13 wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
  • the prediction means compares the measured value with a predetermined reference value, and when the measured value is out of the range of the reference value, predicts that the subject has a phosphorus metabolism abnormality, or The device according to 14.
  • Item 17 A program that, when run on a computer, causes the computer to perform the following processing for predicting phosphorus metabolism abnormality in a subject: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of The prediction process which predicts the said phosphorus metabolism abnormality based on the measured value acquired by the said acquisition process.
  • Item 18 A program that, when run on a computer, causes the computer to perform the following processing for predicting phosphorus metabolism abnormality in a subject: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of The prediction process which predicts the said phosphorus metabolism abnormality based on the measured value acquired by the said acquisition
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 Item 18.
  • Item 19 In the prediction processing, the measured value is compared with a predetermined reference value, and when the measured value is out of the range of the reference value, it is predicted that the subject has a phosphorus metabolism abnormality, or The program described in 18. Item 20.
  • Item 21 A method of predicting abnormal phosphorus metabolism in a subject comprising the following steps: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of A prediction step of predicting the phosphorus metabolism abnormality based on the measurement value acquired in the acquisition step. Item 22.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 22.
  • the prediction step compares the measured value with a predetermined reference value, and predicts that the subject has a phosphorus metabolism abnormality if the measured value is out of the range of the reference value.
  • Item 25 Device for predicting FGF23 activation in a subject, having the following means: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of A prediction unit that predicts the activation of the FGF23 based on the measurement values acquired by the acquisition unit. Item 26.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1
  • the prediction means compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or Item 25.
  • FGF23 is activated in the subject, provided that at least one biomarker selected from the group consisting of Aldh112, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the reference value range, Or the apparatus described in 26. Item 28.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 29.
  • the measured value is compared with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or Item 28.
  • Item 31. A method of predicting FGF23 activation in a subject comprising the steps of: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of A prediction step of predicting activation of the FGF23 based on the measurement value acquired in the acquisition step.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 32.
  • the prediction step compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, Il22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or It is predicted that FGF23 is activated in the subject, provided that at least one biomarker selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value, Or the method according to 32.
  • Item 34 Screening apparatus for candidate active ingredients for suppressing the functional expression of FGF23, having the following means: Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin; A first measurement value acquiring unit for acquiring a measurement value of a biomarker protein of at least one test substance-treated sample and / or a measurement value of mRNA of the protein; A determination means for determining that the test substance is a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquisition means. Item 35.
  • a second measurement value acquiring means for acquiring a measurement value of a corresponding biomarker protein in at least one unprocessed sample to be selected and / or a measurement value of mRNA of said protein, Measured value comparison means for comparing the measured value of the test substance-treated sample with the measured value of the untreated sample, And have 35.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7
  • the device according to Item 35 which is at least one selected from the group consisting of: Col1a1 and Defb8. Item 37.
  • a screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for suppressing the functional expression of FGF23: Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin
  • a first measurement value acquisition process for acquiring a measurement value of a biomarker protein in at least one test substance-treated sample and / or a measurement value of mRNA of said protein, and The determination process which determines that the said test substance is a candidate substance of an active ingredient based on the measured value acquired by said 1st measured value acquisition process.
  • the test substance comprises a sample collected from the skin of a subject not treated with the test substance (except for human beings), a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin
  • a second measurement value acquisition process for acquiring a measurement value of a corresponding biomarker protein of at least one untreated sample selected from a group and / or a measurement value of mRNA of said protein,
  • a measured value comparison process comparing the measured value of the test substance-treated sample with the measured value of the untreated sample;
  • the test substance is determined to be a candidate substance of the active ingredient based on the comparison result of the measurement value comparison process.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7
  • the screening method of the candidate substance of the active ingredient for suppressing the functional expression of FGF23 including the following processes: (I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein, (II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I). Item 41.
  • the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
  • step (I) Before the step (I), (I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance, (Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in the step (i); (Iii) a step of recovering protein and / or mRNA from the sample obtained in the step (ii)
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1
  • the method according to any one of Items 40 to 42 which is at least one selected from the group consisting of: Col1a1 and Defb8. Item 44.
  • Screening apparatus for candidate active ingredients for activating the function of FGF23 having the following means: Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample) A first measurement value acquiring unit for acquiring a measurement value of a protein and / or a measurement value of mRNA of the protein; A determination means for determining that the test substance is a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquisition means. Item 45.
  • a second measurement value acquisition unit for acquiring the measurement value of the protein and / or the mRNA of the protein, and Measured value comparison means for comparing the measured value of the test substance-treated sample with the measured value of the untreated sample, And have The screening device according to Item 44, wherein the determination means determines that the test substance is a candidate substance of the active ingredient based on the comparison result of the measurement value comparison means.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7
  • the device according to Item 45 which is at least one selected from the group consisting of, Col1a1 and Defb8. Item 47.
  • a screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for activating the function of FGF23: Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample)
  • a first measurement value acquisition process for acquiring a measurement value of a protein and / or a measurement value of mRNA of the protein, and The determination process which determines that the said test substance is a candidate substance of an active ingredient based on the measured value acquired by said 1st measured value acquisition process.
  • the bio of a sample collected from the skin of a subject (except for human beings) not treated with the test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (untreated sample) A second measurement value acquisition process for acquiring a measurement value of a marker protein and / or a measurement value of mRNA of said protein, and A measured value comparison process comparing the measured value of the test substance-treated sample with the measured value of the untreated sample;
  • the program according to Item 47 wherein in the determination process, the test substance is determined to be a candidate substance of the active ingredient based on a comparison result of the measurement value comparison process.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7
  • the screening method of the active substance candidate substance for activating the function of FGF23 including the following processes: (I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein, (II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I). Item 51.
  • the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
  • step (I) Before the step (I), (I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance, (Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in the step (i); (Iii) a step of recovering protein and / or mRNA from the sample obtained in the step (ii) 52.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1
  • the method according to any one of Items 40 to 42 which is at least one selected from the group consisting of: Col1a1 and Defb8. Item 54.
  • Predictor for predicting the involvement of FGF23 in disease comprising the following means: First measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition means A determination means for determining that FGF23 is involved in the disease based on the measurement value obtained by Item 55.
  • a second measurement value acquisition unit for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein; Measurement value comparison means for comparing the measurement value acquired by the first measurement value acquisition means with the measurement value acquired by the second measurement value acquisition means; And have 56.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 56.
  • the device according to item 55 which is at least one selected from the group consisting of: Col1a1 and Defb8. Item 57.
  • a prediction program which, when executed on a computer, causes the computer to carry out the following processing for predicting the involvement of FGF23 in a disease:
  • a first measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition process
  • the decision processing which determines that FGF23 is concerned in the said disease based on the measured value acquired by b. Item 58.
  • a second measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein,
  • a measurement value comparison process comparing the measurement value acquired in the first measurement value acquisition process and the measurement value acquired in the second measurement value acquisition process;
  • the program according to Item 57 wherein in the determination processing, it is determined that FGF23 is involved in the disease based on the result of the comparison.
  • Item 59 wherein in the determination processing, it is determined that FGF23 is involved in the disease based on the result of the comparison.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 60.
  • Methods of predicting FGF23's involvement in disease comprising the following steps: (I) obtaining a measured value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measured value of mRNA of said protein, (II) A step of determining that FGF23 is involved in the disease based on the measurement value obtained in the step (I). Item 61.
  • Step (II) is a step of determining that FGF23 is involved in the disease based on the result of the comparison.
  • the biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1
  • the method according to any one of items 60 to 61, which is at least one selected from the group consisting of: Col1a1 and Defb8. Item 63.
  • a skin biomarker for predicting involvement of renal function, abnormal phosphorus metabolism, or FGF23 comprising at least one selected from the group consisting of
  • the present invention as an effect, it is possible to detect a decrease in renal function using a skin-derived sample.
  • the present invention can predict, as one effect, a metabolic abnormality of phosphorus using a skin-derived sample.
  • the present invention can predict the activation of FGF23 in a subject using a skin-derived sample as one effect.
  • the present invention can screen for a candidate substance of an active ingredient for suppressing functional expression of FGF23 using skin derived specimen as one effect.
  • the present invention can screen for a candidate substance of an active ingredient for activating the function of FGF23 using skin derived specimen as one effect.
  • the present invention can, as one effect, predict the involvement of FGF23 in disease using skin-derived specimens.
  • FIG. 1 is a schematic view of a system 100 according to first to third embodiments of the present invention. It is a figure showing hardware constitutions of system 100 concerning the 1st-the 3rd embodiment of the present invention.
  • FIG. 6 is a block diagram for explaining the functions of arithmetic units 1 to 3 according to first to third embodiments of the present invention of the present invention. It is a flow chart which shows operation of arithmetic unit 1 concerning the 1st mode of the present invention. It is a flow chart which shows operation of arithmetic unit 2 concerning the 2nd mode of the present invention. It is a flow chart which shows operation of arithmetic unit 3 concerning the 3rd mode of the present invention.
  • FIG. 1 is a schematic view of a system 100 according to first to third embodiments of the present invention. It is a figure showing hardware constitutions of system 100 concerning the 1st-the 3rd embodiment of the present invention.
  • FIG. 6 is a block diagram for explaining the functions of arithmetic units 1 to
  • FIG. 10 is a schematic view of a system 200 according to the fourth and fifth embodiments of the present invention. It is a figure which shows the hardware constitutions of the system 200 which concerns on the 4th, 5th embodiment of this invention. It is a block diagram for demonstrating the function of the screening apparatuses 4 and 5 which concern on the 4th, 5th embodiment of this invention of this invention. It is a flowchart which shows operation
  • FIG. 16 is a schematic view of a system 300 according to a sixth embodiment of the present invention.
  • FIG. 17A shows the sequences of gRNA and ssODNs.
  • FIG. 17B shows the genotype of mutant mice. The expression of biomarkers in the skin of UNx / HPi, WT3W / HP1W, WT3W / LP1W, WT3W / ND1W mouse models is shown.
  • the present invention includes the first embodiment for solving the problem of predicting kidney disease using a skin-derived sample.
  • the present invention includes a second embodiment for solving the problem of predicting metabolic abnormality of phosphorus using a skin-derived sample.
  • the present invention includes the third embodiment for solving the problem of predicting the activation of FGF23 in a subject.
  • the present invention includes the fourth embodiment for solving the problem of screening a candidate substance of an active ingredient for suppressing the functional expression of FGF23.
  • the present invention includes the fifth embodiment for solving the problem of screening candidate substances for active ingredients for activating the function of FGF23.
  • the present invention includes the sixth embodiment for solving the problem of predicting the involvement of FGF23 in diseases.
  • the present invention includes the seventh embodiment of the test reagent and the test kit.
  • the present invention includes the eighth embodiment according to the skin biomarker.
  • renal disease is any abnormality or disease of the kidney causing renal function decline, and is not particularly limited as long as the kidney has some functional or physical disorder.
  • acute renal failure acute pyelonephritis, acute glomerulonephritis (glomerulonephritis associated with hemolytic streptococcal infection, rapidly progressive glomerulonephritis, etc.), renal disease associated with heart disease
  • Acute renal diseases such as acute diseases (type 1 cardiorenal syndrome etc.); chronic pyelonephritis, reflux nephropathy, interstitial nephritis, polycystic kidney disease, chronic glomerulonephritis (IgA nephropathy, systemic lupus erythematosus glomeruli Nephritis (Lupus Nephritis) etc.
  • Chronic disease among chronic kidney disease associated with heart disease Type 2 cardio-renal syndrome etc
  • Chronic nephritis such as diabetic n
  • kidney function reduction means, for example, at least one type of kidney disease marker (preferably urine) shown in the following Tables 1-1 to 1-4 which are generally measured by clinical examination in the case of human. The condition where the value is outside the standard value range).
  • kidney disease marker can be measured by a known method disclosed in the Clinical Test Method Revision Edition 32 (Kanai Masamitsu Edited by Kanahara Publishing Co., Ltd.) and the like.
  • acute renal failure is a disease in which renal function is rapidly reduced, for example, those in which the serum creatinine level rapidly increases to 2.0 to 2.5 mg / dl or more (based on the kidney If there is a decline in function, serum creatinine level increases by 50% or more from the previous level), or serum creatinine level is 0.5 mg / dl / day or more, and urea nitrogen is 10 mg / dl / day or more I say something that increases.
  • Acute renal failure includes (1) prerenal acute renal failure due to decreased renal blood flow, (2) renal acute renal failure with impaired renal parenchyma, and (3) renal failure after renal failure Although post-renal acute renal failure is included, preferred acute renal failure for application of the present invention is prerenal acute renal failure and renal acute renal failure, preferably prerenal acute renal failure.
  • Renal disease associated with heart disease is also called cardiorenal syndrome.
  • Cardiorenal syndromes including acute and chronic conditions, are classified into multiple types. Of these, types 1 and 2 start with heart disease, type 1 is acute cardiorenal syndrome, and type 2 is chronic cardiorenal syndrome. Type 1 may be triggered by ischemic heart disease or the like.
  • Type 1 cardiorenal syndrome refers to any heart disease resulting in one of the following stages 1-3: Stage 1: Serum creatinine level increases to about 1.5 to 1.9 times the standard value, or in the same individual, serum creatinine level increases by 0.3 mg / dl or more over the previous level, and urine volume Stage 2: The blood pressure is about 0.5 mL / kg / hour over 6 hours to 12 hours: The serum creatinine level increases to 2.0 to 2.9 times the standard value, and the urine volume is 0.
  • Stage 3 The condition of less than 5 mL / kg / hour continues: Does the serum creatinine level increase to about 3 times the standard value, or does the serum creatinine level increase by 4.0 mg / dL or more over the previous level in the same individual , Or if renal replacement therapy is started or eGFR drops to less than 35 mL / min / 1.73 m 2 in patients younger than 18 years, urine volume is 24 in addition to any of the above four conditions. 0.3 mL / kg / hour or more If the condition of less than hour continues or the condition of urinelessness continues for 12 hours or more.
  • chronic kidney disease means, when the subject is a human, according to “CKD Diagnosis Guide 2012 (The Japanese Society of Nephrology), renal disorders (urinary abnormalities such as proteinuria including micro albuminuria), Abnormal urinary sediment, abnormal imaging such as single kidney and multiple cystic kidney, decreased renal function such as increase in serum creatinine level, abnormal electrolyte such as hypokalemia due to renal tubular disorder, abnormal pathology in renal biopsy etc. Etc.), or estimated GFR (glomerular filtration rate) of 60 mL / min / 1.73 m 2 or less is a state in which renal function decline lasts for 3 months or more.
  • CKD Diagnosis Guide 2012 The Japanese Society of Nephrology
  • renal disorders urinary abnormalities such as proteinuria including micro albuminuria
  • Abnormal urinary sediment abnormal imaging such as single kidney and multiple cystic kidney
  • decreased renal function such as increase in serum creatinine level
  • abnormal electrolyte such as hypokalemia due to renal tubular disorder, abnormal pathology in
  • the estimated GFR (eGFR) can be calculated by the estimation formula (eGFR Creat) from the serum creatinine value shown in Table 2 below.
  • the serum cystatin C estimation formula (eGFRcys) can be applied.
  • chronic renal disease can be diagnosed when urine protein of 0.15 g / g Cr or more continues by urinalysis three months ago and recent.
  • chronic kidney disease it is possible to diagnose chronic kidney disease as a case where diabetes continues more than 3 months ago and 30 mg / g Cr or more continues in the recent albuminuria test.
  • eGFR in% (0.3 x height (m) / subject's serum Cr value) x 100
  • chronic kidney disease can be predicted from the average daily water intake or specific gravity of urine.
  • the severity of chronic kidney disease can be determined based on Table 3 below (Table 3 is Table 2 of “CKD Diagnosis Guide 2012”).
  • the disease associated with FGF23 is not limited as long as it is a disease that develops with overexpression of FGF23.
  • Hypophosphatemia rickets / osteomalacia autosomal dominant hypophosphatemia rickets / osteomalacia, autosomal recessive hypophosphatemia rickets / osteomalacia, X-chromosomal dominant hypophosphate Rickets / osteomalacia, neoplastic rickets / osteomalacia, secondary hyperparathyroidism, abnormal phosphorus metabolism in acute kidney disease or chronic kidney disease, bone-mineral metabolism disorder (CKD-MBD) Etc. can be mentioned.
  • CKD-MBD bone-mineral metabolism disorder
  • the "individual” is not particularly limited, but the individual includes humans and mammals other than humans. Examples of mammals other than human include cows, horses, sheep, goats, pigs, dogs, cats, rabbits, monkeys and the like. Preferred are humans, cats and dogs. Also, the age and gender of the individual do not matter.
  • the “subject” may or may not be an individual having renal function decline or a history of other renal diseases.
  • the subject may be an individual with symptoms such as polyuria, thirst, increased water intake, excessive gastric juice, vomiting, hematuria, general fatigue, or even an asymptomatic individual Good.
  • subjects include subjects suspected of having renal disorder or chronic kidney disease according to known diagnostic methods such as medical interviews, urinalysis, blood biochemistry, renal imaging, renal biopsy etc. Be
  • the skin-derived sample is not limited as long as it is derived from the skin of a living body.
  • the skin-derived sample also includes sweat, secretions from the skin, and the like.
  • the method of collecting the skin is also not limited, and examples thereof include a biopsy material, skin adhering to a perforated needle when a hole in a piercing is made, abrasion on the skin surface, and the like.
  • the sample may be fresh or stored.
  • it can be stored in a room temperature environment, a refrigerated environment or a frozen environment, but is preferably frozen.
  • Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1, Aldh1b1 shown in Table 4 are included. And at least one selected from the group consisting of Sparc, Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8, variants thereof, orthologs and the like.
  • At least one biomarker (referred to as group 1) is likely to be under the control of FGF23. Therefore, when the measured value of one group of biomarkers is out of the range of the reference value, for example, it can be predicted that FGF23 is involved in, for example, renal diseases and other diseases, or abnormal phosphorus metabolism.
  • At least one biomarker selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 is likely not to be regulated by FGF23. Therefore, if one group of biomarkers is out of the range of the reference value, and if the measured values of the two groups of biomarkers are within the range of the reference value, for example, renal disease or other diseases, or phosphorus metabolism In the abnormality, it can be predicted more reliably that FGF23 is involved.
  • FGF23 Whether or not FGF23 is involved is preferably determined in consideration of both the measured values of one group of biomarkers and the measured values of two groups of biomarkers.
  • the “measurement value of at least one type of protein selected from the group consisting of biomarkers” refers to a value reflecting the amount or concentration of at least one type of protein selected from the group consisting of biomarkers.
  • amount although it may be molar or mass, it is preferable to label by mass.
  • concentration when a value is expressed as “concentration”, it may be a molar concentration or a mass ratio (mass / volume) per a fixed volume of an analyte, but is preferably mass / volume.
  • the intensity of a signal such as fluorescence or luminescence may be used.
  • the “measured value of at least one mRNA selected from the group consisting of biomarkers” may be represented by the copy number (absolute amount) of biomarker mRNA present in a certain amount of sample, or ⁇ 2- It may be a value reflecting the expression level relative to the expression level of housekeeping genes such as microglobulin mRNA, GAPDH mRNA, Maea mRNA and ⁇ -actin mRNA. Moreover, it may be expressed by the intensity of signals such as fluorescence and luminescence.
  • the "predetermined reference value" of the measured value of the biomarker protein is based on the measured value of the biomarker protein in the sample of the individual who has developed renal function decline and / or the measured value of the biomarker protein in the sample of the healthy individual Reference value to be determined.
  • a measured value of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from a subject, and / or contained in a sample collected from the subject A group consisting of a measured value (also referred to as a test measured value) of at least one mRNA selected from the group consisting of the biomarkers, and a biomarker contained in a skin-derived sample collected from a healthy individual corresponding to the test measured value And / or a measured value of at least one mRNA selected from the group consisting of a biomarker included in a sample collected from the subject (also referred to as a healthy measurement value)
  • the reference value is determined by known methods on the basis of.
  • "corresponding" is intended to be a homogeneous biomarker.
  • the most accurately determined value is the sensitivity and / or specificity, the positive predictive value, the negative predictive value, the first quartile range of the quartile range, which are determined from the ROC curve depending on the purpose of the test. It can set suitably based on indices, such as the 2nd quartile (median value) and the 3rd quartile.
  • the highest measured value may be used as the reference value.
  • the reference value is determined based on the measured value of the biomarker protein in the sample of an individual who has developed renal function
  • the biotechniques in the samples of multiple individuals who have developed renal function have been described.
  • the lowest measurement value can be determined as the threshold value.
  • the reference value may be a measured value of the biomarker protein itself in a sample of a healthy individual, or an average, median or mode of measured values of a plurality of biomarker proteins of a healthy individual. it can.
  • a measurement value of a past biomarker protein (which may be a single value or an average value of a plurality of values, which is the same subject and obtained when the subject is in a healthy state) Values, modes, etc. may also be used.
  • the “reference value” of the measured value of the biomarker mRNA is also determined using the measured value of the biomarker mRNA instead of the measured value of the biomarker protein, as in the “reference value” of the measured value of the biomarker protein described above. can do.
  • the “range of reference value” means that the reference range is equal to or less than the reference value as long as the biomarker protein has a property that expression and activity increase with the decrease in renal function. If the biomarker protein is of a nature that decreases in expression and activity as renal function declines, the reference range is above the reference value.
  • Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Aldh1l2, and Col5a1 have decreased expression of renal function.
  • the expression of C1qtnf6, Sparc, Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8 decreases with the decrease in renal function.
  • the "healthy individual” is not particularly limited.
  • the term preferably refers to a human or non-human mammal described in the "individual” section, which does not show abnormal data in biochemical tests, blood tests, urine tests, serological tests, physiological tests and the like.
  • the age and sex of healthy individuals are not particularly limited.
  • the “plural samples” are two or more, preferably five or more, and more preferably ten or more. These may be specimens collected from different individuals, or may be a plurality of specimens of the same individual different in collection time.
  • Multiple values are measurements of biomarker protein of 2 or more, preferably 5 or more, more preferably 10 or more and biomarker mRNA.
  • the "plural individuals” are 2 or more, preferably 5 or more, more preferably 10 or more individuals.
  • the individual who obtains the measurement value of the biomarker protein and the measurement value of the biomarker mRNA to determine the reference value is not necessarily the same as the species, age, sex, etc. of the subject, but is preferably the same.
  • the individual is of the same age and / or the same gender as the subject.
  • the "anti-biomarker antibody” is not particularly limited as long as it specifically binds to at least one protein selected from the group consisting of the aforementioned biomarkers, and at least one protein selected from the group consisting of biomarkers or one of them Any of polyclonal antibodies, monoclonal antibodies, and fragments thereof (for example, Fab, F (ab) 2 etc.) obtained by immunizing animals other than human with an antibody as an antigen can be used. Also, the class and subclass of immunoglobulin are not particularly limited. It may also be a chimeric antibody. Furthermore, it may be scFv or the like.
  • biomarker protein used as an antigen used to produce an anti-biomarker antibody all or a part of at least one type of protein selected from the group consisting of the above-mentioned biomarkers can be mentioned.
  • the “biomarker mRNA detection nucleic acid” in the present invention is not limited as long as it contains at least one mRNA selected from the group consisting of the above-mentioned biomarkers, or a sequence specifically hybridizing with the reverse transcription product of the mRNA.
  • the detection nucleic acid may be DNA or RNA, and the nucleotides contained in the detection nucleic acid may be natural nucleotides or artificially synthesized nucleotides.
  • the length of the detection nucleic acid is not particularly limited. If the detection nucleic acid is used as a capture probe in a microarray or the like, the sequence hybridizing to the target nucleic acid is preferably about 100 mer, more preferably about 60 mer, and still more preferably It is about 30 mer.
  • the capture probe can be produced using a known oligonucleotide synthesizer or the like.
  • the capture probe may contain a sequence that does not hybridize to the target nucleic acid.
  • the sequence that hybridizes to the target nucleic acid is preferably about 50 mer, more preferably about 30 mer, and still more preferably about 15 to 25 mer It is.
  • the primers can be produced using a known oligonucleotide synthesizer or the like.
  • the primers may contain sequences that do not hybridize to the target nucleic acid.
  • the primer may be labeled with a fluorescent dye or the like.
  • RT-PCR can also use quantitative probes that are degraded during PCR reactions, which are used for real-time quantification of PCR products.
  • the quantitative probe is also not limited as long as it hybridizes to the target nucleic acid.
  • the quantitative probe is preferably a nucleic acid of about 5 to 20 mer, which contains a sequence that hybridizes to a target nucleic acid.
  • a fluorescent dye be labeled at one end of the quantitative probe, and a quencher of the fluorescent dye be labeled at the other end of the quantitative probe.
  • a method of obtaining a measured value of at least one type of protein selected from the group consisting of biomarkers in the present invention and at least one type of mRNA measurement value selected from the group consisting of biomarkers It is not limited as long as the measured value can be obtained. For example, it can be obtained according to the method described below.
  • an anti-biomarker antibody for capturing an antigen can be immobilized in advance on a solid phase such as a microplate to form a complex of the immobilized anti-biomarker antibody and a biomarker protein in a sample. . Measuring the amount or concentration of the biomarker protein contained in the sample by detecting the complex immobilized on the solid phase or the complex formed on the solid phase by a method known in the art Can.
  • the method of fixing the anti-biomarker antibody for antigen capture to the solid phase is not particularly limited. It can be carried out directly or indirectly via another substance using known methods. Examples of direct binding include physical adsorption and the like. Preferably, the anti-biomarker antibody can be physically bound to the microplate directly using an immuno plate or the like.
  • the material of the solid phase is not particularly limited, and examples thereof include polystyrene and polypropylene.
  • the shape of the solid phase is not particularly limited, and examples thereof include microplates, microtubes, test tubes and the like.
  • an operation of washing the solid phase may be included.
  • PBS containing a surfactant or the like can be used.
  • the detection of the complex is carried out using a detection anti-biomarker antibody labeled with a labeling substance, or an unlabeled anti-biomarker antibody and the unlabeled anti-biomarker antibody are bound to each other.
  • a detection anti-biomarker antibody labeled with a labeling substance or an unlabeled anti-biomarker antibody and the unlabeled anti-biomarker antibody are bound to each other.
  • it can be carried out using an anti-immunoglobulin antibody etc. labeled with a labeling substance that can be used, it is preferable to use a labeled detection anti-biomarker antibody.
  • the epitope in the biomarker protein of the detection anti-biomarker antibody and the epitope in the biomarker protein of the anti-biomarker antibody for antigen capture are different.
  • the labeling substance used for the detection anti-biomarker antibody or the labeled anti-immunoglobulin antibody is not particularly limited as long as a detectable signal is generated.
  • fluorescent substances, radioactive isotopes, enzymes and the like can be mentioned.
  • the enzyme alkaline phosphatase, peroxidase and the like can be mentioned.
  • fluorescent substances include fluorescein isothiocyanate (FITC), rhodamine, fluorescent dyes such as Alexa Fluor (registered trademark), fluorescent proteins such as GFP, and the like.
  • FITC fluorescein isothiocyanate
  • rhodamine fluorescent dyes such as Alexa Fluor (registered trademark)
  • fluorescent proteins such as GFP, and the like.
  • radioactive isotopes 125 I, 14 C, 32 P and the like can be mentioned.
  • alkaline phosphatase or peroxidase is preferable as a labeling substance.
  • the anti-biomarker antibody for detection is obtained by labeling the anti-biomarker antibody with the above-mentioned labeling substance by a labeling method known in the art. Moreover, you may label using a commercially available labeling kit etc. Also, the labeled immunoglobulin antibody may use the same method as the labeling of the anti-biomarker antibody, or a commercially available one may be used.
  • the measurement value of the biomarker contained in the sample can be obtained by detecting the signal generated by the labeled substance of the labeled anti-biomarker antibody contained in the complex.
  • detecting the signal includes qualitatively detecting the presence or absence of the signal, quantifying the signal intensity, and detecting the intensity of the signal semi-quantitatively.
  • Semi-quantitative detection means that the intensity of a signal is indicated stepwise such as “no signal generation”, “weak”, “medium”, “strong” and the like. In this step, it is preferable to detect the intensity of the signal quantitatively or semi-quantitatively.
  • the method of detecting a signal can use a well-known method.
  • a measurement method can be appropriately selected according to the type of signal derived from the above-mentioned labeling substance.
  • the labeling substance is an enzyme
  • the signal such as light or color generated by reacting a substrate for the enzyme may be measured by using a known device such as a luminometer or a spectrophotometer. it can.
  • the substrate for the enzyme can be appropriately selected from known substrates depending on the type of the enzyme.
  • CDP-Star registered trademark
  • 4-chloro-3- methoxyspiro [1,2-dioxetane-3,2 '-(5'-chloro) trixilo] is used as a substrate.
  • Chemiluminescent substrates such as [3. 3. 1.
  • the labeling substance is peroxidase, tetramethylbenzidine (TMB) etc. can be mentioned.
  • the labeling substance is a radioactive isotope
  • radiation as a signal can be measured using a known device such as a scintillation counter.
  • fluorescence as a signal can be measured using a known device such as a fluorescence microplate reader.
  • an excitation wavelength and a fluorescence wavelength can be suitably determined according to the kind of fluorescent substance used.
  • the detection result of the signal can be used as a measurement value of a biomarker protein.
  • the measured value of the signal intensity itself or a value calculated from the measured value of the signal intensity can be used as a measured value of the protein of the biomarker.
  • Obtaining a Measured Value of a Biomarker Gene In order to obtain a measured value of at least one mRNA selected from the group consisting of a biomarker (hereinafter sometimes abbreviated as "measured value of a biomarker mRNA" in the present specification)
  • known methods such as microarray method, RNA-seq analysis method, quantitative RT-PCR method can be used.
  • the probes used in the microarray method may be synthesized by using a self-selected probe or a known probe, or a commercially available microarray chip may be used.
  • RNA or mRNA extracted from the sample may be used.
  • Samples used for total RNA and mRNA extraction are collected from an individual and immediately subjected to RNA extraction, or collected from an individual and frozen immediately (preferably under an atmosphere of -196 ° C. or less (quenched in liquid nitrogen ) And stored at -80 ° C. or less until RNA extraction.
  • the method for extracting total RNA and mRNA from the sample is not particularly limited, and known extraction methods can be used.
  • the quantification by the microarray method can be performed according to a known method, and the expression amount of the biomarker mRNA may be expressed as a relative expression amount relative to the expression amount of a housekeeping gene, and is expressed as a measurement value of signal intensity of a fluorescent dye or the like be able to.
  • the expression level of the biomarker mRNA may be expressed as a relative expression level relative to the expression level of the housekeeping gene, or may be expressed as a measurement value of the intensity of a signal such as a fluorescent dye.
  • RNA extracted from a sample is fragmented, and this is used as a template to synthesize cDNA by reverse transcription reaction and create a library.
  • the nucleotide sequence of a fragment contained in each library is determined by a next-generation sequencer, the information is mapped to a reference gene sequence, and the amount of mRNA expression is represented as RPKM (Reads Per Killobases per Million).
  • RPKM may be expressed as the intensity of a signal such as a heat map.
  • the detection result of the above signal can be used as the expression level of biomarker mRNA.
  • the measured value of the signal intensity itself or a value calculated from the measured value of the signal intensity can be used as the expression level of the biomarker mRNA.
  • the signal intensity of the positive control sample As a value calculated from the measurement value of the signal intensity, for example, a value obtained by subtracting the measurement value of the signal intensity of the negative control sample from the measurement value of the signal intensity, the signal intensity of the positive control sample And the combination thereof.
  • negative control samples include samples of healthy persons.
  • the positive control sample includes a sample containing biomarker mRNA at a predetermined expression level.
  • the measurement value of may be obtained as a measurement value reflecting the function of each biomarker.
  • FIG. 1 is a schematic view of a system 100 according to first to third embodiments of the present invention
  • FIG. 2 is a block diagram showing a hardware configuration of the system 100.
  • the system 100 refers to arithmetic devices having the same hardware configuration and having different functional configurations as described later, and therefore, the system 100 generically refers to the arithmetic devices having these different functional configurations. For this purpose, it is provided with “calculation devices 1, 2, 3”, the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
  • the arithmetic units 1, 2, and 3 are, for example, general-purpose personal computers, and include a CPU 101 that performs data processing to be described later, a memory 102 used for a data processing work area, and a recording unit 103 that records processing data.
  • a bus 104 for transmitting data between the respective units, and an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device are provided.
  • the input unit 8 and the display unit 9 are connected to the arithmetic devices 1, 2, 3, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like.
  • the input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel.
  • the arithmetic units 1, 2, and 3 do not have to be integrated units, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate
  • the arithmetic devices 1, 2, 3, the measuring device 5a, and the measuring device 5b do not necessarily need to be disposed at one place, but a system in which devices provided at different places are communicably connected by a network. May be configured.
  • the processing performed by arithmetic units 1, 2, 3 is actually based on the program stored in recording unit 103 or memory 102, and CPU 101 of arithmetic units 1, 2, 3 actually Means the process to be performed.
  • the CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
  • the measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53.
  • the sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased.
  • the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done.
  • measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
  • measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
  • the measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54.
  • the sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
  • the measuring devices 5a and 5b are connected to the computing devices 1, 2 and 3 by wire or wireless.
  • the measuring device 5a performs A / D conversion of the measured value of the protein, and transmits it as digital data to the computing devices 1, 2, 3.
  • the measuring device 5b A / D converts the measured value of mRNA, and transmits it as digital data to the computing devices 1, 2, 3.
  • the computing devices 1, 2, 3 can acquire the measured value of the protein and the measured value of the mRNA as digital data that can be processed.
  • the measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the computing devices 1, 2, 3 can acquire the measurement value of the kidney disease marker as digital data.
  • the method for predicting renal disease in a subject in this embodiment is a measurement value of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from the subject And / or an acquisition step of acquiring a measurement value of at least one mRNA selected from the group consisting of biomarkers contained in a specimen collected from the subject, and the measurement value acquired in the acquisition step And a prediction step of predicting the renal disease on the basis of the above.
  • the measured value obtained by the above-mentioned “2. Method of obtaining each measured value” is compared with a predetermined reference value corresponding to each biomarker of the measured value, and the measured value is obtained. Is determined to be outside the reference value range, it can be determined that the subject has a renal disease.
  • the measured value of one group of biomarkers when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is involved in the renal disease. In addition, when the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the renal disease is related to FGF23. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, the renal disease involves FGF23. You may decide to
  • the process may be performed by the CPU 101 described later, or may be performed by an examiner.
  • the method for predicting renal disease may include a step of performing treatment (eg, diet therapy such as a low phosphorus diet) to improve the renal disease when it is predicted to be a renal disease.
  • treatment eg, diet therapy such as a low phosphorus diet
  • the first embodiment includes a device for predicting renal disease in a subject, which is controlled by the CPU 101 executing the following computing means according to a program described later: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the renal disease based on the measurement value acquired by the acquisition means.
  • prediction of renal disease can be performed by the system 100 (FIGS. 1 and 2) provided with the arithmetic device 1 as the above-mentioned device.
  • FIG. 3 is a block diagram for explaining the function of the arithmetic device 1 according to the first embodiment.
  • the arithmetic device 1 includes a measurement value acquisition unit 11, a reference value acquisition unit 12, a measurement value comparison unit 13, and a prediction unit 14. These functional blocks are realized by, for example, installing the program according to the present invention in the recording unit 103 or the memory 102 of the arithmetic device 1 and executing the program by the CPU 101.
  • the acquisition unit and the prediction unit described in the claims correspond to the measurement value acquisition unit 11 and the prediction unit 14 shown in FIG. 3, respectively.
  • the reference value acquisition unit 12 and the measurement value comparison unit 13 may have any configuration.
  • the measured value M11 of the biomarker protein is taken into the arithmetic device 1 from the measuring device 5a
  • the measured value M21 of the biomarker mRNA is taken into the arithmetic device 1 from the measuring device 5b.
  • the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are recorded outside the computing device 1, and are taken into the computing device 1 via the Internet, for example.
  • the measured value M11 of the biomarker protein and the measured value M21 of the biomarker mRNA may be taken from a medical institution (not shown) via a network. Further, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA may be recorded in advance in the recording unit 103 or the memory 102 of the arithmetic device 1.
  • each function block of the measurement value acquisition unit 11, the reference value acquisition unit 12, the measurement value comparison unit 13, and the prediction unit 14 does not necessarily need to be executed by a single CPU, and a plurality of CPUs It may be distributed and processed.
  • the functions of the measurement value acquisition unit 11, the reference value acquisition unit 12, and the measurement value comparison unit 13 are performed by the CPU of the first computer, and the function of the prediction unit 14 is performed by the CPU of another second computer It may be configured as such.
  • the arithmetic device 1 executes the program according to the present invention in, for example, an executable format (for example, converted from a programming language by a compiler).
  • the arithmetic unit 1 performs processing using the program recorded in the recording unit 103.
  • the program may be installed in the arithmetic device 1 from a tangible computer readable non-temporary storage medium 109 such as a CD-ROM, or the arithmetic device 1 may be installed on the Internet (not shown). You may connect and download the program code of the program via the Internet.
  • kidney disease prediction method in the first embodiment is the same as the calculation device 1 according to the first embodiment in executing the following kidney disease prediction method of the present invention by the following program Good.
  • FIG. 4 is a flowchart showing an operation of the arithmetic device 1 according to the first aspect of the present invention. 4 by the measured value acquisition unit 11 shown in FIG. 3, step S12 by FIG. 4 by the reference value acquisition unit 12, step S13 by FIG. 4 by the measured value comparison unit 13, and FIG. The processes of steps S15 and S16 shown in FIG.
  • step S11 first, an input of start of acquisition of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started.
  • the measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S11 corresponds to the acquisition process described in the claims.
  • step S12 the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 or according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner.
  • the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
  • the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
  • step S14 when the comparison result in step S13 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is not a renal disease (step S15). Further, when the comparison result in step S13 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is a renal disease (step S16). Steps S14, S15, and S16 correspond to the prediction processing described in the claims.
  • the obtained prediction result is displayed on the display unit 9 of the arithmetic device 1 (step S17) or recorded in the recording unit 103 in the arithmetic device 1. Alternatively, it may be displayed on a display unit of a computer terminal outside the arithmetic device 1 connected via the Internet, for example, in a medical institution.
  • the computer program for predicting kidney disease includes a program that causes the CPU 101 of the arithmetic unit 1 to execute the steps S11 to S17.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
  • the method of predicting phosphorus metabolism abnormality in a subject in this embodiment comprises measuring at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from the subject Acquisition step of acquiring a measurement value of at least one mRNA selected from the group consisting of a value and / or a biomarker contained in a specimen collected from the subject, and the measurement value acquired in the acquisition step And a prediction step of predicting the phosphorus metabolism abnormality based on
  • the present embodiment compares the measured values obtained by the above-mentioned “2. Method for obtaining each measured value” with a predetermined reference value corresponding to each measured value, and the measured value is the reference. If out of the value range, the subject can be determined to be abnormal in phosphorus metabolism.
  • the measured value of one group of biomarkers when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is involved in the phosphorus metabolism abnormality. In addition, when the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the above-mentioned phosphorus metabolism disorder is related to FGF23. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, FGF23 is involved in the phosphorus metabolism abnormality. You may decide to
  • the process may be performed by the CPU 101 described later, or may be performed by an examiner.
  • a step of performing treatment eg, diet therapy such as low phosphorus diet
  • phosphorus metabolism abnormality is included.
  • the second embodiment includes an apparatus for predicting phosphorus metabolism abnormality of a subject, which is controlled by the CPU 101 by executing the following arithmetic function by a program described later: From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the phosphorus metabolism abnormality based on the measurement value acquired by the acquisition means.
  • FIG. 1 The block diagram for demonstrating the function of the calculating
  • the configuration of the arithmetic unit 2 is the same as the above 4-2. It is the same as the arithmetic unit 1 described above.
  • the method for predicting phosphorus metabolism abnormality in the second embodiment is such that the arithmetic device 2 according to the second embodiment executes the following method for predicting phosphorus metabolism abnormality of the present invention by the following program May be
  • FIG. 5 is a flowchart showing the operation of the arithmetic device 2 according to the second aspect of the present invention.
  • the step S21 in FIG. 5 is performed by the measured value acquiring unit 11 shown in FIG. 3
  • the step S22 in FIG. 5 is performed by the reference value acquiring unit 12, the step S23 in FIG.
  • step S21 first, an input of start of acquisition of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started.
  • the measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S21 corresponds to the acquisition process described in the claims.
  • the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner.
  • the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
  • the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
  • step S24 when the comparison result in step S23 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is not abnormal in phosphorus metabolism (step S25). Further, when the comparison result in step S23 is outside the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is abnormal in phosphorus metabolism (step S26). Steps S24, S25, and S26 correspond to the prediction process described in the claims.
  • the obtained prediction result is displayed on the display unit 9 of the computing device 2 (step S 27) or recorded in the recording unit 103 in the computing device 2. Alternatively, it may be displayed on a display unit of a computer terminal outside the computing device 2 connected via the Internet, for example, in a medical institution.
  • a computer program for predicting phosphorus metabolism abnormality includes a program that causes the CPU 101 of the arithmetic unit 2 to execute the steps S21 to S27.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
  • the method of predicting FGF23 activation in a subject is a method of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from a subject
  • the present embodiment compares the measured values obtained by the above-mentioned “2. Method for obtaining each measured value” with a predetermined reference value corresponding to each measured value, and the measured value is the reference. If out of the value range, the subject can be determined to have renal disease.
  • the measured value of one group of biomarkers when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is activated in the subject. In addition, when measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that FGF23 is activated in the subject. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, FGF23 is activated in the subject You may decide to
  • the process may be performed by the CPU 101 described later, or may be performed by an examiner.
  • a treatment for improving FGF23 activation eg, administration of anti-FGF antibody, high phosphorus diet, etc.
  • Dietary treatment may be included.
  • a third embodiment includes a device for predicting activation of FGF23 in a subject, which is controlled by the CPU 101 executing the following arithmetic function by a program described later: It consists of a measured value of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from subjects, and / or biomarkers contained in samples collected from said subjects Acquisition means for acquiring a measurement value of at least one type of mRNA selected from a group, and prediction means for predicting the activation of the FGF23 based on the measurement value acquired by the acquisition means.
  • FIG. 3 The block diagram for demonstrating the function of the arithmetic unit 3 which concerns on a 3rd embodiment is shown in FIG.
  • the configuration of the arithmetic unit 3 is the same as the above 4-2. It is the same as the arithmetic unit 1 described above.
  • the method of predicting FGF23 activation in the third embodiment is as follows:
  • the arithmetic device 3 according to the third embodiment uses the following program to predict the following FGF23 activation method of the present invention It may be executed.
  • FIG. 6 is a flowchart showing the operation of the arithmetic device 3 according to the third aspect of the present invention. 6 by the measured value acquisition unit 11 shown in FIG. 3, step S32 by FIG. 6 by the reference value acquisition unit 12, step S33 by FIG. 6 by the measured value comparison unit 13, and FIG. The processes of steps S35 and S36 shown in 6 are respectively executed.
  • step S31 first, an acceptance start input of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started.
  • the measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S31 corresponds to the acquisition process described in the claims.
  • step S ⁇ b> 32 the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 or according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner.
  • the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
  • step S33 the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or the bio The marker mRNA is compared with the reference value R22.
  • step S34 when the comparison result in step S33 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is not activated in the subject (step S35). . Further, when the comparison result in step S33 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is activated in the subject (step S36). Steps S34, S35, and S36 correspond to the prediction process described in the claims.
  • the obtained prediction result is displayed on the display unit 9 of the arithmetic device 3 (step S37) or recorded in the recording unit 103 in the arithmetic device 3. Alternatively, it may be displayed on a display unit of a computer terminal outside the arithmetic device 3 connected via the Internet, for example, in a medical institution.
  • the computer program for predicting the activation of FGF23 includes a program that causes the CPU 101 of the arithmetic unit 3 to execute the steps S31 to S37.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
  • a "subject” is an individual to whom a test substance is administered, preferably a human is excluded.
  • mammals other than human include cows, horses, sheep, goats, pigs, dogs, cats, rabbits, monkeys and the like. Preferred are humans, cats and dogs. Also, the age and gender of the individual do not matter.
  • the subject may or may not be an individual having a history of impaired renal function or other renal disease. Also, the subject may or may not be a subject suffering from a disease associated with FGF23.
  • the sample is at least one selected from the group consisting of a sample collected from the skin, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin included.
  • the sample collected from the skin is not limited as long as it originates from the skin of a living body.
  • the skin-derived sample also includes sweat, secretions from the skin, and the like.
  • the method of collecting the skin is also not limited, and it is possible to increase the biopsy material, the skin adhering to the piercing needle when the piercing hole is made, the abrasion of the skin surface, and the like.
  • the sample may be fresh or stored.
  • it can be stored in a room temperature environment, a refrigerated environment or a frozen environment, but is preferably frozen.
  • the “test tissue” is a tissue to which a test substance is to be administered, for example, it is collected from an individual who can be the subject, and survives outside the body by being cultured outside the body. It is an organization.
  • the tissue may be an entire organ or a part of an organ.
  • the “test cell” is a cell to which a test substance is to be administered, for example, it is collected from an individual who can be the subject, and survives outside the body by being cultured in vitro. It is a cell.
  • the cell may be a cell having limited passage such as primary culture, or may be a so-called cultured cell whose passage is maintained. Also, these may be cells produced by genetic engineering.
  • the “test substance” is a substance that can be a subject to be evaluated as to whether it is a candidate substance of the active ingredient and is not particularly limited.
  • compounds, proteins, peptides, nucleic acids, lipids, carbohydrates, glycolipids, glycoproteins, metals and the like can be mentioned.
  • the administration method of the test substance is also not particularly limited.
  • the test substance can be administered, for example, at 1 pg / ml to 1 mg / ml in the culture medium.
  • the test substance can be administered at 1 ng / kg to 1 g / kg per day.
  • the period from the administration of the test substance to the collection of the sample is not particularly limited as long as the effect of the test substance can be obtained.
  • test substance-treated sample a subject that has been treated with a test substance, a test tissue or a sample collected from test cells
  • test substance-treated sample a subject that has not been treated with a test substance, a subject collected from a subject tissue or a subject cell
  • nonaive subject a subject that has not been treated with a test substance, a subject collected from a subject tissue or a subject cell
  • kidney disease “fGF23 related disease”, “biomarker”, “measured value of at least one protein selected from the group consisting of biomarkers”, “selected from the group consisting of biomarkers” Value of at least one type of mRNA, “predetermined reference value”, “healthy individual”, “multiple samples”, “multiple values”, “anti-biomarker antibody”, “biomarker mRNA detection nucleic acid”
  • predetermined reference value “healthy individual”, “multiple samples”, “multiple values”, “anti-biomarker antibody”, “biomarker mRNA detection nucleic acid”
  • I.I. 1 A description of the terms of is incorporated herein.
  • methods for obtaining “a measured value of at least one type of protein selected from the group consisting of biomarkers” and “a measured value of at least one type of mRNA selected from the group consisting of biomarkers” are described above in I. 2. It conforms to.
  • FIG. 7 is a schematic view of a system 200 according to fourth and fifth embodiments of the present invention
  • FIG. 8 is a block diagram showing a hardware configuration of the system 200.
  • the system 200 refers to a screening apparatus (in the following, having the same hardware configuration and referring to screening apparatuses having different functional configurations as will be described later).
  • the "screening devices 4 and 5" are provided, the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
  • the screening devices 4 and 5 are, for example, general-purpose personal computers, and include a CPU 101 that performs data processing to be described later, a memory 102 used for a data processing work area, a recording unit 103 that records processing data, And an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device.
  • the input unit 8 and the display unit 9 are connected to the screening devices 4 and 5, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like.
  • the input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel.
  • the screening devices 4 and 5 do not have to be integrated devices, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate
  • the screening devices 4 and 5, the measuring device 5a, and the measuring device 5b do not necessarily have to be disposed at one place, and a system in which devices provided at different places are communicably connected via a network is configured. May be.
  • the processing performed by the screening devices 4 and 5 actually means the processing performed by the CPU 101 of the screening devices 4 and 5 based on the program stored in the recording unit 103 or the memory 102. Do.
  • the CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
  • the measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53.
  • the sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased.
  • the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done.
  • measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
  • measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
  • the measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54.
  • the sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
  • the measuring devices 5a and 5b are connected to the screening devices 4 and 5 by wire or wirelessly.
  • the measuring device 5a performs A / D conversion of the measured value of the protein, and transmits it to the screening device 4, 5 as digital data.
  • the measuring device 5b converts the measured value of mRNA into a digital signal, and transmits it to the screening devices 4 and 5 as digital data.
  • the screening devices 4 and 5 can acquire the measurement value of the protein and the measurement value of the mRNA as digital data that can be processed.
  • the measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the screening devices 4 and 5 can acquire the measurement value of the kidney disease marker as digital data.
  • the method of screening for a candidate substance of the active ingredient for suppressing the functional expression of FGF23 in the subject in the present embodiment is the method of collecting the substance from the skin of the subject (except human) treated with the test substance Of the biomarker protein of at least one test substance-treated sample selected from the group consisting of a tested sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin and And / or acquiring the measurement value of mRNA of the protein, and determining the test substance as a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquiring means. .
  • a test sample obtained from the skin of a subject (except for human beings) not treated with the test substance a test derived from the skin according to the above-mentioned "I. Measurement value of corresponding biomarker protein and / or mRNA of said protein in at least one unprocessed sample selected from the group consisting of a sample collected from a tissue and a sample collected from a test cell derived from skin Including the step of obtaining a measurement value.
  • the measured value of each biomarker obtained from the test substance-treated sample is compared with the corresponding measured value in the unsampled process, and compared with the measured value of the biomarker of the untreated sample to be treated with the test substance If the measured value of the biomarker of the sample has changed, it can be determined that the test substance is an active ingredient that suppresses the functional expression of FGF23.
  • the functional expression of FGF23 means that the original function of FGF23 is exhibited.
  • An example of the functional expression of FGF23 is, for example, lowering the concentration of inorganic phosphorus in blood.
  • functional expression of FGF23 includes an increase in expression of FGF23.
  • At least one selected from the group consisting of Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1 and Lrrc2 (referred to as the third group) Is up-regulated in FGF23 knockout mice as compared to mice receiving a high phosphorus diet.
  • At least one type (group 4) selected from the group consisting of Col15a1, Sparc, Col11a1, Clec11a, Serpinb6d, and Defb8 has a decreased expression in FGF23 knockout mice as compared to mice receiving a high phosphorus diet.
  • at least one type (the above-mentioned two groups) selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 does not change the expression in FGF23 knockout mice.
  • the measured value of the biomarker of the test substance-treated sample is increased compared to the measured value of the biomarker of the untreated sample. Then, the test substance can be determined to suppress the functional expression of FGF23. Also, for the 4 groups of biomarkers, if the mouse receiving a high phosphorus diet is the subject, the measured value of the biomarker of the test substance-treated sample is lower than the measured value of the biomarker of the untreated sample. The test substance can be determined to suppress the functional expression of FGF23.
  • test substance can be determined to act specifically on the function of FGF23.
  • the measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of the protein is higher than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of the protein
  • the measured value of the test substance-treated sample is, for example, 115% or more, preferably 130% or more, more preferably 150% or more of the measured value of the untreated sample
  • the test substance it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is rising.
  • the measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of said protein is lower than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of said protein
  • the measured value of the test substance-treated sample is, for example, 85% or less, preferably 70% or less, more preferably 50% or less of the measured value of the untreated sample, the test substance
  • the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is decreasing.
  • the test substance is administered in the administration step of (i) administering the test substance to the subject, the test tissue or the test cell, and (ii) the step (i)
  • the method may include the steps of collecting the sample from the subject, the test tissue or the test cells, and (iii) recovering the protein and / or the mRNA from the sample obtained in the step (ii).
  • the step (ii) and the step (iii) do not necessarily need to be performed continuously in the same organization, and for example, the sample collected in the step (ii) is sent to a third party organization iii) The following may be implemented.
  • step (iii) and step (I) do not necessarily have to be performed continuously in the same organization, for example, the step of sending the protein and / or mRNA recovered in step (iii) to a third party organization (Iii) The following may be implemented.
  • a fourth embodiment is a candidate substance of an active ingredient for suppressing the functional expression of FGF23 having the following arithmetic means, which is controlled by the CPU 101 executing the following arithmetic function according to a program described later
  • the screening device 4 includes: Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Means for acquiring the measurement value of the biomarker protein of at least one test substance-treated sample and / or the measurement value of mRNA of the protein, and the measurement acquired by the first measurement value acquisition means
  • screening can be performed by the system 200 (FIGS. 7 and 8) provided with the screening device 4 as the above-mentioned device.
  • FIG. 9 is a block diagram for explaining the function of the screening device 4 according to this embodiment.
  • the screening device 4 includes a first measurement value acquisition unit 31, a second measurement value acquisition unit 32, a measurement value comparison unit 33, and a candidate substance determination unit 34.
  • the second measurement value acquisition unit 32 is an arbitrary configuration. These functional blocks are realized by installing the screening program according to the present invention in the recording unit 103 or the memory 102 of the screening device 4 shown in FIG. 8 and the CPU 101 executing this screening program.
  • the first measured value acquiring unit, the second measured value acquiring unit, the measured value comparing unit, and the determining unit according to the claims are the first measured value acquiring unit 31 and the second measured value illustrated in FIG. 9.
  • the acquisition unit 32, the measurement value comparison unit 33, and the candidate substance determination unit 34 correspond to each other.
  • the measured value M31 of the biomarker protein of the test substance-treated sample is taken from the measuring device 5a to the screening device 4, and the measured value M41 of the biomarker mRNA of the test substance-treated sample is measured from the measuring device 5b to the screening device Captured in 4.
  • the measured value M32 of the biomarker protein of the untreated sample and the measured value M42 of the biomarker mRNA of the untreated sample are recorded outside the screening device 4, and are taken into the screening device 4 via the Internet, for example.
  • the measured value M31 of the biomarker protein of the test substance-treated sample and the measured value M41 of the biomarker mRNA of the test substance-treated sample may be taken from a medical institution (not shown) via a network.
  • the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample may be recorded in advance in the recording unit 103 or the memory 102 of the cleaning device 4.
  • each functional block of the first measurement value acquisition unit 31, the second measurement value acquisition unit 32, the measurement value comparison unit 33, and the candidate substance determination unit 34 may be executed by a single CPU. It is not necessarily necessary, and processing may be distributed and performed by a plurality of CPUs.
  • the functions of the first measurement value acquisition unit 31, the second measurement value acquisition unit 32, and the measurement value comparison unit 33 are executed by the CPU of the first computer, and the function of the candidate substance determination unit 34 is It may be configured to be executed by the CPU of another second computer.
  • the screening device 4 executes the program according to the present invention in, for example, an execution form (for example, converted from a programming language by a compiler and generated) in order to perform the processes of steps S41 to S47 described in FIG.
  • the screening device 4 performs the process using the program recorded in the recording unit 103.
  • the above program may be installed in the screening device 4 from a tangible computer readable non-temporary recording medium 109 such as a CD-ROM, or the screening device 4 may be connected to the Internet (not shown). You may connect and download the program code of the program via the Internet.
  • FIG. 10 is a flowchart showing the operation of the screening device 4 according to the fourth aspect of the present invention. 10 is performed by the first measurement value acquisition unit 11 shown in FIG. 9, step S42 of FIG. 10 is performed by the second measurement value acquisition unit 32, and step S43 of FIG. 10 is performed by the measurement value comparison unit 33.
  • the candidate substance determining unit 34 executes the processing of steps S45 and S46 shown in FIG.
  • step S41 first, an input for starting acquisition of the measured value M31 of the biomarker protein of the test substance-treated sample or the measured value M41 of the biomarker mRNA of the test substance-treated sample from the examiner is received from the input unit 8 Alternatively, upon receiving a measurement start instruction from the measurement device 5a or 5b, the first measurement value acquisition unit 31 starts acquisition of the measurement value. Step S41 corresponds to the acquisition process described in the claims.
  • step S42 the second measurement value acquisition unit 32 acquires the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample.
  • step S43 the measurement value comparison unit 33 compares the measurement value M31 of the biomarker protein of the test substance-treated sample with the measurement value M32 of the biomarker protein of the untreated sample respectively corresponding to the test substance treatment
  • the measured value M41 of the biomarker mRNA of the sample is compared with the measured value M42 of the biomarker mRNA of the untreated sample corresponding to each.
  • step S44 if the comparison result in step S43 indicates a change, the candidate substance determination unit 34 determines that the test substance is a candidate substance of the active ingredient (step S45). In addition, when the result of the comparison in step S43 shows no change, the candidate substance determination unit 34 determines that the test substance is not a candidate substance of the active ingredient (step S46). Steps S44, S45, and S46 correspond to the prediction process described in the claims.
  • the obtained determination result is displayed on the display unit 9 of the screening device 4 (step S47) or recorded in the recording unit 103 in the screening device 4. Alternatively, it may be displayed on a display unit of a computer terminal outside the screening device 4 connected via the Internet, for example, at a medical institution.
  • the computer program for screening a candidate substance of an active ingredient for suppressing the functional expression of FGF23 includes a program for causing the CPU 101 of the screening device 4 to execute the steps S41 to S47.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the screening device can read the program.
  • the method of screening for a candidate substance of an active ingredient for activating the function of FGF23 in a subject is a method for collecting a substance from the skin of a subject (excluding human) treated with a test substance Of the biomarker protein of at least one test substance-treated sample selected from the group consisting of a tested sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin and And / or acquiring the measurement value of mRNA of the protein, and determining the test substance as a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquiring means. .
  • a test sample obtained from the skin of a subject (except for human beings) not treated with the test substance a test derived from the skin according to the above-mentioned "I. Measurement value of corresponding biomarker protein and / or mRNA of said protein in at least one unprocessed sample selected from the group consisting of a sample collected from a tissue and a sample collected from a test cell derived from skin Including the step of obtaining a measurement value.
  • the measured value of each biomarker obtained from the test substance-treated sample is compared with the corresponding measured value in the unsampled process, and compared with the measured value of the biomarker of the untreated sample to be treated with the test substance If the measured value of the biomarker of the sample has changed, it can be determined that the test substance is an active ingredient that activates the function of FGF23.
  • the activation of the function of FGF23 means that the original function of FGF23 is activated.
  • An example of functional activation of FGF23 is, for example, lowering the concentration of inorganic phosphorus in blood.
  • activation of the function of FGF23 includes an increase in the expression of FGF23.
  • the measured value of the biomarker of the test substance-treated sample is reduced compared to the measured value of the biomarker of the untreated sample.
  • the test substance can be determined to activate the function of FGF23.
  • the test substance is The function of FGF23 can be determined as activation.
  • test substance can be determined to act specifically on the function of FGF23.
  • the measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of the protein is higher than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of the protein
  • the measured value of the test substance-treated sample is, for example, 115% or more, preferably 130% or more, more preferably 150% or more of the measured value of the untreated sample
  • the test substance it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is rising.
  • the measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of said protein is lower than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of said protein
  • the measured value of the test substance-treated sample is, for example, 85% or less, preferably 70% or less, more preferably 50% or less of the measured value of the untreated sample, the test substance
  • the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is decreasing.
  • the test substance is administered in the administration step of (i) administering the test substance to the subject, the test tissue or the test cell, and (ii) the step (i)
  • the method may include the steps of collecting the sample from the subject, the test tissue or the test cells, and (iii) recovering the protein and / or the mRNA from the sample obtained in the step (ii).
  • the step (ii) and the step (iii) do not necessarily need to be performed continuously in the same organization, and for example, the sample collected in the step (ii) is sent to a third party organization iii) The following may be implemented.
  • step (iii) and step (I) do not necessarily have to be performed continuously in the same organization, for example, the step of sending the protein and / or mRNA recovered in step (iii) to a third party organization (Iii) The following may be implemented.
  • a fifth embodiment is a candidate substance of an active ingredient for activating the function of FGF23, having the following calculation means, which is controlled by the CPU 101 executing the following calculation function by a program described later
  • the screening device 5 of: Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Means for acquiring the measurement value of the biomarker protein of at least one test substance-treated sample and / or the measurement value of mRNA of the protein, and the measurement acquired by the first measurement value acquisition means
  • screening can be performed by the system 200 (FIGS. 7 and 8) provided with the screening device 5 as the above-mentioned device.
  • FIG. 5 The block diagram for demonstrating the function of the screening apparatus 5 which concerns on this embodiment is shown in FIG.
  • the configuration of the screening device 5 is the same as that of the screening device 4 described in the above II.3-2.
  • the screening device 5 uses the following program to screen the candidate substances of the active ingredient for activating the function of FGF23. Screening of candidate active ingredients for activating the function of FGF23 may be performed.
  • FIG. 11 is a flowchart showing the operation of the screening device 5 according to the fifth embodiment of the present invention. Note that step S51 in FIG. 11 is performed by the first measurement value acquisition unit 31 shown in FIG. 9, step S52 in FIG. 11 is performed by the second measurement value acquisition unit 32, and step S53 in FIG.
  • the candidate substance determining unit 34 executes the processing of steps S55 and S56 shown in FIG.
  • step S51 first, an input for starting acquisition of the measured value M31 of the biomarker protein of the test substance-treated sample or the measured value M41 of the biomarker mRNA of the test substance-treated sample from the examiner is received from the input unit 8 Alternatively, upon receiving a measurement start instruction from the measurement device 5a or 5b, the first measurement value acquisition unit 31 starts acquisition of the measurement value.
  • Step S51 corresponds to the acquisition process described in the claims.
  • step S52 the second measurement value acquisition unit 32 acquires the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample.
  • step S53 the measurement value comparison unit 33 compares the measurement value M31 of the biomarker protein of the test substance-treated sample with the measurement value M32 of the biomarker protein of the untreated sample respectively corresponding to the test substance treatment
  • the measured value M41 of the biomarker mRNA of the sample is compared with the measured value M42 of the biomarker mRNA of the untreated sample corresponding to each.
  • step S54 when the result of comparison in step S53 is changing, the candidate substance determination unit 34 determines that the test substance is a candidate substance of the active ingredient (step S55). In addition, when the result of the comparison in step S53 is not changed, the candidate substance determination unit 34 determines that the test substance is not a candidate substance of the active ingredient (step S56). Steps S54, S55, and S56 correspond to the prediction process described in the claims.
  • the obtained determination result is displayed on the display unit 9 of the screening device 4 (step S57) or recorded in the recording unit 103 in the screening device 4. Alternatively, it may be displayed on a display unit of a computer terminal outside the screening device 4 connected via the Internet, for example, at a medical institution.
  • the computer program of the screening of the candidate substance of the active ingredient for suppressing the functional expression of FGF23 includes a program that causes the CPU 101 of the screening device 4 to execute the steps S51 to S57.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the screening device can read the program.
  • the "disease” is not limited.
  • the disease may include any disease or disorder that may develop in each organ of the individual.
  • it includes abnormalities specific to the specific disease (also referred to as “pre-lesion”) that occur before the disease is reached.
  • Preferred diseases include ischemic diseases such as thrombosis, embolism, and stenosis (in particular, heart, brain, lung, large intestine, etc.); circulatory disorders such as aneurysm, varicose, congestion, hemorrhage, etc.
  • aorta, vein, lung Liver, spleen, retina etc. allergic diseases such as allergic bronchitis, glomerulonephritis; degenerative diseases such as dementia, Parkinson's disease, amyotrophic lateral sclerosis, myasthenia gravis, etc. Muscles, etc .; Tumors (benign epithelial tumors, benign non-epithelial tumors, malignant epithelial tumors, malignant non-epithelial tumors); metabolic diseases (glycometabolic disorders, dyslipidemia, electrolytic abnormalities); infections (bacterial) , Viruses, rickettsial, chlamydia, fungi etc., protozoa, parasites etc.
  • the diseases also include diseases causing systemic symptoms.
  • diseases that cause systemic symptoms include autoimmune diseases such as systemic lupus erythematosus and multiple sclerosis; metabolic abnormalities such as hereditary mucopolysaccharidosis and the like.
  • FIG. 12 is a schematic view of a system 300 according to a sixth embodiment of the present invention.
  • the system 300 refers to arithmetic devices having the same hardware configuration and different functional configurations as described later, and therefore, the system 300 collectively refers to the arithmetic devices having these different functional configurations.
  • the arithmetic unit 6 the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
  • the arithmetic device 6 is constituted by, for example, a general-purpose personal computer, and is located between a CPU 101 for performing data processing described later, a memory 102 used for a data processing work area, a recording unit 103 for recording processing data, And an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device.
  • the input unit 8 and the display unit 9 are connected to the arithmetic device 6, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like.
  • the input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel.
  • the arithmetic device 6 does not have to be an integrated device, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate
  • the arithmetic device 6, the measuring device 5a, and the measuring device 5b are not necessarily arranged at one place, and a system in which devices provided at different places are communicably connected by a network may be configured. .
  • the processing performed by the arithmetic device 6 actually means the processing performed by the CPU 101 of the arithmetic device 6 based on the program stored in the recording unit 103 or the memory 102.
  • the CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
  • the measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53.
  • the sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased.
  • the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done.
  • measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
  • measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
  • the measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54.
  • the sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
  • the measuring devices 5a and 5b are connected to the computing device 6 by wire or wirelessly.
  • the measuring device 5a performs A / D conversion of the measured value of the protein, and transmits the converted value as digital data to the computing device 6.
  • the measuring device 5b A / D converts the measured value of mRNA, and transmits it to the computing device 6 as digital data.
  • the arithmetic unit 6 can acquire the measurement value of the protein and the measurement value of the mRNA as digital data that can be processed.
  • the measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the arithmetic unit 6 can acquire the measurement value of the kidney disease marker as digital data.
  • a process for obtaining a measured value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measured value of mRNA of the protein Determining based on the value that FGF23 is involved in the disease.
  • the present embodiment compares the measured values obtained by the above-mentioned “I.2. Acquisition method of each measured value” with a predetermined reference value corresponding to each measured value, and the measured value When it is outside the reference value range, it can be determined that the disease is related to FGF23.
  • the disease is related to FGF23.
  • the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the disease is related to FGF23. More preferably, when the measured value of one group of biomarkers is out of the range of the reference value and the measured value of the two groups of biomarkers is within the range of the reference value, May be determined to be involved.
  • the three groups of biomarkers mentioned are up-regulated in FGF23 knockout mice as compared to mice inoculated with a high phosphorus diet. Also, the four groups of biomarkers have reduced expression in FGF23 knockout mice as compared to mice inoculated with a high phosphorus diet. Furthermore, at least one type (the above-mentioned two groups) selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 does not change the expression in FGF23 knockout mice.
  • the functional expression of FGF23 is suppressed in the disease.
  • the functional expression of FGF23 is suppressed in the disease. It can be decided.
  • the two groups of biomarkers are examined in the same manner, if the measured value in the subject does not change compared to the reference value, the function of FGF23 in the disease is It can be determined that it is acting specifically.
  • the process may be performed by the CPU 101 described later, or may be performed by an examiner.
  • a treatment for improving the functional expression of FGF23 for example, a low phosphorus diet (e.g., a low phosphorus diet) Etc. may be included.
  • the sixth embodiment relates to the involvement of FGF23 in a disease in which the following means are controlled by the CPU 101 executing the following arithmetic function by a program described later Including a predictor to predict: First measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition means A determination means for determining that FGF23 is involved in the disease based on the measurement value obtained by
  • the involvement of FGF23 in a disease can be predicted by the system 300 (FIGS. 12 and 13) provided with the arithmetic device 6 as the above-mentioned device.
  • FIG. 14 is a block diagram for explaining the function of the arithmetic device 6 according to the sixth embodiment.
  • the configuration of the arithmetic unit 6 is the same as that of the above-described I. 4-2. It is the same as the arithmetic unit 1 described above.
  • the method for predicting the involvement of FGF23 in a disease according to the sixth embodiment is that the arithmetic device 6 according to the sixth embodiment relates FGF23 to the following disease of the present invention by the following program.
  • the prediction method may be performed.
  • FIG. 15 is a flowchart showing the operation of the arithmetic device 6 according to the sixth aspect of the present invention.
  • the step S61 in FIG. 15 is performed by the measured value acquiring unit 11 shown in FIG. 14, the step S62 in FIG. 15 is performed by the reference value acquiring unit 12, the step S63 in FIG.
  • the processes of steps S65 and S66 shown in 15 are respectively executed.
  • step S61 first, an acceptance start input of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started.
  • the measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S61 corresponds to the acquisition process described in the claims.
  • the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner.
  • the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
  • the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
  • step S64 when the comparison result in step S63 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is not involved in the disease (step S65). . Further, when the comparison result in step S63 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is involved in the disease (step S66). Steps S64, S65, and S66 correspond to the prediction process described in the claims.
  • the obtained prediction result is displayed on the display unit 9 of the arithmetic device 6 (step S67) or recorded in the recording unit 103 in the arithmetic device 6.
  • it may be displayed on a display unit of a computer terminal, for example, in a medical institution, which is connected via the Internet and which is external to the arithmetic device 6.
  • a computer program according to the sixth embodiment for predicting whether or not FGF23 is involved in a disease includes a program that causes the CPU 101 of the arithmetic unit 6 to execute the steps S61 to S67.
  • the computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
  • test Reagent and Test Kit An example of the seventh embodiment of the present invention relates to a test reagent comprising the anti-biomarker antibody used in the above-mentioned "I.2.
  • anti-biomarker antibody those described above in "I.1. Explanation of terms" can be used.
  • the test reagent of this embodiment may contain at least one or more anti-biomarker antibodies.
  • the anti-biomarker antibody is a polyclonal antibody, it may be a polyclonal antibody obtained by immunizing with one type of antigen, or may be obtained by immunizing the same individual with two or more types of antigens in parallel. It may be a polyclonal antibody. Furthermore, each polyclonal antibody obtained by inoculating different animals with two or more kinds of antigens may be mixed.
  • the anti-biomarker antibody when the anti-biomarker antibody is a monoclonal antibody, it may be a monoclonal antibody produced from one type of hybridoma, but is a monoclonal antibody produced from two or more types of hybridomas, and each monoclonal antibody is Two or more types of multiple monoclonal antibodies that recognize the same or different epitopes may be included. In addition, one or more kinds of polyclonal antibodies and monoclonal antibodies may be mixed and contained.
  • the form of the anti-biomarker antibody contained in the test reagent is not particularly limited, and may be in a dry state or liquid state such as an antiserum containing anti-biomarker antibody or ascites fluid.
  • the form of the anti-biomarker antibody may be a dried state or an aqueous solution of a purified anti-biomarker antibody, an immunoglobulin fraction containing the anti-biomarker antibody, or an IgG fraction containing the anti-biomarker antibody.
  • a stabilizer such as ⁇ -mercaptoethanol and DTT
  • a protective agent such as albumin
  • polyoxyethylene (20) sorbitan It may contain at least one of surfactants such as monolaurate, polyoxyethylene (10) octyl phenyl ether, and preservatives such as sodium azide.
  • the form of the anti-biomarker antibody is a purified anti-biomarker antibody, an immunoglobulin fraction containing the anti-biomarker antibody, or a dried state or an aqueous solution of an IgG fraction containing the anti-biomarker antibody, phosphate buffer Buffer component such as liquid; Stabilizing agent such as ⁇ -mercaptoethanol and DTT; Protective agent such as albumin; Salt such as sodium chloride; polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (10) octyl phenyl ether And the like, and may contain at least one preservative such as sodium azide.
  • the anti-biomarker antibody may be unlabeled or labeled with the above-mentioned labeling substance, but is preferably labeled with the above-mentioned labeling substance.
  • the labeling substance those exemplified in the above-mentioned section “I.2. Measurement value acquisition method” can be used.
  • the anti-biomarker antibody for antigen capture may be provided in a state of being immobilized on a solid phase surface or the like. The solid phase and the immobilization are as described above in the section “I.2. Method of obtaining measured value”.
  • the solid phase is a microplate.
  • test reagent may be provided as a kit.
  • the test kit of this embodiment comprises a solid phase on which the antigen-capture anti-biomarker antibody is immobilized, and a detection anti-bio And a marker antibody.
  • the labeling substance is an enzyme, it may contain the substrate solution.
  • the aforementioned test kit is, for example, a kit as shown in FIG.
  • the test kit 9 includes an external box 94, a microplate 92 on which an antibody for capturing an antigen is immobilized, a first container 91a containing an anti-biomarker antibody for detection labeled with a labeling substance, and a substrate solution that reacts with an enzyme. And the package insert 93 of the test kit.
  • the handling method of the test kit, storage conditions and the like can be described.
  • a container or the like containing an aqueous medium for washing may be packaged in the packaging box 94.
  • Another example of the seventh embodiment relates to a test reagent containing the biomarker mRNA detection nucleic acid used in the above-mentioned "I.2. Method for obtaining measurement value".
  • biomarker mRNA detection nucleic acid those described above in “I.1. Explanation of terms” can be used.
  • the test reagent containing the biomarker mRNA detection nucleic acid used for the microarray may be in a lyophilized state or in a solution containing a buffer such as Tris-HCl, EDTA, salt and the like. When there are a plurality of target biomarker mRNAs, it is preferable to place each detection nucleic acid in a separate container.
  • the biomarker mRNA detection nucleic acid may be immobilized on a substrate and provided as a microarray chip.
  • the substrate of the microarray is not particularly limited as long as it can immobilize the detection nucleic acid, and examples thereof include glass, polymers such as polypropylene, and nylon membranes.
  • the method for immobilizing the detection nucleic acid on the substrate can also be performed according to a known method, and for example, a spacer or crosslinker containing a reactive group for immobilizing the detection nucleic acid can be used.
  • test reagent containing a biomarker mRNA detection nucleic acid is provided as a kit together with the reagent, a medium such as paper, compact disc or the like on which information of the nucleic acid or information for accessing such information is recorded. It is also good.
  • the test reagent containing a biomarker mRNA detection nucleic acid used for RT-PCR may be in a lyophilised state or in a solution containing a buffer such as Tris-HCl, EDTA, salts and the like.
  • the primers may be provided in separate containers provided with the forward primer and the reverse primer, or may be provided in a mixed state.
  • the quantitative probe may be provided in a separate container with each primer, and each primer and the quantitative probe may be provided in a mixed state.
  • a test reagent containing a biomarker mRNA detection nucleic acid may be provided in the form of a kit, which comprises a forward primer and a reverse primer, or a forward primer, a reverse primer and a quantitative probe, and optionally a package insert.
  • the kit may include reagents for quantitative PCR.
  • test reagent containing the biomarker mRNA detection nucleic acid may be provided as a kit.
  • test reagent and the test kit according to the seventh embodiment can be used to obtain each measurement value in the first to sixth embodiments.
  • the eighth embodiment of the present invention is Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, Il22ra2, Rbfox1, Lrrc2, Col15a1, Aldh1l, Elc1
  • the present invention relates to a method for use as a biomarker of skin for predicting involvement of renal function, abnormal phosphorus metabolism, or FGF23, comprising at least one selected from the group consisting of Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8.
  • biomarkers are included in each sample.
  • the donor oligo DNA was designed to have a stop codon immediately below the Leucine coding sequence at the planned cleavage site for Fgf23-gRNA1 and Fgf23-gRNA2 (FIG. 17A).
  • ssODNs for Fgf23-gRNA1, 2 CGGATAGGCTCTAGCAGTGCCCAAGCTGCAGACAGTGCAAGAGCAGCGCCCAGGAGTCTcctcagaagaactcgtcaagaagCAGGTCCAGTCATGGCACAGCACTGAGTGGCTAATGCTGAGTTTGAATCTGACAA (SEQ ID NO: 3)
  • F2 heterozygous mice were produced by in vitro fertilization of the obtained F1 heterozygous male mice and wild type mouse females
  • F3 mice were produced by in vitro fertilization of the obtained F2 heterozygous female mice and F1 heterozygous male mice.
  • F4 mice were prepared by in vitro fertilization of F3 homozygous female mice and F3 heterozygous female mice and F1 heterozygous male mice. For the experiments, homozygotes from the F3 generation onward were used.
  • Genotype of each mouse was determined by direct sequence.
  • mice II-1 Preparation of UNx / HPi Model Mouse and Extraction of Tissue UNx / HPi mouse (one nephrectomy-high phosphorus diet mouse) was prepared by rearing on a high phosphorus diet after single nephrectomy. As a control, mice were raised on a low phosphorus diet after a sham operation.
  • mice that had one-sided nephrectomy were given a 2% inorganic phosphorus-containing high phosphorus diet (TD. 10662, Oriental Bioservice) (following, renal disease) Also called a group). Sham-operated mice were given a low phosphorus diet (TD. 10662 variant, Oriental Bioservice) containing 0.35% inorganic phosphorus (hereinafter also referred to as Sham group).
  • This chronic kidney disease model is a modification of the method described in Hu MC et al. (J Am Soc Nephrol 22, 124-136, 2011).
  • Hu MC et al. Added ischemia reperfusion injury to the remaining kidney (left kidney) at the time of half nephrectomy described in (1) above, but in this modification, ischemia reperfusion was not performed. Tissues were removed after 1 week (E), 4 weeks (M) and 8 weeks (L) after initiation of the high phosphorus diet (low phosphorus diet in the Sham group).
  • the animals from which the tissue is removed are bled from the orbital to the EDTA-added tube under anesthesia by intraperitoneal administration of Avertin (250 mg / kg), and cervical dislocation is euthanized, and organs and tissues (bone marrow, brain, The skin, heart, kidney, liver, lung, pancreas, skeletal muscle, spleen, testis, thymus, fat, large intestine, stomach, adrenal gland, aorta, eye, ileum, jejunum, pituitary, skull, salivary gland and thyroid) were removed.
  • the excised organs and tissues were measured for wet weight and immediately frozen with liquid nitrogen and stored at -80 ° C.
  • the collected blood was centrifuged at 1200 g for 10 minutes at room temperature.
  • the supernatant plasma was collected after centrifugation and stored at -80.degree.
  • Experiment III Measurement of inorganic phosphorus in plasma E- / M- / L-UNx / HPi, E- / M- / L-Sham, 4-week-old Fgf23 mutant mouse, WT3W / HP1W, WT3W / LP1W, WT3W In each mouse of / ND1W, 100 ⁇ l each of the cryopreserved plasma sample was used to measure the inorganic phosphorus level in plasma by the enzyme method.
  • Experimental Example IV Gene expression analysis in each tissue IV-1. Extraction of RNA from each tissue Cryopreserved tissue is homogenized in TRIzol Reagent (Thermo Fisher Scientific, MA, USA) for 10 seconds at 15,000 rpm with PT10-35 GT Polytron homogenizer (KINEMATICA, Luzern, Switzerland) After grinding and drying in liquid nitrogen using a mortar and pestle, homogenize for 10 seconds at 15,000 rpm with PT10-35 GT Polytron homogenizer in TRIzol Reagent, or use Cell Destroyer PS1000 or PS2000 (Bio Medical Science Inc., Homogenize at 4 ° C.
  • the recovered RNA was subjected to quality and concentration confirmation with Nanodrop (Thermo Fisher Scientific, MA, USA).
  • RNA expression analysis (1) Acquisition of RNAseq Data Using the above sample, data of RNAseq were acquired according to the following procedure. a. Quality inspection The quality inspection of the receiving sample was performed in the following items. -Measurement of concentration using Nanodrop (spectrophotometer)-Measurement of concentration using Agilent 2100 Bioanalyzer-Confirmation of quality b. Sample Preparation A library for the next generation sequencer HiSeq was prepared according to the following steps using SureSelect Strand-Specific RNA Library Preparation Kit. i. Recover poly (A) + RNA (mRNA) from Total RNA using oligo (dT) magnetic beads ii.
  • RNA iii cDNA synthesis iv. Double-stranded cDNA synthesis v. End repair, phosphorylation, A tail addition vi. Indexed Adapter Ligation vii. 13 cycle PCR viii. Purification by magnetic beads c. Data acquisition by next-generation sequencer Using the next-generation sequencer HiSeq 2500 or 4000 (illumina), sequencing was performed according to the following steps. i. Addition of sequencing reagent Reagent: TruSeq PE Cluster Kit v3-cBot-HS (1 flowcell) ⁇ PE-401-3001> (illumina) Reagents: TruSeq SBS Kit v3-HS (200 cycle) ⁇ FC-401-3001> (illumina) ii.
  • RNAseq data (2) Analysis of RNAseq data (2) -1. Analysis of Next-Generation Sequencer Output Data The following information processing was performed on the output data.
  • Base call Text data of the base sequence was acquired from the output analysis raw data (image data).
  • Filtering Screening of lead data by predetermined filtering was performed.
  • Sorting by Index arrangement Each sample data was sorted by Index information.
  • qRT-PCR CDNA was synthesized according to the standard protocol of Superscrtipt III First-Strand Synthesis Supermix (Thermo Fisher Scientific, MA, USA) using Oligo dT20 primers using 0.5-1 ⁇ g of total RNA obtained from skull and skin as a template for cDNA synthesis.
  • the synthesized cDNA is diluted 20-fold with TE buffer (10 mM Tris-HCl pH 8.0, 0.1 mM EDTA), and then LightCycler 480 II (Roche) according to the standard protocol of LightCycler 480 SYBR Green I Master (Roche, Basel, Switzerland) Real-time PCR was performed to measure Cp value.
  • the Cp value obtained for each gene was compared with the Cp value of Maea as a reference gene to quantify the relative expression level of each gene relative to the reference gene.
  • the primer pairs used in real time PCR are as shown in Table 5. All primers were designed by Primer-BLAST (NCBI).
  • Genes whose expression is controlled by Model I mechanism Expression is elevated in the skin due to renal disease and / or hyperphosphatemia. There are two pathways in which the expression of these genes is suppressed by renal disease and / or hyperphosphorus condition due to the elevated expression of FGF23 in bone and the pathway not affected by the elevated expression of FGF23.
  • Renal disease and / or high phosphorus status suppresses expression in the skin.
  • Renal disease and / or hyperphosphatemia suppresses expression in the skin.
  • the expression of these genes is suppressed by the upregulation of FGF23 in bone due to renal disease and / or hyperphosphatemia.
  • Renal disease and / or hyperphosphatemia suppresses expression in the skin.
  • the expression of these genes is not affected by the elevated expression of FGF23 in bone due to renal disease and / or hyperphosphate status.
  • Arithmetic unit 1 Arithmetic unit 1 2 Arithmetic unit 2 3 Arithmetic unit 3 4 Screening device 4 5 Screening device 5 6 Arithmetic device 6 11 measurement value acquisition unit 12 reference value acquisition unit 13 measurement value comparison unit 14 prediction unit 31 first measurement value acquisition unit 32 second measurement value acquisition unit 33 measurement value comparison unit 34 candidate substance determination unit

Abstract

One problem addressed by the present invention is to predict a kidney disease using a skin-derived sample. A device 1 that predicts a kidney disease in a subject and has an acquisition means and a prediction means. The acquisition means acquires a measured value for at least one type of protein selected from among biomarkers included in a skin-derived sample taken from the subject and/or a measured value for at least one type of mRNA selected from among the biomarkers included in the sample taken from the subject. The prediction means predicts the kidney disease on the basis of the measured value(s) acquired by the acquisition means.

Description

皮膚のバイオマーカーSkin biomarkers
 本発明は、皮膚由来検体を用いて腎疾患を予測すること、リンの代謝異常を予測すること、被験体におけるFGF23の活性化を予測すること、FGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングすること、FGF23の機能を活性化するための有効成分の候補物質をスクリーニングすること、疾患におけるFGF23の関与を予測することに関する。 The present invention relates to predicting renal disease using a skin-derived sample, predicting metabolic abnormality of phosphorus, predicting activation of FGF23 in a subject, and active ingredients for suppressing functional expression of FGF23. The present invention relates to screening of candidate substances, screening of candidate active ingredients for activating the function of FGF23, and predicting the involvement of FGF23 in diseases.
 疾患には、可逆的に治療できる状態とそうでない状態(つまり不可逆的な状態)がある。可逆的な状態中に、異常をいち早く検出し治療する、あるいはそのような状態にすらならないように予防することが健康を維持することに不可欠である。また、可逆的な状態であっても、疾患の早期発見は、より軽度な治療方法、より短期の治療期間、またより良い予後の健康状態へ直結する。また、心臓疾患、脳疾患、がん、糖尿病に代表されるように、一つの器官や組織の異常が他の器官の疾患を招く(一般に合併症と呼ばれている)ことはよく知られており、そのような疾患においては、ひとつの器官・組織の異常から他の器官・組織の疾患が引き起こされるのを出来るだけ早い段階で防ぐことが必須となる。 Diseases may or may not be reversibly treatable (i.e., irreversible). It is essential to maintain good health that during the reversible condition, the abnormality is detected and treated promptly or prevented from becoming even such condition. Also, even in reversible conditions, early detection of the disease leads directly to milder treatment regimens, shorter treatment periods, and better prognosis health. In addition, as represented by heart disease, brain disease, cancer and diabetes, it is well known that abnormalities in one organ or tissue lead to diseases in other organs (generally called complications). In such diseases, it is essential to prevent the disease of one organ or tissue from being caused by another organ or tissue as early as possible.
 人をふくめた全ての動物において、個々の器官や組織は個別の部品ではなく、それぞれが機能的なネットワークを形成することにより、個体レベルでの品質管理がなされている。全身に張り巡らされている血管ネットワークによるホルモンなどの内分泌因子の運搬、神経ネットワークによる各器官機能の協調的な調整は「多器官連関システム」の代表的な例であり、生理学、内分泌学として体系づけられている。 In all animals including humans, individual organs and tissues are not individual parts, and each form a functional network to achieve quality control at the individual level. The transport of endocrine factors such as hormones by vascular networks distributed throughout the body, and coordinated coordination of each organ function by neural networks are representative examples of "multi-organ linkage system", and systems as physiology and endocrinology It is attached.
 一方、透析や腎移植を必要とする末期腎不全患者(ESKD)は、世界的にも増加傾向にあり、1990~2000年の10年間で、ESKD患者数は、43万人から106.5万人に増加し、さらに2008年には、少なくとも165万人程度に増加している(非特許文献1)。慢性腎疾患(Chronic Kidney disease:CKD)は、ESKDの予備軍であるが、腎臓は、「沈黙の臓器」と呼ばれ、腎障害が起こってもその状態が臨床データ等に現れにくく、慢性腎疾患を来す前の腎機能低下は、早期発見が困難であるのが現状である。 On the other hand, the number of end-stage renal failure patients (ESKD) requiring dialysis and kidney transplantation is also on the rise worldwide, and the number of ESKD patients is 430,000 to 1,065,000 in 10 years from 1990 to 2000. The number has increased to at least 1.65 million in 2008 (non-patent document 1). Chronic kidney disease (Chronic Kidney disease: CKD) is a reserve arm of ESKD, but the kidneys are called "silent organs", and even if kidney damage occurs, the condition is less likely to appear in clinical data etc. Renal dysfunction prior to the onset of the disease is currently difficult to detect early.
 FGF23は、血中リン濃度を低下させるホルモンであり、腎近位尿細管におけるリンの再吸収、及び腸管からのリンの吸収を抑制することにより血中のリン濃度を低下させることが知られている。また、FGF23の濃度上昇は、慢性腎疾患に伴う骨-ミネラル代謝異常(CKD-MBD)を惹起することが知られている。 FGF23 is a hormone that lowers the level of phosphorus in blood, and is known to lower the level of phosphorus in blood by suppressing the reabsorption of phosphorus in the renal proximal tubule and the absorption of phosphorus from the intestinal tract. There is. It is also known that elevated levels of FGF23 cause bone-mineral metabolism disorder (CKD-MBD) associated with chronic kidney disease.
 本発明は、皮膚由来検体を用いて腎疾患を予測することを一つの課題とする。本発明は、皮膚由来検体を用いてリンの代謝異常を予測することを一つの課題とする。本発明は、被験体におけるFGF23の活性化を予測することを一つの課題とする。本発明は、FGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングすることを一つの課題とする。本発明は、FGF23の機能を活性化するための有効成分の候補物質をスクリーニングすることを一つの課題とする。本発明は、疾患におけるFGF23の関与を予測することを一つの課題とする。 An object of the present invention is to predict kidney disease using a skin-derived sample. An object of the present invention is to predict metabolic disorders of phosphorus using a skin-derived sample. The present invention makes it one task to predict the activation of FGF23 in a subject. One object of the present invention is to screen candidate substances of the active ingredient for suppressing the functional expression of FGF23. One object of the present invention is to screen candidate substances for active ingredients for activating the function of FGF23. The present invention makes it one task to predict the involvement of FGF23 in diseases.
 本発明者は、鋭意研究を重ねたところ、腎疾患動物モデルにおいて、ある種のバイオマーカーの発現が皮膚で病態に応じて変化することを見出した。また、これらのバイオマーカーでは、FGF23に関連するものと、関連しないものが存在することを見出した。 As a result of intensive studies, the inventor found that expression of certain biomarkers changes in the skin depending on the pathological condition in a kidney disease animal model. In addition, it was found that among these biomarkers, those related to FGF23 and those not related were present.
 本発明は、当該知見に基づいて完成されたものであり、以下の態様を含む。
項1.
 下記の手段を有する、被験体の腎疾患を予測する装置:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記腎疾患を予測する予測手段。
項2.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項1に記載の装置。
項3.
 前記予測手段は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、項1又は2に記載の装置。
項4.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記腎疾患に、FGF23が関与していると決定する、項3に記載の装置。
項5.
 コンピュータに実行させたときに、被験体の腎疾患を予測するための下記の処理を当該コンピュータに実施させるプログラム:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
 前記取得処理で取得された測定値に基づいて、前記腎疾患を予測する予測処理。
項6.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項5に記載のプログラム。
項7.
 前記予測処理では、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、項5又は6に記載のプログラム。
項8.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記腎疾患に、FGF23が関与していると決定する、項7に記載のプログラム。
項9.
 下記の工程を有する、被験体の腎疾患を予測する方法:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
 前記取得工程で取得された測定値に基づいて、前記腎疾患を予測する予測工程。
項10.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項9に記載の方法。
項11.
 前記予測工程は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、項9又は10に記載の方法。
項12.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記腎疾患に、FGF23が関与していると決定する、項11に記載の方法。
項13.
 下記の手段を有する、被験体におけるリン代謝異常を予測する装置:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記リン代謝異常を予測する予測手段。
項14.
前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項13に記載の装置。
項15.
 前記予測手段は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、項13又は14に記載の装置。
項16.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記リン代謝異常に、FGF23が関与していると決定する、項15に記載の装置。
項17.
 コンピュータに実行させたときに、被験体におけるリン代謝異常を予測するための下記の処理を当該コンピュータに実施させるプログラム:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
 前記取得処理で取得された測定値に基づいて、前記リン代謝異常を予測する予測処理。
項18.
前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項17に記載のプログラム。
項19.
 前記予測処理では、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、項17又は18に記載のプログラム。
項20.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記リン代謝異常に、FGF23が関与していると決定する、項19に記載のプログラム。
項21.
 下記の工程を有する、被験体におけるリン代謝異常を予測する方法:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
 前記取得工程で取得された測定値に基づいて、前記リン代謝異常を予測する予測工程。
項22.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項21に記載の方法。
項23.
 前記予測工程は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、項21又は22に記載の方法。
項24.
 さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
には、前記リン代謝異常に、FGF23が関与していると決定する、項23に記載の方法。
項25.
 下記の手段を有する、被験体におけるFGF23の活性化を予測する装置:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記FGF23の活性化を予測する予測手段。
項26.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項25に記載の装置。
項27.
 前記予測手段は、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、項25又は26に記載の装置。
項28.
 コンピュータに実行させたときに、被験体におけるFGF23の活性化を予測するための下記の処理を当該コンピュータに実施させるプログラム:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
 前記取得処理で取得された測定値に基づいて、前記FGF23の活性化を予測する予測処理。
項29.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項28に記載のプログラム。
項30.
 前記予測処理では、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、項28又は29に記載のプログラム。
項31.
 下記の工程を有する、被験体におけるFGF23の活性化を予測する方法:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
 前記取得工程で取得された測定値に基づいて、前記FGF23の活性化を予測する予測工程。
項32.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項31に記載の方法。
項33.
 前記予測工程は、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
 Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、項31又は32に記載の方法。
項34.
 下記の手段を有する、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング装置:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び
 前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
項35.
 被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
 被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較手段、
をさらに有し、
 前記決定手段は、前記測定値比較手段の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、項34に記載の装置。
項36.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項35に記載の装置。
項37.
 コンピュータに実行させたときに、FGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングするための下記の処理を当該コンピュータに実施させるスクリーニングプログラム:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び
 前記第1の測定値取得処理で取得された測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定処理。
項38.
 さらに、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
 被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較処理、
を前記コンピュータに実施させ、
 前記決定処理では、前記測定値比較処理の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、項37に記載のプログラム。
項39.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項37又は38に記載のプログラム。
項40.
 以下の工程を含む、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング方法:
 (I)被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
 (II)前記工程(I)で得られた測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程。
項41.
 前記工程(I)と(II)の間に、前記工程(I)で取得された被験物質処理検体の測定値と、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程をさらに含み、
 工程(II)が、前記比較結果に基づいて、前記被験物質が有効成分の候補物質であると決定する工程である、
をさらに含む、項40に記載の方法。
項42.
 前記工程(I)の前に、
(i)被験体(ヒトを除く)、皮膚に由来する被験組織又は被験細胞を、被験物質で処理する工程、
(ii)前記工程(i)において被験物質で処理された前記被験体、被験組織又は被験細胞から検体を採取する工程、及び
(iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程
を含む、項40又は41に記載の方法。
項43.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項40~42のいずれか一項に記載の方法。
項44.
 下記の手段を有する、FGF23の機能を活性化するための有効成分の候補物質のスクリーニング装置:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(被験物質処理検体)中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び
 前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
項45.
 被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(未処理検体)のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
 被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較手段、
をさらに有し、
 前記決定手段は、前記測定値比較手段の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、項44に記載のスクリーニング装置。
項46.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項45に記載の装置。
項47.
 コンピュータに実行させたときに、FGF23の機能を活性化するための有効成分の候補物質をスクリーニングするための下記の処理を当該コンピュータに実施させるスクリーニングプログラム:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(被験物質処理検体)中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び
 前記第1の測定値取得処理で取得された測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定処理。
項48.
 さらに、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(未処理検体)のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
 被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較処理、
を前記コンピュータに実施させ、
 前記決定処理では、前記測定値比較処理の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、項47に記載のプログラム。
項49.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項47又は48に記載のプログラム。
項50.
 以下の工程を含む、FGF23の機能を活性化するための有効成分の候補物質のスクリーニング方法:
 (I)被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
 (II)前記工程(I)で得られた測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程。
項51.
 前記工程(I)と(II)の間に、前記工程(I)で取得された被験物質処理検体の測定値と、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程をさらに含み、
 工程(II)が、前記比較結果に基づいて、前記被験物質が有効成分の候補物質であると決定する工程である、
項50に記載の方法。
項52.
 前記工程(I)の前に、
(i)被験体(ヒトを除く)、皮膚に由来する被験組織又は被験細胞を、被験物質で処理する工程、
(ii)前記工程(i)において被験物質で処理された前記被験体、被験組織又は被験細胞から検体を採取する工程、及び
(iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程
を含む、項50又は51に記載の方法。
項53.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項40~42のいずれか一項に記載の方法。
項54.
 下記の手段を有する、疾患におけるFGF23の関与を予測する予測装置:
 疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び 前記第1の測定値取得手段が取得した測定値に基づいて、前記疾患にFGF23が関与していると決定する決定手段。
項55.
 前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
 前記第1の測定値取得手段が取得した測定値及び前記第2の測定値取得手段が取得した測定値を比較する測定値比較手段、
をさらに有し、
 前記決定手段は、前記比較の結果に基づいて前記疾患にFGF23が関与していると決定する、項54に記載の装置。
項56.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項55に記載の装置。
項57.
 コンピュータに実行させたときに、疾患におけるFGF23の関与を予測するための下記の処理を当該コンピュータに実施させる予測プログラム:
 疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び 前記第1の測定値取得処理で取得された測定値に基づいて、前記疾患にFGF23が関与していると決定する決定処理。
項58.
 さらに、前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
 第1の測定値取得処理で取得された測定値及び第2の測定値取得処理で取得された測定値を比較する測定値比較処理、
を前記コンピュータに実施させ、
 前記決定処理では、前記比較の結果に基づいて前記疾患にFGF23が関与していると決定する、項57に記載のプログラム。
項59.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項57又は58に記載のプログラム。
項60.
 以下の工程を含む、疾患におけるFGF23の関与を予測する予測方法:
 (I)疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
 (II)前記工程(I)で得られた測定値に基づいて、前記疾患にFGF23が関与していると決定する工程。
項61.
 前記工程(I)と(II)の間に、前記工程(I)で取得された測定値と、前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程
をさらに含み、
 工程(II)が、前記比較の結果に基づいて、前記疾患にFGF23が関与していると決定する工程である、
項60に記載の方法。
項62.
 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、項60~61のいずれか一項に記載の方法。
項63.
 Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種を含む、腎機能、リン代謝異常、又はFGF23の関与を予測するための皮膚のバイオマーカー。
The present invention has been completed based on the above findings, and includes the following aspects.
Item 1.
Apparatus for predicting renal disease in a subject having the following means:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of
Prediction means for predicting the renal disease based on the measurement values acquired by the acquisition means.
Item 2.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to item 1, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 3.
The prediction means compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is outside the range of the reference value. The device described in.
Item 4.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
The device according to Item 3, wherein it is determined that FGF23 is involved in the renal disease.
Item 5.
A program that, when executed on a computer, causes the computer to perform the following processing to predict renal disease in a subject:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of
The prediction process which predicts the said renal disease based on the measured value acquired by the said acquisition process.
Item 6.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 6. The program according to item 5, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 7.
In the prediction process, the measured value is compared with a predetermined reference value, and if the measured value is out of the range of the reference value, the subject is determined to have a renal disease. The program described in.
Item 8.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
8. The program according to item 7, wherein it is determined that FGF23 is involved in the renal disease.
Item 9.
A method of predicting renal disease in a subject comprising the following steps:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of
A prediction step of predicting the renal disease based on the measurement value acquired in the acquisition step.
Item 10.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 10. The method according to item 9, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 11.
The prediction step compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is out of the range of the reference value. The method described in.
Item 12.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
Item 12. The method according to Item 11, wherein it is determined that FGF23 is involved in the renal disease.
Item 13.
Apparatus for predicting abnormal phosphorus metabolism in a subject, having the following means:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of
Prediction means for predicting the phosphorus metabolism abnormality based on the measurement value acquired by the acquisition means.
Item 14.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 14. The device according to item 13, wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 15.
The prediction means compares the measured value with a predetermined reference value, and when the measured value is out of the range of the reference value, predicts that the subject has a phosphorus metabolism abnormality, or The device according to 14.
Item 16.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
The device according to Item 15, wherein it is determined that FGF23 is involved in the phosphorus metabolism disorder.
Item 17.
A program that, when run on a computer, causes the computer to perform the following processing for predicting phosphorus metabolism abnormality in a subject:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of
The prediction process which predicts the said phosphorus metabolism abnormality based on the measured value acquired by the said acquisition process.
Item 18.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 Item 18. The program according to Item 17, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 19.
In the prediction processing, the measured value is compared with a predetermined reference value, and when the measured value is out of the range of the reference value, it is predicted that the subject has a phosphorus metabolism abnormality, or The program described in 18.
Item 20.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
20. The program according to Item 19, wherein it is determined that FGF23 is involved in the phosphorus metabolism disorder.
Item 21.
A method of predicting abnormal phosphorus metabolism in a subject comprising the following steps:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of
A prediction step of predicting the phosphorus metabolism abnormality based on the measurement value acquired in the acquisition step.
Item 22.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 22. The method according to item 21, which is at least one selected from the group consisting of, Col1a1 and Defb8.
Item 23.
The prediction step compares the measured value with a predetermined reference value, and predicts that the subject has a phosphorus metabolism abnormality if the measured value is out of the range of the reference value. The method described in 22.
Item 24.
Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, And / or if the measured value of one of the biomarkers is outside the range of the reference value, and / or
When the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value
24. The method according to item 23, wherein it is determined that FGF23 is involved in the phosphorus metabolism disorder.
Item 25.
Device for predicting FGF23 activation in a subject, having the following means:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring measurement values of at least one mRNA selected from the group consisting of
A prediction unit that predicts the activation of the FGF23 based on the measurement values acquired by the acquisition unit.
Item 26.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The device according to Item 25, which is at least one selected from the group consisting of, Col1a1 and Defb8.
Item 27.
The prediction means compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or
Item 25. Predicting that FGF23 is activated in the subject, provided that at least one biomarker selected from the group consisting of Aldh112, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the reference value range, Or the apparatus described in 26.
Item 28.
A program that, when run on a computer, causes the computer to perform the following processing to predict FGF23 activation in a subject:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring measurement values of at least one mRNA selected from the group consisting of
The prediction process which predicts activation of said FGF23 based on the measured value acquired by the said acquisition process.
Item 29.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 29. The program according to Item 28, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 30.
In the prediction processing, the measured value is compared with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or
Item 28. Predicting that FGF23 is activated in the subject, provided that at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the reference value range, Or the program described in 29.
Item 31.
A method of predicting FGF23 activation in a subject comprising the steps of:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one mRNA selected from the group consisting of
A prediction step of predicting activation of the FGF23 based on the measurement value acquired in the acquisition step.
Item 32.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 32. The method according to Item 31, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 33.
The prediction step compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, Il22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, And / or when the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or
It is predicted that FGF23 is activated in the subject, provided that at least one biomarker selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 is within the range of the reference value, Or the method according to 32.
Item 34.
Screening apparatus for candidate active ingredients for suppressing the functional expression of FGF23, having the following means:
Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A first measurement value acquiring unit for acquiring a measurement value of a biomarker protein of at least one test substance-treated sample and / or a measurement value of mRNA of the protein;
A determination means for determining that the test substance is a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquisition means.
Item 35.
From a group consisting of a sample collected from the skin of a subject (except for human beings) not treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A second measurement value acquiring means for acquiring a measurement value of a corresponding biomarker protein in at least one unprocessed sample to be selected and / or a measurement value of mRNA of said protein,
Measured value comparison means for comparing the measured value of the test substance-treated sample with the measured value of the untreated sample,
And have
35. The apparatus according to Item 34, wherein the determination means determines that the test substance is a candidate substance of the active ingredient based on the comparison result of the measurement value comparison means.
Item 36.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to Item 35, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 37.
A screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for suppressing the functional expression of FGF23:
Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A first measurement value acquisition process for acquiring a measurement value of a biomarker protein in at least one test substance-treated sample and / or a measurement value of mRNA of said protein, and
The determination process which determines that the said test substance is a candidate substance of an active ingredient based on the measured value acquired by said 1st measured value acquisition process.
Item 38.
Furthermore, it comprises a sample collected from the skin of a subject not treated with the test substance (except for human beings), a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A second measurement value acquisition process for acquiring a measurement value of a corresponding biomarker protein of at least one untreated sample selected from a group and / or a measurement value of mRNA of said protein,
A measured value comparison process comparing the measured value of the test substance-treated sample with the measured value of the untreated sample;
On the computer,
The program according to Item 37, wherein in the determination process, the test substance is determined to be a candidate substance of the active ingredient based on the comparison result of the measurement value comparison process.
Item 39.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to Item 37 or 38, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 40.
The screening method of the candidate substance of the active ingredient for suppressing the functional expression of FGF23 including the following processes:
(I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein,
(II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I).
Item 41.
Between the steps (I) and (II), the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with
Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
The method according to Item 40, further comprising
Item 42.
Before the step (I),
(I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance,
(Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in the step (i);
(Iii) a step of recovering protein and / or mRNA from the sample obtained in the step (ii)
The method according to Item 40 or 41, comprising
Item 43.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The method according to any one of Items 40 to 42, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 44.
Screening apparatus for candidate active ingredients for activating the function of FGF23, having the following means:
Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample) A first measurement value acquiring unit for acquiring a measurement value of a protein and / or a measurement value of mRNA of the protein;
A determination means for determining that the test substance is a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquisition means.
Item 45.
Biomarker protein of a sample collected from the skin of a subject (except for human beings) not treated with the test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (untreated sample) A second measurement value acquisition unit for acquiring the measurement value of the protein and / or the mRNA of the protein, and
Measured value comparison means for comparing the measured value of the test substance-treated sample with the measured value of the untreated sample,
And have
The screening device according to Item 44, wherein the determination means determines that the test substance is a candidate substance of the active ingredient based on the comparison result of the measurement value comparison means.
Item 46.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to Item 45, which is at least one selected from the group consisting of, Col1a1 and Defb8.
Item 47.
A screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for activating the function of FGF23:
Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample) A first measurement value acquisition process for acquiring a measurement value of a protein and / or a measurement value of mRNA of the protein, and
The determination process which determines that the said test substance is a candidate substance of an active ingredient based on the measured value acquired by said 1st measured value acquisition process.
Item 48.
Furthermore, the bio of a sample collected from the skin of a subject (except for human beings) not treated with the test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (untreated sample) A second measurement value acquisition process for acquiring a measurement value of a marker protein and / or a measurement value of mRNA of said protein, and
A measured value comparison process comparing the measured value of the test substance-treated sample with the measured value of the untreated sample;
On the computer,
The program according to Item 47, wherein in the determination process, the test substance is determined to be a candidate substance of the active ingredient based on a comparison result of the measurement value comparison process.
Item 49.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to Item 47 or 48, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 50.
The screening method of the active substance candidate substance for activating the function of FGF23 including the following processes:
(I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein,
(II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I).
Item 51.
Between the steps (I) and (II), the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with
Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
The method according to Item 50.
Item 52.
Before the step (I),
(I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance,
(Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in the step (i);
(Iii) a step of recovering protein and / or mRNA from the sample obtained in the step (ii)
52. A method according to item 50 or 51, comprising
Item 53.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The method according to any one of Items 40 to 42, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 54.
Predictor for predicting the involvement of FGF23 in disease comprising the following means:
First measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition means A determination means for determining that FGF23 is involved in the disease based on the measurement value obtained by
Item 55.
A second measurement value acquisition unit for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein;
Measurement value comparison means for comparing the measurement value acquired by the first measurement value acquisition means with the measurement value acquired by the second measurement value acquisition means;
And have
56. A device according to item 54, wherein the determination means determines that FGF23 is involved in the disease based on the result of the comparison.
Item 56.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 56. The device according to item 55, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 57.
A prediction program which, when executed on a computer, causes the computer to carry out the following processing for predicting the involvement of FGF23 in a disease:
A first measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition process The decision processing which determines that FGF23 is concerned in the said disease based on the measured value acquired by b.
Item 58.
Furthermore, a second measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein,
A measurement value comparison process comparing the measurement value acquired in the first measurement value acquisition process and the measurement value acquired in the second measurement value acquisition process;
On the computer,
60. The program according to Item 57, wherein in the determination processing, it is determined that FGF23 is involved in the disease based on the result of the comparison.
Item 59.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 60. The program according to Item 57 or 58, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 60.
Methods of predicting FGF23's involvement in disease comprising the following steps:
(I) obtaining a measured value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measured value of mRNA of said protein,
(II) A step of determining that FGF23 is involved in the disease based on the measurement value obtained in the step (I).
Item 61.
Between the steps (I) and (II), the measured value obtained in the step (I) and the measured value of the biomarker protein in a sample collected from the skin of a subject not having the disease And / or comparing the measured value of mRNA of said protein
Further include
Step (II) is a step of determining that FGF23 is involved in the disease based on the result of the comparison.
61. A method according to item 60.
Item 62.
The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The method according to any one of items 60 to 61, which is at least one selected from the group consisting of: Col1a1 and Defb8.
Item 63.
Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4 dip, I22ra2, Rbfox1, Lrcc2, Col1a1, Col5a1, C1qtnChIh, a C1qtnChIh, a C1qtnCh, a backgrip A skin biomarker for predicting involvement of renal function, abnormal phosphorus metabolism, or FGF23, comprising at least one selected from the group consisting of
 本発明によれば、一つの効果として、皮膚由来検体を用いて腎機能の低下を発見することができる。本発明は、一つの効果として、皮膚由来検体を用いてリンの代謝異常を予測することができる。本発明は、一つの効果として、皮膚由来検体を用いて被験体におけるFGF23の活性化を予測することができる。本発明は、一つの効果として、皮膚由来検体を用いてFGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングすることができる。本発明は、一つの効果として、皮膚由来検体を用いてFGF23の機能を活性化するための有効成分の候補物質をスクリーニングすることができる。本発明は、一つの効果として、皮膚由来検体を用いて疾患におけるFGF23の関与を予測することができる。 According to the present invention, as an effect, it is possible to detect a decrease in renal function using a skin-derived sample. The present invention can predict, as one effect, a metabolic abnormality of phosphorus using a skin-derived sample. The present invention can predict the activation of FGF23 in a subject using a skin-derived sample as one effect. The present invention can screen for a candidate substance of an active ingredient for suppressing functional expression of FGF23 using skin derived specimen as one effect. The present invention can screen for a candidate substance of an active ingredient for activating the function of FGF23 using skin derived specimen as one effect. The present invention can, as one effect, predict the involvement of FGF23 in disease using skin-derived specimens.
本発明の第1~第3の実施態様に係るシステム100の概観図である。FIG. 1 is a schematic view of a system 100 according to first to third embodiments of the present invention. 本発明の第1~第3の実施態様に係るシステム100のハードウェア構成を示す図である。It is a figure showing hardware constitutions of system 100 concerning the 1st-the 3rd embodiment of the present invention. 本発明の本発明の第1~第3の実施態様に係る演算装置1~3の機能を説明するためのブロック図である。FIG. 6 is a block diagram for explaining the functions of arithmetic units 1 to 3 according to first to third embodiments of the present invention of the present invention. 本発明の第1の態様に係る演算装置1の動作を示すフローチャートである。It is a flow chart which shows operation of arithmetic unit 1 concerning the 1st mode of the present invention. 本発明の第2の態様に係る演算装置2の動作を示すフローチャートである。It is a flow chart which shows operation of arithmetic unit 2 concerning the 2nd mode of the present invention. 本発明の第3の態様に係る演算装置3の動作を示すフローチャートである。It is a flow chart which shows operation of arithmetic unit 3 concerning the 3rd mode of the present invention. 本発明の第4、第5の実施態様に係るシステム200の概観図である。FIG. 10 is a schematic view of a system 200 according to the fourth and fifth embodiments of the present invention. 本発明の第4、第5の実施態様に係るシステム200のハードウェア構成を示す図である。It is a figure which shows the hardware constitutions of the system 200 which concerns on the 4th, 5th embodiment of this invention. 本発明の本発明の第4、第5の実施態様に係るスクリーニング装置4、5の機能を説明するためのブロック図である。It is a block diagram for demonstrating the function of the screening apparatuses 4 and 5 which concern on the 4th, 5th embodiment of this invention of this invention. 本発明の第4の態様に係るスクリーニング装置4の動作を示すフローチャートである。It is a flowchart which shows operation | movement of the screening apparatus 4 which concerns on the 4th aspect of this invention. 本発明の第5の態様に係るスクリーニング装置5の動作を示すフローチャートである。It is a flowchart which shows operation | movement of the screening apparatus 5 which concerns on the 5th aspect of this invention. 本発明の第6の実施態様に係るシステム300の概観図である。FIG. 16 is a schematic view of a system 300 according to a sixth embodiment of the present invention. 本発明の第6の実施態様に係るシステム300のハードウェア構成を示す図である。It is a figure which shows the hardware constitutions of the system 300 which concerns on the 6th embodiment of this invention. 本発明の本発明の第6の実施態様に係る演算装置6の機能を説明するためのブロック図である。It is a block diagram for demonstrating the function of the arithmetic unit 6 which concerns on the 6th embodiment of this invention of this invention. 本発明の第6の態様に係る演算装置6の動作を示すフローチャートである。It is a flowchart which shows operation | movement of the arithmetic unit 6 which concerns on the 6th aspect of this invention. 検査キットの一例を示す概観図である。It is an outline figure showing an example of a test kit. 図17Aは、gRNA及びssODNsの配列を示す。図17Bは変異マウスのジェノタイプを示す。FIG. 17A shows the sequences of gRNA and ssODNs. FIG. 17B shows the genotype of mutant mice. UNx/HPi、WT3W/HP1W、WT3W/LP1W、WT3W/ND1Wマウスモデルの皮膚におけるバイオマーカーの発現を示す。The expression of biomarkers in the skin of UNx / HPi, WT3W / HP1W, WT3W / LP1W, WT3W / ND1W mouse models is shown. E-/M-/L-UNx/HPi、及びWT3W/HP1W、WT3W/LP1W、WT3W/ND1Wの頭蓋骨においけるFgf23の発現を示す図である。It is a figure which shows expression of Fgf23 in the skull of E- / M- / L-UNx / HPi and WT3W / HP1W, WT3W / LP1W, WT3W / ND1W. 各モデルマウスの血中無機リン濃度を示す。The blood inorganic phosphorus concentration of each model mouse is shown. Fgf23遺伝子変異マウスとWT3W/HP1W、WT3W/LP1W、WT3W/ND1Wにおけるバイオマーカーの発現の比較結果を示す。The comparison result of the expression of the biomarker in Fgf23 gene mutant mouse and WT3W / HP1W, WT3W / LP1W, WT3W / ND1W is shown. Fgf23遺伝子機能発現に依存又は非依存性の4タイプのモデルを示す。4 shows four types of models dependent or independent on Fgf23 gene functional expression.
 本発明は、皮膚由来検体を用いて腎疾患を予測するという課題を解決するための第1の実施態様を含む。本発明は、皮膚由来検体を用いてリンの代謝異常を予測するという課題を解決するための第2の実施態様を含む。本発明は、被験体におけるFGF23の活性化を予測するという課題を解決するための第3の実施態様を含む。本発明は、FGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングするという課題を解決するための第4の実施態様を含む。本発明は、FGF23の機能を活性化するための有効成分の候補物質をスクリーニングするという課題を解決するための第5の実施態様を含む。本発明は、疾患におけるFGF23の関与を予測するという課題を解決するための第6の実施態様を含む。本発明は、検査試薬及び検査キットに係る第7の実施態様を含む。本発明は皮膚のバイオマーカーに係る第8の実施態様を含む。 The present invention includes the first embodiment for solving the problem of predicting kidney disease using a skin-derived sample. The present invention includes a second embodiment for solving the problem of predicting metabolic abnormality of phosphorus using a skin-derived sample. The present invention includes the third embodiment for solving the problem of predicting the activation of FGF23 in a subject. The present invention includes the fourth embodiment for solving the problem of screening a candidate substance of an active ingredient for suppressing the functional expression of FGF23. The present invention includes the fifth embodiment for solving the problem of screening candidate substances for active ingredients for activating the function of FGF23. The present invention includes the sixth embodiment for solving the problem of predicting the involvement of FGF23 in diseases. The present invention includes the seventh embodiment of the test reagent and the test kit. The present invention includes the eighth embodiment according to the skin biomarker.
I.第1から第3の実施態様
1.用語の説明
 はじめに、第1から第3の実施態様において、本明細書、請求の範囲、要約で使用される用語について説明する。特に記載がない限り、第1から第3の実施態様に関して本明細書、請求の範囲、要約で使用されている用語は、本項の規定に従う。但し、第4から第8の実施態様の説明においても、本項の説明が援用される場合がある。
I. First to third embodiments
1. Description of Terms First, in the first to third embodiments, terms used in the specification, claims, and summary will be described. Unless otherwise stated, the terms used in the specification, claims and summary with respect to the first to third embodiments conform to the provisions of this section. However, also in the description of the fourth to eighth embodiments, the explanation of this section may be incorporated.
 本明細書において「腎疾患」とは、腎機能低下を来す腎臓の何らかの異常又は疾患であり、腎臓に何らかの機能的、又は物理的な障害がある状態である限り特に制限されない。具体的には、急性腎不全、急性腎盂腎炎、急性糸球体腎炎(溶血連鎖球菌感染症に付随して起こる糸球体腎炎、急速進行性糸球体腎炎等)、心疾患に付随する腎疾患のうち急性の疾患(タイプ1心腎症候群等)等の急性腎疾患;慢性腎盂腎炎、逆流性腎症、間質性腎炎、多発性嚢胞腎、慢性糸球体腎炎(IgA腎症、全身性エリテマトーデス糸球体腎炎(ループス腎炎)等)、心疾患に付随する腎疾患のうち慢性の疾患(タイプ2心腎症候群等)、糖尿病性腎症、腎糸球体線維沈着症等の慢性腎炎;ネフローゼ症候群;腎腫瘍等の慢性腎疾患等が挙げられる。好ましくは、急性腎疾患である。また、別の態様として、好ましくは虚血性心疾患に付随する腎疾患であり、特に虚血性心疾患に付随する急性腎疾患(タイプ1心腎症候群等)である。 In the present specification, “renal disease” is any abnormality or disease of the kidney causing renal function decline, and is not particularly limited as long as the kidney has some functional or physical disorder. Specifically, among acute renal failure, acute pyelonephritis, acute glomerulonephritis (glomerulonephritis associated with hemolytic streptococcal infection, rapidly progressive glomerulonephritis, etc.), renal disease associated with heart disease Acute renal diseases such as acute diseases (type 1 cardiorenal syndrome etc.); chronic pyelonephritis, reflux nephropathy, interstitial nephritis, polycystic kidney disease, chronic glomerulonephritis (IgA nephropathy, systemic lupus erythematosus glomeruli Nephritis (Lupus Nephritis) etc., Chronic disease among chronic kidney disease associated with heart disease (Type 2 cardio-renal syndrome etc), Chronic nephritis such as diabetic nephropathy, glomerular fibrosis, etc .; Nephrotic syndrome; Renal tumor Chronic kidney disease and the like. Preferably, it is acute kidney disease. In another embodiment, it is preferably renal disease associated with ischemic heart disease, particularly acute renal disease associated with ischemic heart disease (type 1 cardiorenal syndrome, etc.).
 本明細書において、「腎機能低下」とは、例えばヒトの場合、一般的に臨床検査で測定されている下記表1-1から表1-4に示す少なくとも一種の腎疾患マーカー(好ましくは尿タンパク質以外)が基準値範囲外となる状態をいう。腎疾患マーカーとして、より好ましくは、血清尿素窒素、血清クレアチニン、血清無機リン、尿中フィブリノーゲン、クレアチニンクリアランス、24時間クレアチニンクリアランス推定糸球体ろ過量(eGFR)、尿素クリアランス、イヌリンクリアランス、チオ硫酸ナトリウムクリアランス、腎血漿流量、ろ過率、ナトリウム排泄率、リチウム排泄率、フェノールスルホンフタレイン試験、濃縮試験、希釈試験、自由水クリアランス、自由水再吸収、尿細管排泄極量、尿細管再吸収極量、リン酸再吸収率、β2-ミクログロブリン、及びα1-ミクログロブリンからなる群から選択される少なくとも一種である。 In the present specification, “renal function reduction” means, for example, at least one type of kidney disease marker (preferably urine) shown in the following Tables 1-1 to 1-4 which are generally measured by clinical examination in the case of human. The condition where the value is outside the standard value range). As renal disease markers, more preferably serum urea nitrogen, serum creatinine, serum inorganic phosphorus, urinary fibrinogen, creatinine clearance, 24 hour creatinine clearance estimated glomerular filtration rate (eGFR), urea clearance, inulin clearance, sodium thiosulfate clearance , Renal plasma flow rate, filtration rate, sodium excretion rate, lithium excretion rate, phenolsulfonephthalein test, concentration test, dilution test, free water clearance, free water resorption, tubular excretory pole volume, tubular resorption peak volume, It is at least one selected from the group consisting of phosphoric acid resorption rate, β2-microglobulin, and α1-microglobulin.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004
 上記腎疾患マーカーは、臨床検査法提要改訂第32版(金井正光編集 金原出版株式会社)等に掲載の公知の方法によって測定することができる。 The above-mentioned kidney disease marker can be measured by a known method disclosed in the Clinical Test Method Revision Edition 32 (Kanai Masamitsu Edited by Kanahara Publishing Co., Ltd.) and the like.
 本明細書において、「急性腎不全」とは、急激に腎機能が低下する疾患であり、たとえば、血清クレアチニン値が2.0~2.5mg/dl以上へ急速に増加したもの(基礎に腎機能低下がある場合には血清クレアチニン値が前値に対して50%以上増加したもの)、又は血清クレアチニン値が0.5mg/dl/day以上,尿素窒素が10mg/dl/day以上の速度で増加するものをいう。急性腎不全には、(1)腎血流量の減少が原因となる腎前性急性腎不全、(2)腎実質に障害がある腎性急性腎不全、(3)腎以降の尿流障害による腎後性急性腎不全が含まれるが、本発明の適用対象として好ましい急性腎不全は、腎前性急性腎不全及び腎性急性腎不全であり、好ましくは腎前性急性腎不全である。 In the present specification, “acute renal failure” is a disease in which renal function is rapidly reduced, for example, those in which the serum creatinine level rapidly increases to 2.0 to 2.5 mg / dl or more (based on the kidney If there is a decline in function, serum creatinine level increases by 50% or more from the previous level), or serum creatinine level is 0.5 mg / dl / day or more, and urea nitrogen is 10 mg / dl / day or more I say something that increases. Acute renal failure includes (1) prerenal acute renal failure due to decreased renal blood flow, (2) renal acute renal failure with impaired renal parenchyma, and (3) renal failure after renal failure Although post-renal acute renal failure is included, preferred acute renal failure for application of the present invention is prerenal acute renal failure and renal acute renal failure, preferably prerenal acute renal failure.
 心疾患に付随する腎疾患は、心腎症候群とも呼ばれる。心腎症候群は、急性及び慢性の病態を含み、複数のタイプに分類されている。このうち心疾患がきっかけで発症するのは、タイプ1とタイプ2であり、タイプ1は急性心腎症候群、タイプ2は慢性心腎症候群である。タイプ1は虚血性心疾患等が引き金となって発症することがある。 Renal disease associated with heart disease is also called cardiorenal syndrome. Cardiorenal syndromes, including acute and chronic conditions, are classified into multiple types. Of these, types 1 and 2 start with heart disease, type 1 is acute cardiorenal syndrome, and type 2 is chronic cardiorenal syndrome. Type 1 may be triggered by ischemic heart disease or the like.
 タイプ1心腎症候群は、何らかの心疾患により、以下の病期1~3のいずれかの病期に該当するようになった状態をいう:
病期1:血清クレアチニン値が基準値の1.5~1.9倍程度に増加するか、又は同一個体において血清クレアチニン値が前値に対して0.3mg/dl以上増加し、かつ尿量が6時間~12時間にわたって0.5mL/kg/時程度である
病期2:血清クレアチニン値が基準値の2.0~2.9倍程度に増加し、かつ12時間以上尿量が0.5mL/kg/時未満の状態が続く
病期3:血清クレアチニン値が基準値の3倍程度まで増加するか、同一個体において血清クレアチニン値が前値に対して4.0mg/dL以上増加するか、腎代替療法が開始されるか、又は18歳未満の患者の場合eGFRが35mL/min/1.73m未満に低下した場合であり、前記4つのいずれかの状態に加え、尿量が24時間以上0.3mL/kg/時未満である状態が続いているか、又は12時間以上無尿の状態が続いている場合。
Type 1 cardiorenal syndrome refers to any heart disease resulting in one of the following stages 1-3:
Stage 1: Serum creatinine level increases to about 1.5 to 1.9 times the standard value, or in the same individual, serum creatinine level increases by 0.3 mg / dl or more over the previous level, and urine volume Stage 2: The blood pressure is about 0.5 mL / kg / hour over 6 hours to 12 hours: The serum creatinine level increases to 2.0 to 2.9 times the standard value, and the urine volume is 0. Stage 3: The condition of less than 5 mL / kg / hour continues: Does the serum creatinine level increase to about 3 times the standard value, or does the serum creatinine level increase by 4.0 mg / dL or more over the previous level in the same individual , Or if renal replacement therapy is started or eGFR drops to less than 35 mL / min / 1.73 m 2 in patients younger than 18 years, urine volume is 24 in addition to any of the above four conditions. 0.3 mL / kg / hour or more If the condition of less than hour continues or the condition of urinelessness continues for 12 hours or more.
 本明細書において、「慢性腎疾患」とは、被験体がヒトである場合、「CKD診断ガイド2012(日本腎臓学会編)に従い、腎臓の障害(微量アルブミン尿を含む蛋白尿などの尿異常、尿沈渣の異常、片腎や多発性のう胞腎などの画像異常、血清クレアチニン値上昇などの腎機能低下、尿細管障害による低カリウム血症などの電解質異常、腎生検などで病理組織検査の異常等)、もしくは推定GFR(糸球体濾過量)60mL/分/1.73m未満の腎機能低下が3カ月以上持続する状態をいう。 In the present specification, “chronic kidney disease” means, when the subject is a human, according to “CKD Diagnosis Guide 2012 (The Japanese Society of Nephrology), renal disorders (urinary abnormalities such as proteinuria including micro albuminuria), Abnormal urinary sediment, abnormal imaging such as single kidney and multiple cystic kidney, decreased renal function such as increase in serum creatinine level, abnormal electrolyte such as hypokalemia due to renal tubular disorder, abnormal pathology in renal biopsy etc. Etc.), or estimated GFR (glomerular filtration rate) of 60 mL / min / 1.73 m 2 or less is a state in which renal function decline lasts for 3 months or more.
 ここで、推算GFR(eGFR)は、下記表2に示す血清クレアチニン値からの推算式(eGFRcreat)で算出ことができる。また、下肢切断者等の筋肉量の極端に少ない者に対しいては、血清シスタチンCの推算式(eGFRcys)を適用することができる。 Here, the estimated GFR (eGFR) can be calculated by the estimation formula (eGFR Creat) from the serum creatinine value shown in Table 2 below. In addition, for those with extremely low muscle mass, such as lower extremity amputees, the serum cystatin C estimation formula (eGFRcys) can be applied.
Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000005
 例えば、タンパクを指標とする場合、3カ月以上前と最近の尿検査で尿蛋白0.15g/gCr以上が続いている場合を、慢性腎疾患であると診断することができる。また、糖尿病で3カ月以上前と最近のアルブミン尿検査で30mg/gCr以上が続いている場合を、慢性腎疾患であると診断することができる。 For example, in the case where protein is used as an index, chronic renal disease can be diagnosed when urine protein of 0.15 g / g Cr or more continues by urinalysis three months ago and recent. In addition, it is possible to diagnose chronic kidney disease as a case where diabetes continues more than 3 months ago and 30 mg / g Cr or more continues in the recent albuminuria test.
 小児に関しては、日本人小児の酵素法による血清クレアチニン(Cr)の基準値が作成され,これを使用して腎機能異常者の評価を行うことができる。例えば、%表示のeGFRは、2歳以上11歳以下の小児については、下記式1で表すことができる。
[数式1]
eGFR(%)=(0.3×身長(m)/被験体の血清Cr値)×100
For children, a standard value of serum creatinine (Cr) is prepared by the enzymatic method in Japanese children, and it can be used to evaluate people with renal dysfunction. For example, the eGFR in% can be represented by the following formula 1 for children between 2 and 11 years old.
[Equation 1]
eGFR (%) = (0.3 x height (m) / subject's serum Cr value) x 100
 また、ネコ、イヌ等のヒト以外の哺乳動物の場合、1日平均引水量、又は尿比重等から慢性腎疾患であることを予測することができる。 In addition, in the case of mammals other than humans, such as cats and dogs, chronic kidney disease can be predicted from the average daily water intake or specific gravity of urine.
 慢性腎疾患の重症度は、例えば、ヒトの場合、下記表3に基づいて決定することができる(表3は、「CKD診断ガイド2012」の表2)。 For example, in the case of humans, the severity of chronic kidney disease can be determined based on Table 3 below (Table 3 is Table 2 of “CKD Diagnosis Guide 2012”).
Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006
 FGF23が関連する疾患としては、FGF23の過剰発現に伴って発症する疾患である限り、制限されない。低リン血症性くる病/骨軟化症、常染色体性優性低リン血症性くる病/骨軟化症、常染色体性劣性低リン血症性くる病/骨軟化症、X染色体性優性低リン血症性くる病/骨軟化症、腫瘍性くる病/骨軟化症、二次性副甲状腺機能亢進症、急性腎疾患又は慢性腎疾患におけるリン代謝異常、骨-ミネラル代謝異常(CKD-MBD)等を挙げることができる。 The disease associated with FGF23 is not limited as long as it is a disease that develops with overexpression of FGF23. Hypophosphatemia rickets / osteomalacia, autosomal dominant hypophosphatemia rickets / osteomalacia, autosomal recessive hypophosphatemia rickets / osteomalacia, X-chromosomal dominant hypophosphate Rickets / osteomalacia, neoplastic rickets / osteomalacia, secondary hyperparathyroidism, abnormal phosphorus metabolism in acute kidney disease or chronic kidney disease, bone-mineral metabolism disorder (CKD-MBD) Etc. can be mentioned.
 本発明において、「個体」は、特に制限されないが、個体にはヒト及びヒト以外の哺乳動物が含まれる。ヒト以外の哺乳動物としては、例えばウシ、ウマ、ヒツジ、ヤギ、ブタ、イヌ、ネコ、ウサギ、サル等が挙げられる。好ましくは、ヒト、ネコ、イヌである。また、個体の年齢、性別は問わない。 In the present invention, the "individual" is not particularly limited, but the individual includes humans and mammals other than humans. Examples of mammals other than human include cows, horses, sheep, goats, pigs, dogs, cats, rabbits, monkeys and the like. Preferred are humans, cats and dogs. Also, the age and gender of the individual do not matter.
 また、「被験体」は、腎機能低下やその他の腎疾患の既往を有している個体であっても、有していない個体であってもよい。また、被験体は、多尿、喉の渇き、飲水量の増加、胃液過多、嘔吐、血尿、及び全身倦怠感等の症状がある個体であってもよく、また無症状の個体であってもよい。さらに、被験体には、医療面接、尿検査、血液の生化学的検査、腎像画像診断、腎生検等の公知の診断方法に従って、腎障害や慢性腎疾患が疑われた被験体も含まれる。 In addition, the “subject” may or may not be an individual having renal function decline or a history of other renal diseases. In addition, the subject may be an individual with symptoms such as polyuria, thirst, increased water intake, excessive gastric juice, vomiting, hematuria, general fatigue, or even an asymptomatic individual Good. Furthermore, subjects include subjects suspected of having renal disorder or chronic kidney disease according to known diagnostic methods such as medical interviews, urinalysis, blood biochemistry, renal imaging, renal biopsy etc. Be
 本発明において、「皮膚由来検体」は、生体の皮膚に由来する限り制限されない。また、皮膚由来検体には、汗、皮膚からの分泌液等が含まれる。皮膚の採取方法も制限されず、生検材料、ピアスの穴を空けた際に穿孔針に付着する皮膚、皮膚表面の擦過物等を挙げることができる。 In the present invention, the "skin-derived sample" is not limited as long as it is derived from the skin of a living body. The skin-derived sample also includes sweat, secretions from the skin, and the like. The method of collecting the skin is also not limited, and examples thereof include a biopsy material, skin adhering to a perforated needle when a hole in a piercing is made, abrasion on the skin surface, and the like.
 さらに、検体は、新鮮なものであっても、保存されていたものであってもよい。検体を保存する場合には、室温環境、冷蔵環境又は冷凍環境において保存することができるが、好ましくは冷凍保存である。 Furthermore, the sample may be fresh or stored. When the sample is stored, it can be stored in a room temperature environment, a refrigerated environment or a frozen environment, but is preferably frozen.
 本発明の「バイオマーカー」には、表4に示されるHamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種及びこれらのバリアント、オーソログ等が含まれる。 In the “biomarker” of the present invention, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1, Aldh1b1 shown in Table 4 are included. And at least one selected from the group consisting of Sparc, Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8, variants thereof, orthologs and the like.
Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007
 ここで、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカー(1群と呼ぶ)については、FGF23の制御を受ける可能性が高い。したがって、1群のバイオマーカーの測定値が基準値の範囲外である場合には、例えば、腎疾患その他の疾患、又はリン代謝異常において、FGF23が関与していると予測することができる。 Here, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, & At least one biomarker (referred to as group 1) is likely to be under the control of FGF23. Therefore, when the measured value of one group of biomarkers is out of the range of the reference value, for example, it can be predicted that FGF23 is involved in, for example, renal diseases and other diseases, or abnormal phosphorus metabolism.
 一方、Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種バイオマーカー(2群と呼ぶ)はFGF23の制御を受けない可能性が高い。したがって、1群のバイオマーカーが基準値の範囲外となっており、かつ2群のバイオマーカーの測定値が基準値の範囲内である場合には、例えば、腎疾患その他の疾患、又はリン代謝異常において、FGF23が関与していることをより確実に予測することができる。 On the other hand, at least one biomarker selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 (referred to as group 2) is likely not to be regulated by FGF23. Therefore, if one group of biomarkers is out of the range of the reference value, and if the measured values of the two groups of biomarkers are within the range of the reference value, for example, renal disease or other diseases, or phosphorus metabolism In the abnormality, it can be predicted more reliably that FGF23 is involved.
 FGF23が関与しているか否かは、1群のバイオマーカーの測定値と2群のバイオマーカーの測定値の双方を考慮して決定することが好ましい。 Whether or not FGF23 is involved is preferably determined in consideration of both the measured values of one group of biomarkers and the measured values of two groups of biomarkers.
 「バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値」とは、バイオマーカーからなる群から選択される少なくとも一種のタンパク質の量又は濃度を反映した値をいう。当該測定値を「量」で標記する場合には、モルであっても質量であってもよいが、質量で標記することが好ましい。また、値を「濃度」で表記する場合には、モル濃度であっても検体の一定容量あたりの質量の割合(質量/容量)であってもよいが、好ましくは質量/容量である。量又は濃度を反映する値としては、上記の他に、蛍光や発光などのシグナルの強度であってもよい。 The “measurement value of at least one type of protein selected from the group consisting of biomarkers” refers to a value reflecting the amount or concentration of at least one type of protein selected from the group consisting of biomarkers. When the said measured value is labeled by "amount", although it may be molar or mass, it is preferable to label by mass. In addition, when a value is expressed as “concentration”, it may be a molar concentration or a mass ratio (mass / volume) per a fixed volume of an analyte, but is preferably mass / volume. As the value reflecting the amount or concentration, in addition to the above, the intensity of a signal such as fluorescence or luminescence may be used.
 「バイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値」とは、一定量の検体中に存在するバイオマーカー mRNAのコピー数(絶対量)で表されてもよく、又は、β2-ミクログロブリンmRNA、GAPDH mRNA、Maea mRNA及びβ-アクチン mRNA等のハウスキーピング遺伝子の発現量に対する相対発現量を反映した値であってもよい。また、蛍光や発光などのシグナルの強度で表されてもよい。 The “measured value of at least one mRNA selected from the group consisting of biomarkers” may be represented by the copy number (absolute amount) of biomarker mRNA present in a certain amount of sample, or β2- It may be a value reflecting the expression level relative to the expression level of housekeeping genes such as microglobulin mRNA, GAPDH mRNA, Maea mRNA and β-actin mRNA. Moreover, it may be expressed by the intensity of signals such as fluorescence and luminescence.
 バイオマーカータンパク質の測定値の「所定の基準値」は、腎機能低下を来した個体の検体中のバイオマーカータンパク質の測定値、及び/又は健常個体の検体中のバイオマーカータンパク質の測定値に基づいて決定される基準値をいう。具体的には、被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値(被験測定値ともいう)と、被験測定値に対応する健常個体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値(健常測定値ともいう)と、に基づいて、基準値は公知の方法により決定される。ここで「対応する」とは、同種のバイオマーカーであることを意図する。 The "predetermined reference value" of the measured value of the biomarker protein is based on the measured value of the biomarker protein in the sample of the individual who has developed renal function decline and / or the measured value of the biomarker protein in the sample of the healthy individual Reference value to be determined. Specifically, it is contained in a measured value of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from a subject, and / or contained in a sample collected from the subject A group consisting of a measured value (also referred to as a test measured value) of at least one mRNA selected from the group consisting of the biomarkers, and a biomarker contained in a skin-derived sample collected from a healthy individual corresponding to the test measured value And / or a measured value of at least one mRNA selected from the group consisting of a biomarker included in a sample collected from the subject (also referred to as a healthy measurement value) The reference value is determined by known methods on the basis of. Here, "corresponding" is intended to be a homogeneous biomarker.
 たとえば、腎機能低下を来した複数の個体の検体を用いて測定したバイオマーカータンパク質の測定値と、複数の健常個体の検体を用いて測定したバイオマーカータンパク質の測定値とを取得する。これらの複数の値に基づいて腎機能低下の有無を最も精度よく判定できる値を「基準値」とすることができる。ここで、「最も精度よく判定できる値」は、検査の目的によってROC曲線から求められる感度及び/又は特異度、陽性的中率、陰性的中率、四分位範囲の第1四分位、第2四分位(中央値)、第3四分位などの指標に基づいて適宜設定することができる。 For example, measurement values of biomarker proteins measured using specimens of a plurality of individuals having decreased renal function and measurements of biomarker proteins measured using specimens of a plurality of healthy individuals are obtained. A value that can most accurately determine the presence or absence of renal function deterioration based on the plurality of values can be used as the “reference value”. Here, "the most accurately determined value" is the sensitivity and / or specificity, the positive predictive value, the negative predictive value, the first quartile range of the quartile range, which are determined from the ROC curve depending on the purpose of the test. It can set suitably based on indices, such as the 2nd quartile (median value) and the 3rd quartile.
 例えば一態様として複数の健常個体から得られたそれぞれの検体中のバイオマーカータンパク質の測定値の中で、最も高い測定値を基準値としてもよい。 For example, among the measured values of the biomarker protein in each sample obtained from a plurality of healthy individuals in one aspect, the highest measured value may be used as the reference value.
 また、別の態様として、腎機能低下を来した個体の検体中のバイオマーカータンパク質の測定値に基づいて基準値を決定する場合には、腎機能低下を来した複数の個体の検体中のバイオマーカータンパク質の測定値の中から、最も低い測定値を閾値と決定することができる。 In another embodiment, when the reference value is determined based on the measured value of the biomarker protein in the sample of an individual who has developed renal function, the biotechniques in the samples of multiple individuals who have developed renal function have been described. Among the measurement values of the marker protein, the lowest measurement value can be determined as the threshold value.
 また別の態様として、基準値は、健常個体の検体中のバイオマーカータンパク質の測定値そのもの、又は健常個体の複数のバイオマーカータンパク質の測定値の平均値、中央値又は最頻値とすることもできる。 In another embodiment, the reference value may be a measured value of the biomarker protein itself in a sample of a healthy individual, or an average, median or mode of measured values of a plurality of biomarker proteins of a healthy individual. it can.
 またさらに、基準値として、同一被験体であって当該被験体が健常な状態である時に取得された過去のバイオマーカータンパク質の測定値(一つの値でもよいし、複数の値の平均値、中央値、最頻値などであってもよい)を使用することもできる。 Furthermore, as a reference value, a measurement value of a past biomarker protein (which may be a single value or an average value of a plurality of values, which is the same subject and obtained when the subject is in a healthy state) Values, modes, etc. may also be used.
 バイオマーカー mRNAの測定値の「基準値」についても、上記バイオマーカータンパク質の測定値の「基準値」と同様に、バイオマーカータンパク質の測定値に代えてバイオマーカー mRNAの測定値を使用して決定することができる。 The “reference value” of the measured value of the biomarker mRNA is also determined using the measured value of the biomarker mRNA instead of the measured value of the biomarker protein, as in the “reference value” of the measured value of the biomarker protein described above. can do.
 ここで「基準値の範囲」とは、バイオマーカータンパク質が腎機能の低下に伴って発現や活性が上昇する性質のものであれば、基準範囲は基準値以下である。バイオマーカータンパク質が腎機能の低下に伴って発現や活性が低下する性質のものであれば、基準範囲は基準値以上である。例えば、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、及びCol5a1は、腎機能低下に伴って発現が上昇する。C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8は、腎機能低下に伴って発現が低下する。 Here, the “range of reference value” means that the reference range is equal to or less than the reference value as long as the biomarker protein has a property that expression and activity increase with the decrease in renal function. If the biomarker protein is of a nature that decreases in expression and activity as renal function declines, the reference range is above the reference value. For example, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Aldh1l2, and Col5a1 have decreased expression of renal function. The expression of C1qtnf6, Sparc, Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8 decreases with the decrease in renal function.
 「健常個体」とは、特に制限されない。好ましくは「個体」の項に記載されたヒト又はヒト以外の哺乳動物であって、生化学的検査、血液検査、尿検査、血清検査、生理学的検査等において異常データを示さない個体をいう。健常個体の年齢、性別は特に制限されない。 The "healthy individual" is not particularly limited. The term preferably refers to a human or non-human mammal described in the "individual" section, which does not show abnormal data in biochemical tests, blood tests, urine tests, serological tests, physiological tests and the like. The age and sex of healthy individuals are not particularly limited.
 「複数の検体」とは、2以上、好ましくは5以上、より好ましくは10以上の検体である。これらは、異なる個体から採取された検体であってもよいし、採取時が異なる同一個体の複数の検体であってもよい。 The “plural samples” are two or more, preferably five or more, and more preferably ten or more. These may be specimens collected from different individuals, or may be a plurality of specimens of the same individual different in collection time.
 「複数の値」とは、2以上、好ましくは5以上、より好ましくは10以上のバイオマーカータンパク質の測定値及びバイオマーカー mRNAの測定値である。 "Multiple values" are measurements of biomarker protein of 2 or more, preferably 5 or more, more preferably 10 or more and biomarker mRNA.
 「複数の個体」とは、2以上、好ましくは5以上、より好ましくは10以上の個体である。 The "plural individuals" are 2 or more, preferably 5 or more, more preferably 10 or more individuals.
 基準値を決定するためにバイオマーカータンパク質の測定値及びバイオマーカー mRNAの測定値を取得する個体と、被験体の種、年齢、性別等を必ずしも同じくする必要はないが、好ましくは同種である。また、前記個体は被験体と同年代及び/又は同一性別であることが好ましい。 The individual who obtains the measurement value of the biomarker protein and the measurement value of the biomarker mRNA to determine the reference value is not necessarily the same as the species, age, sex, etc. of the subject, but is preferably the same. Preferably, the individual is of the same age and / or the same gender as the subject.
 「抗バイオマーカー抗体」は、上述したバイオマーカーからなる群から選択される少なくとも一種のタンパク質と特異的に結合する限り制限はなく、バイオマーカーからなる群から選択される少なくとも一種のタンパク質又はその一部を抗原としてヒト以外の動物に免疫して得られたポリクローナル抗体、モノクローナル抗体、及びそれらの断片(例えば、Fab、F(ab)等)のいずれも用いることができる。また、免疫グロブリンのクラス及びサブクラスは特に制限されない。また、キメラ抗体であってもよい。さらに、scFv等であってもよい。 The "anti-biomarker antibody" is not particularly limited as long as it specifically binds to at least one protein selected from the group consisting of the aforementioned biomarkers, and at least one protein selected from the group consisting of biomarkers or one of them Any of polyclonal antibodies, monoclonal antibodies, and fragments thereof (for example, Fab, F (ab) 2 etc.) obtained by immunizing animals other than human with an antibody as an antigen can be used. Also, the class and subclass of immunoglobulin are not particularly limited. It may also be a chimeric antibody. Furthermore, it may be scFv or the like.
 抗バイオマーカー抗体を作製するために用いられる、抗原となるバイオマーカータンパク質としては、上述したバイオマーカーからなる群から選択される少なくとも一種のタンパク質の全体、又は一部を挙げることができる。 As a biomarker protein used as an antigen used to produce an anti-biomarker antibody, all or a part of at least one type of protein selected from the group consisting of the above-mentioned biomarkers can be mentioned.
 本発明における「バイオマーカー mRNA検出核酸」は、上述したバイオマーカーからなる群から選択される少なくとも一種のmRNA、又は当該mRNAの逆転写産物と特異的にハイブリダイズする配列を含む限り制限されない。検出核酸は、DNAであっても、RNAであってもよく、また、検出核酸に含まれるヌクレオチドは、天然のヌクレオチドであっても人工的に合成されたヌクレオチドであってもよい。 The “biomarker mRNA detection nucleic acid” in the present invention is not limited as long as it contains at least one mRNA selected from the group consisting of the above-mentioned biomarkers, or a sequence specifically hybridizing with the reverse transcription product of the mRNA. The detection nucleic acid may be DNA or RNA, and the nucleotides contained in the detection nucleic acid may be natural nucleotides or artificially synthesized nucleotides.
 検出核酸の長さは、特に制限されない。検出核酸が、マイクロアレイ等においてキャプチャープローブとして使用されるものであれば、標的核酸とハイブリダイズする配列が、好ましくは100 mer程度であり、より好ましくは60 mer程度であり、さらに好ましくは、20~30 mer程度である。キャプチャープローブは、公知のオリゴヌクレオチド合成機等を使用して製造することができる。キャプチャープローブには、標的核酸とハイブリダイズしない配列が含まれていてもよい。 The length of the detection nucleic acid is not particularly limited. If the detection nucleic acid is used as a capture probe in a microarray or the like, the sequence hybridizing to the target nucleic acid is preferably about 100 mer, more preferably about 60 mer, and still more preferably It is about 30 mer. The capture probe can be produced using a known oligonucleotide synthesizer or the like. The capture probe may contain a sequence that does not hybridize to the target nucleic acid.
 検出核酸が、PCR反応に使用されるプライマーであれば、標的核酸とハイブリダイズする配列が、好ましくは50 mer程度であり、より好ましくは30 mer程度であり、さらに好ましくは、15~25 mer程度である。プライマーは、公知のオリゴヌクレオチド合成機等を使用して製造することができる。プライマーには、標的核酸とハイブリダイズしない配列が含まれていてもよい。また、プライマーは、蛍光色素等で標識されていてもよい。 If the detection nucleic acid is a primer used in a PCR reaction, the sequence that hybridizes to the target nucleic acid is preferably about 50 mer, more preferably about 30 mer, and still more preferably about 15 to 25 mer It is. The primers can be produced using a known oligonucleotide synthesizer or the like. The primers may contain sequences that do not hybridize to the target nucleic acid. Also, the primer may be labeled with a fluorescent dye or the like.
 また、RT-PCRには、プライマーの他にPCR産物のリアルタイムの定量のために使用される、PCR反応中に分解される定量用プローブを使用することもできる。定量用プローブも標的核酸とハイブリダイズする限り、制限されない。定量用プローブは、標的核酸とハイブリダイズする配列を含む、5~20 mer程度の核酸であることが好ましい。さらに定量用プローブの一端には、蛍光色素が標識され、定量用プローブのもう一端には、当該蛍光色素のクエンチャーが標識されていることが好ましい。 In addition to primers, RT-PCR can also use quantitative probes that are degraded during PCR reactions, which are used for real-time quantification of PCR products. The quantitative probe is also not limited as long as it hybridizes to the target nucleic acid. The quantitative probe is preferably a nucleic acid of about 5 to 20 mer, which contains a sequence that hybridizes to a target nucleic acid. Furthermore, it is preferable that a fluorescent dye be labeled at one end of the quantitative probe, and a quencher of the fluorescent dye be labeled at the other end of the quantitative probe.
2.各測定値の取得方法
 本発明におけるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及びバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値の取得方法は、それぞれの測定値が取得できる限り制限されない。例えば、以下に述べる方法に従って取得することができる。
2. Method of Obtaining Each Measured Value A method of obtaining a measured value of at least one type of protein selected from the group consisting of biomarkers in the present invention and at least one type of mRNA measurement value selected from the group consisting of biomarkers It is not limited as long as the measured value can be obtained. For example, it can be obtained according to the method described below.
2-1.バイオマーカーのタンパク質の測定値の取得
 バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値(以下、本明細書において「バイオマーカータンパク質の測定値」と略記することもある)を取得する場合、本工程では、当該測定値を取得するために、上記「1.用語の説明」で述べた抗バイオマーカー抗体を用いることが可能である。また、後述する抗バイオマーカー抗体を含む検査試薬を使用してもよい。
2-1. Acquisition of measurement value of protein of biomarker The measurement value of at least one protein selected from the group consisting of biomarkers (hereinafter, sometimes abbreviated as "measurement value of biomarker protein" in the present specification) is obtained In this case, in this step, it is possible to use the anti-biomarker antibody described in the above "1. Description of terms" to obtain the measurement value. Moreover, you may use the test reagent containing the anti-biomarker antibody mentioned later.
 バイオマーカータンパク質の測定値を取得する方法としては、公知のELISA法等を使用して行うことができる。 As a method of acquiring the measured value of a biomarker protein, it can carry out using well-known ELISA method etc.
 本実施態様では、あらかじめ抗原捕捉用の抗バイオマーカー抗体をマイクロプレート等の固相上に固定化し、固定化された抗バイオマーカー抗体と検体中のバイオマーカータンパク質の複合体を形成させることができる。固相上に固定化された当該複合体又は固相上で形成された複合体を、当該技術において公知の方法で検出することにより、検体に含まれるバイオマーカータンパク質の量又は濃度を測定することができる。 In this embodiment, an anti-biomarker antibody for capturing an antigen can be immobilized in advance on a solid phase such as a microplate to form a complex of the immobilized anti-biomarker antibody and a biomarker protein in a sample. . Measuring the amount or concentration of the biomarker protein contained in the sample by detecting the complex immobilized on the solid phase or the complex formed on the solid phase by a method known in the art Can.
 抗原捕捉用の抗バイオマーカー抗体の固相への固定の方法は、特に限定されない。公知の方法を使用して、直接的に、又は別の物質を介して間接的に行うことができる。直接の結合としては、例えば、物理的吸着などが挙げられる。好ましくは、イムノプレート等を使用して、直接抗バイオマーカー抗体をマイクロプレートに物理的に結合させることができる。 The method of fixing the anti-biomarker antibody for antigen capture to the solid phase is not particularly limited. It can be carried out directly or indirectly via another substance using known methods. Examples of direct binding include physical adsorption and the like. Preferably, the anti-biomarker antibody can be physically bound to the microplate directly using an immuno plate or the like.
 固相の素材は特に限定されず、例えば、ポリスチレン、ポリプロピレンなどが挙げられる。固相の形状は特に限定されず、例えば、マイクロプレート、マイクロチューブ、試験管などが挙げられる。 The material of the solid phase is not particularly limited, and examples thereof include polystyrene and polypropylene. The shape of the solid phase is not particularly limited, and examples thereof include microplates, microtubes, test tubes and the like.
 本方法においては、前記複合体の形成に続いて、固相を洗浄する操作を含んでもよい。洗浄する場合には、界面活性剤等を含むPBS等を使用することができる。 In the present method, following the formation of the complex, an operation of washing the solid phase may be included. In the case of washing, PBS containing a surfactant or the like can be used.
 本方法において、前記複合体の検出は、標識物質で標識された検出用抗バイオマーカー抗体を使用して行うか、未標識の抗バイオマーカー抗体と当該未標識の抗バイオマーカー抗体と結合することができる標識物質で標識された抗イムノグロブリン抗体等を使用して行うことができるが、標識された検出用抗バイオマーカー抗体を使用することが好ましい。また、検出用抗バイオマーカー抗体のバイオマーカータンパク質におけるエピトープと抗原捕捉用抗バイオマーカー抗体のバイオマーカータンパク質におけるエピトープとは異なることが好ましい。 In this method, the detection of the complex is carried out using a detection anti-biomarker antibody labeled with a labeling substance, or an unlabeled anti-biomarker antibody and the unlabeled anti-biomarker antibody are bound to each other. Although it can be carried out using an anti-immunoglobulin antibody etc. labeled with a labeling substance that can be used, it is preferable to use a labeled detection anti-biomarker antibody. Furthermore, it is preferable that the epitope in the biomarker protein of the detection anti-biomarker antibody and the epitope in the biomarker protein of the anti-biomarker antibody for antigen capture are different.
 検出用抗バイオマーカー抗体又は標識抗イムノグロブリン抗体に用いられる標識物質は、検出可能なシグナルが生じるかぎり、特に限定されない。例えば、蛍光物質、放射性同位元素、及び酵素等が挙げられる。酵素としては、アルカリホスファターゼ、ペルオキシダーゼ等が挙げられる。蛍光物質としては、フルオレセインイソチオシアネート(FITC)、ローダミン、Alexa Fluor(登録商標)などの蛍光色素、GFPなどの蛍光タンパク質などが挙げられる。放射性同位元素としては、125I、14C、32Pなどが挙げられる。それらの中でも、標識物質として、アルカリホスファターゼ、又はペルオキシダーゼが好ましい。 The labeling substance used for the detection anti-biomarker antibody or the labeled anti-immunoglobulin antibody is not particularly limited as long as a detectable signal is generated. For example, fluorescent substances, radioactive isotopes, enzymes and the like can be mentioned. As the enzyme, alkaline phosphatase, peroxidase and the like can be mentioned. Examples of fluorescent substances include fluorescein isothiocyanate (FITC), rhodamine, fluorescent dyes such as Alexa Fluor (registered trademark), fluorescent proteins such as GFP, and the like. As radioactive isotopes, 125 I, 14 C, 32 P and the like can be mentioned. Among them, alkaline phosphatase or peroxidase is preferable as a labeling substance.
 検出用抗バイオマーカー抗体は、当該技術において公知の標識方法により、抗バイオマーカー抗体を上記の標識物質で標識して得られる。また、市販のラベリングキットなどを用いて標識してもよい。また、標識イムノグロブリン抗体は、抗バイオマーカー抗体の標識と同じ手法を用いてもよいし、市販のものを使用してもよい。 The anti-biomarker antibody for detection is obtained by labeling the anti-biomarker antibody with the above-mentioned labeling substance by a labeling method known in the art. Moreover, you may label using a commercially available labeling kit etc. Also, the labeled immunoglobulin antibody may use the same method as the labeling of the anti-biomarker antibody, or a commercially available one may be used.
 本実施態様では、複合体に含まれる標識抗バイオマーカー抗体の標識物質により生じるシグナルを検出することにより、検体に含まれるバイオマーカーの測定値を取得できる。ここで、「シグナルを検出する」とは、シグナルの有無を定性的に検出すること、シグナル強度を定量すること、及び、シグナルの強度を半定量的に検出することを含む。半定量的な検出とは、シグナルの強度を、「シグナル発生せず」、「弱」、「中」、「強」などのように段階的に示すことをいう。本工程では、シグナルの強度を定量的又は半定量的に検出することが好ましい。 In this embodiment, the measurement value of the biomarker contained in the sample can be obtained by detecting the signal generated by the labeled substance of the labeled anti-biomarker antibody contained in the complex. Here, "detecting the signal" includes qualitatively detecting the presence or absence of the signal, quantifying the signal intensity, and detecting the intensity of the signal semi-quantitatively. Semi-quantitative detection means that the intensity of a signal is indicated stepwise such as "no signal generation", "weak", "medium", "strong" and the like. In this step, it is preferable to detect the intensity of the signal quantitatively or semi-quantitatively.
 シグナルを検出する方法は、公知の方法を使用することができる。本方法では、上記の標識物質に由来するシグナルの種類に応じた測定方法を適宜選択することができる。例えば、標識物質が酵素である場合、該酵素に対する基質を反応させることによって発生する光、色などのシグナルを、ルミノメーター、分光光度計などの公知の装置を用いて測定することにより行うことができる。 The method of detecting a signal can use a well-known method. In this method, a measurement method can be appropriately selected according to the type of signal derived from the above-mentioned labeling substance. For example, when the labeling substance is an enzyme, the signal such as light or color generated by reacting a substrate for the enzyme may be measured by using a known device such as a luminometer or a spectrophotometer. it can.
 酵素の基質は、該酵素の種類に応じて公知の基質から適宜選択できる。例えば、酵素としてアルカリホスファターゼを用いる場合、基質としては、CDP-Star(登録商標)(4-クロロ-3-(メトキシスピロ[1, 2-ジオキセタン-3, 2’-(5’-クロロ)トリクシロ[3. 3. 1. 13, 7]デカン]-4-イル)フェニルリン酸2ナトリウム)等の化学発光基質、5-ブロモ-4-クロロ-3-インドリルリン酸(BCIP)、5-ブロモ-6-クロロ-インドリルリン酸2ナトリウム、p-ニトロフェニルリン酸等の発色基質が挙げられる。標識物質がペルオキシダーゼである場合には、テトラメチルベンジジン(TMB)等を挙げることができる。 The substrate for the enzyme can be appropriately selected from known substrates depending on the type of the enzyme. For example, when alkaline phosphatase is used as the enzyme, CDP-Star (registered trademark) (4-chloro-3- (methoxyspiro [1,2-dioxetane-3,2 '-(5'-chloro) trixilo] is used as a substrate. Chemiluminescent substrates such as [3. 3. 1. 13, 7] decane] -4-yl) phenyl phosphate (sodium dibasic), 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 5- Chromogenic substrates such as bromo-6-chloro-indolyl phosphate disodium, p-nitrophenyl phosphate and the like can be mentioned. When the labeling substance is peroxidase, tetramethylbenzidine (TMB) etc. can be mentioned.
 標識物質が放射性同位体である場合は、シグナルとしての放射線を、シンチレーションカウンターなどの公知の装置を用いて測定できる。また、標識物質が蛍光物質である場合は、シグナルとしての蛍光を、蛍光マイクロプレートリーダーなどの公知の装置を用いて測定できる。なお、励起波長及び蛍光波長は、用いた蛍光物質の種類に応じて適宜決定できる。 When the labeling substance is a radioactive isotope, radiation as a signal can be measured using a known device such as a scintillation counter. When the labeling substance is a fluorescent substance, fluorescence as a signal can be measured using a known device such as a fluorescence microplate reader. In addition, an excitation wavelength and a fluorescence wavelength can be suitably determined according to the kind of fluorescent substance used.
 シグナルの検出結果は、バイオマーカータンパク質の測定値として用いることができる。例えば、シグナルの強度を定量的に検出する場合は、シグナル強度の測定値自体又は該シグナル強度の測定値から算出される値を、バイオマーカーのタンパク質の測定値として用いることができる。 The detection result of the signal can be used as a measurement value of a biomarker protein. For example, in the case of quantitatively detecting the intensity of a signal, the measured value of the signal intensity itself or a value calculated from the measured value of the signal intensity can be used as a measured value of the protein of the biomarker.
2-2.バイオマーカー遺伝子の測定値の取得
 バイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値(以下、本明細書において「バイオマーカー mRNAの測定値」と略記することもある)を取得するために、マイクロアレイ法、RNA-seq解析法、定量的RT-PCR法等公知の方法を使用することができる。マイクロアレイ法に使用するプローブは、自ら選択したプローブ又は公知のプローブを合成して使用してもよく、また市販のマイクロアレイチップを使用してもよい。
2-2. Obtaining a Measured Value of a Biomarker Gene In order to obtain a measured value of at least one mRNA selected from the group consisting of a biomarker (hereinafter sometimes abbreviated as "measured value of a biomarker mRNA" in the present specification) In addition, known methods such as microarray method, RNA-seq analysis method, quantitative RT-PCR method can be used. The probes used in the microarray method may be synthesized by using a self-selected probe or a known probe, or a commercially available microarray chip may be used.
 ここで、本方法においては、検体から抽出したtotal RNA及びmRNAのいずれを用いてもよい。total RNA及びmRNA抽出に用いる検体は、個体から採取されてすぐにRNA抽出に供されるか、個体から採取されてすぐに凍結(好ましくは、-196℃以下の雰囲気下(液体窒素中で急冷)して、RNA抽出まで-80℃以下で保存されることが好ましい。 Here, in the present method, either total RNA or mRNA extracted from the sample may be used. Samples used for total RNA and mRNA extraction are collected from an individual and immediately subjected to RNA extraction, or collected from an individual and frozen immediately (preferably under an atmosphere of -196 ° C. or less (quenched in liquid nitrogen ) And stored at -80 ° C. or less until RNA extraction.
 検体からのtotal RNA及びmRNAの抽出方法は特に制限されず、公知の抽出方法を使用することができる。 The method for extracting total RNA and mRNA from the sample is not particularly limited, and known extraction methods can be used.
 マイクロアレイ法による定量は、公知の方法に従って行うことができ、バイオマーカー mRNAの発現量は、ハウスキーピング遺伝子の発現量に対する相対発現量として表してもよく、蛍光色素等のシグナル強度の測定値として表すことができる。 The quantification by the microarray method can be performed according to a known method, and the expression amount of the biomarker mRNA may be expressed as a relative expression amount relative to the expression amount of a housekeeping gene, and is expressed as a measurement value of signal intensity of a fluorescent dye or the like be able to.
 RT-PCRによる定量は、検体から抽出したtotal RNA又はmRNAを鋳型として逆転写反応を行い、得られたcDNAを鋳型としてバイオマーカー mRNAの特異的なプライマーを使用してリアルタイムPCR法等で解析することにより行うことができる。また、この場合、バイオマーカー mRNAの発現量は、ハウスキーピング遺伝子の発現量に対する相対発現量として表してもよく、蛍光色素等のシグナルの強度の測定値として表してもよい。 For quantification by RT-PCR, reverse transcription is performed using total RNA or mRNA extracted from the sample as a template, and the obtained cDNA is used as a template for analysis by real-time PCR using specific primers for the biomarker mRNA. It can be done by Also, in this case, the expression level of the biomarker mRNA may be expressed as a relative expression level relative to the expression level of the housekeeping gene, or may be expressed as a measurement value of the intensity of a signal such as a fluorescent dye.
 また、RNA-seq解析法は、検体から抽出したmRNAを断片化し、これを鋳型として逆転写反応によるcDNAの合成とライブラリ作成を行う。各ライブラリに含まれる断片について次世代シークエンサーによる塩基配列を決定し、その情報をリファレンス遺伝子配列へマッピングし、mRNAの発現量をRPKM(Reads Per Killobases per Million)として表す。RPKMは、ヒートマップ等のシグナルの強度として表してもよい。 Also, in the RNA-seq analysis method, mRNA extracted from a sample is fragmented, and this is used as a template to synthesize cDNA by reverse transcription reaction and create a library. The nucleotide sequence of a fragment contained in each library is determined by a next-generation sequencer, the information is mapped to a reference gene sequence, and the amount of mRNA expression is represented as RPKM (Reads Per Killobases per Million). RPKM may be expressed as the intensity of a signal such as a heat map.
 上記シグナルの検出結果は、バイオマーカー mRNAの発現量として用いることができる。例えば、シグナルの強度を定量的に検出する場合は、シグナル強度の測定値自体又は該シグナル強度の測定値から算出される値を、バイオマーカー mRNAの発現量として用いることができる。 The detection result of the above signal can be used as the expression level of biomarker mRNA. For example, in the case of quantitatively detecting the intensity of a signal, the measured value of the signal intensity itself or a value calculated from the measured value of the signal intensity can be used as the expression level of the biomarker mRNA.
 上記シグナル強度の測定値から算出される値としては、例えば、該シグナル強度の測定値から陰性対照試料のシグナル強度の測定値を差し引いた値、該シグナル強度の測定値を陽性対照試料のシグナル強度の測定値で除した値、及びそれらの組み合わせなどが挙げられる。陰性対照試料としては、健常者の検体等が挙げられる。陽性対照試料としては、バイオマーカー mRNAを所定の発現量で含む検体が挙げられる。 As a value calculated from the measurement value of the signal intensity, for example, a value obtained by subtracting the measurement value of the signal intensity of the negative control sample from the measurement value of the signal intensity, the signal intensity of the positive control sample And the combination thereof. Examples of negative control samples include samples of healthy persons. The positive control sample includes a sample containing biomarker mRNA at a predetermined expression level.
 ここで、前記バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値は、各バイオマーカーの機能を反映する測定値として取得されてもよい。 Here, at least one mRNA selected from the group consisting of a measured value of at least one protein selected from the group consisting of the biomarkers, and / or a biomarker contained in a sample collected from the subject The measurement value of may be obtained as a measurement value reflecting the function of each biomarker.
3.ハードウェアの構成
 第1から第3の実施態様で使用されるハードウェアの構成を図1及び図2を用いて説明する。
3. Hardware Configuration The hardware configuration used in the first to third embodiments will be described using FIGS.
 図1は、本発明の第1~第3の実施態様に係るシステム100の概観図であり、図2は、システム100のハードウェア構成を示すブロック図である。システム100は、演算装置(以下では、同様のハードウェア構成を有し、後に説明するような異なる機能構成を有する演算装置に言及することになるため、これら異なる機能構成を有する演算装置を総称するために、「演算装置1,2,3」と表記する)と、入力部8と、表示部9と、測定装置5aと、測定装置5bと、を備える。 FIG. 1 is a schematic view of a system 100 according to first to third embodiments of the present invention, and FIG. 2 is a block diagram showing a hardware configuration of the system 100. The system 100 refers to arithmetic devices having the same hardware configuration and having different functional configurations as described later, and therefore, the system 100 generically refers to the arithmetic devices having these different functional configurations. For this purpose, it is provided with “ calculation devices 1, 2, 3”, the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
 演算装置1,2,3は、例えば汎用のパーソナルコンピュータで構成されており、後述するデータ処理を行うCPU101と、データ処理の作業領域に使用するメモリ102と、処理データを記録する記録部103と、各部の間でデータを伝送するバス104と、外部機器とのデータの入出力を行うインタフェース部105(以下、I/F部と記す)とを備えている。入力部8及び表示部9は、演算装置1,2,3に接続されており、入力部8は、キーボード等で構成され、表示部9は、液晶ディスプレイ等で構成されている。入力部8と表示部9とは、一体化されてタッチパネル付き表示装置として実現されてもよい。なお、演算装置1,2,3は一体の装置である必要はなく、CPU101、メモリ102、記録部103等が別所に配置され、これらがネットワークで接続されていてもよい。また、入力部8や表示部9を省略した操作者を必要としない装置であってもよい。 The arithmetic units 1, 2, and 3 are, for example, general-purpose personal computers, and include a CPU 101 that performs data processing to be described later, a memory 102 used for a data processing work area, and a recording unit 103 that records processing data. A bus 104 for transmitting data between the respective units, and an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device are provided. The input unit 8 and the display unit 9 are connected to the arithmetic devices 1, 2, 3, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like. The input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel. The arithmetic units 1, 2, and 3 do not have to be integrated units, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate | omitted the input part 8 or the display part 9 may be sufficient.
 また、演算装置1,2,3と、測定装置5aと、測定装置5bとについても、一カ所に配置される必要は必ずしもなく、別所に設けられた装置間をネットワークで通信可能に接続したシステムを構成でもよい。 In addition, the arithmetic devices 1, 2, 3, the measuring device 5a, and the measuring device 5b do not necessarily need to be disposed at one place, but a system in which devices provided at different places are communicably connected by a network. May be configured.
 以下の説明においては、特に断らない限り演算装置1,2,3が行う処理は、記録部103又はメモリ102に格納されたプログラムに基づいて、実際には演算装置1,2,3のCPU101が行う処理を意味する。CPU101はメモリ102を作業領域として必要なデータ(処理途中の中間データ等)を一時記憶し、記録部103に演算結果等の長期保存するデータを適宜記録する。 In the following description, unless otherwise stated, the processing performed by arithmetic units 1, 2, 3 is actually based on the program stored in recording unit 103 or memory 102, and CPU 101 of arithmetic units 1, 2, 3 actually Means the process to be performed. The CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
 測定装置5aは、タンパク質を測定するための装置であり、試料置き場51と、反応部52と、検出部53とを備える。試料置き場51にセットされた被験体から採取された検体は、反応部52に設置された抗体捕捉用抗バイオマーカー抗体が固相されたマイクロプレートに分注されインキュベーションされる。必要に応じて未反応の抗原を除去した後、検出抗体がマイクロプレートに分注され、インキュベーションされる。必要に応じて未反応の抗原を除去した後、検出用抗体を検出するための基質がマイクロプレートに分注され、マイクロプレートが検出部53に移動され、基質が反応して発生したシグナルが測定される。また、測定装置5aの別態様は、マイクロアレイ解析によるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロアレイチップ上に分注し、ハイブリダイゼーションを行い、洗浄した後、検出部53に移動させシグナルを検出する。 The measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53. The sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased. After removing the unreacted antigen, if necessary, the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done. Moreover, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
 さらに、測定装置5aの別態様は、RT-PCRによるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロチューブ内に分注し、続いて定量的PCR用試薬をマイクロチューブ内に分注する。反応部52でPCR反応を行いながら、検出部53でチューブ内のシグナルを検出する。 Furthermore, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
 測定装置5bは、mRNAを測定するための装置であり、配列解析部54を備える。RNA-Seq用の反応を行ったサンプルを配列解析部54にセットし、配列解析部54内で、塩基配列の解析をおこなう。 The measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54. The sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
 測定装置5a,5bは、有線又は無線によって演算装置1,2,3に接続されている。測定装置5aは、タンパク質の測定値をA/D変換して、デジタルデータとして演算装置1,2,3に送信する。同様に、測定装置5bは、mRNAの測定値をA/D変換して、デジタルデータとして演算装置1,2,3に送信する。これにより、演算装置1,2,3は、タンパク質の測定値及びmRNAの測定値を、演算処理可能なデジタルデータとして取得することができる。なお、腎疾患マーカーの測定値は、例えば医療機関(図示せず)からインターネットを介してデジタルデータとして送信される。これにより、演算装置1,2,3は、腎疾患マーカーの測定値を、デジタルデータとして取得することができる。 The measuring devices 5a and 5b are connected to the computing devices 1, 2 and 3 by wire or wireless. The measuring device 5a performs A / D conversion of the measured value of the protein, and transmits it as digital data to the computing devices 1, 2, 3. Similarly, the measuring device 5b A / D converts the measured value of mRNA, and transmits it as digital data to the computing devices 1, 2, 3. As a result, the computing devices 1, 2, 3 can acquire the measured value of the protein and the measured value of the mRNA as digital data that can be processed. The measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the computing devices 1, 2, 3 can acquire the measurement value of the kidney disease marker as digital data.
4.腎疾患の予測
4-1.概要
 第1の実施態様においては、上記「2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、被験体の腎疾患を予測する。
4. Predicting kidney disease
4-1. Overview In the first embodiment, the measurement value obtained by performing the method of “2. Method for obtaining each measurement value” is used to predict renal disease in a subject.
 より具体的には、本実施態様における被験体の腎疾患を予測する方法は、被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程と、前記取得工程で取得された測定値に基づいて、前記腎疾患を予測する予測工程とを含む。 More specifically, the method for predicting renal disease in a subject in this embodiment is a measurement value of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from the subject And / or an acquisition step of acquiring a measurement value of at least one mRNA selected from the group consisting of biomarkers contained in a specimen collected from the subject, and the measurement value acquired in the acquisition step And a prediction step of predicting the renal disease on the basis of the above.
 本実施態様は、具体的には、上記「2.各測定値の取得方法」によって取得された前記測定値を、測定値それぞれのバイオマーカーに対応する所定の基準値と比較し、該測定値が前記基準値範囲外である場合には、前記被験体が腎疾患であると決定することができる。 Specifically, in this embodiment, the measured value obtained by the above-mentioned “2. Method of obtaining each measured value” is compared with a predetermined reference value corresponding to each biomarker of the measured value, and the measured value is obtained. Is determined to be outside the reference value range, it can be determined that the subject has a renal disease.
 また、1群のバイオマーカーの測定値が基準値の範囲外である場合には、前記腎疾患に、FGF23が関与していると決定してもよい。また、2群のバイオマーカーの測定値が基準値の範囲内である場合、前記腎疾患に、FGF23が関与していると決定してもよい。より好ましくは、1群のバイオマーカーの測定値が基準値の範囲外であり、2群のバイオマーカーの測定値が基準値の範囲内である場合に、前記腎疾患に、FGF23が関与していると決定してもよい。 In addition, when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is involved in the renal disease. In addition, when the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the renal disease is related to FGF23. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, the renal disease involves FGF23. You may decide to
 前記工程は、後述するCPU101が行ってもよいが、検査者が行ってもよい。 The process may be performed by the CPU 101 described later, or may be performed by an examiner.
 さらに、前記腎疾患の予測方法において、腎疾患であると予測された場合には、腎疾患を改善するための治療(例えば、低リン食等の食事療法)を行う工程を、含めてもよい。 Furthermore, the method for predicting renal disease may include a step of performing treatment (eg, diet therapy such as a low phosphorus diet) to improve the renal disease when it is predicted to be a renal disease. .
4-2.腎機能を予測する装置
 第1の実施態様は、CPU101が後述するプログラムにしたがって下記の演算手段を実行することによって制御される、被験体の腎疾患を予測する装置を含む:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記腎疾患を予測する予測手段。
4-2. Device for Predicting Renal Function The first embodiment includes a device for predicting renal disease in a subject, which is controlled by the CPU 101 executing the following computing means according to a program described later:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the renal disease based on the measurement value acquired by the acquisition means.
 本実施態様では、上記装置として演算装置1を備えたシステム100(図1及び図2)によって、腎疾患の予測を行うことができる。 In this embodiment, prediction of renal disease can be performed by the system 100 (FIGS. 1 and 2) provided with the arithmetic device 1 as the above-mentioned device.
 図3は、第1の実施態様に係る演算装置1の機能を説明するためのブロック図である。演算装置1は、測定値取得部11と、基準値取得部12と、測定値比較部13と、予測部14とを備える。これらの機能ブロックは、本発明に係るプログラムを例えば演算装置1の記録部103又はメモリ102にインストールし、このプログラムをCPU101が実行することにより実現される。なお、特許請求の範囲に記載の取得手段、予測手段が、図3に示す測定値取得部11、予測部14にそれぞれ対応する。基準値取得部12と、測定値比較部13とは、任意の構成であり得る。 FIG. 3 is a block diagram for explaining the function of the arithmetic device 1 according to the first embodiment. The arithmetic device 1 includes a measurement value acquisition unit 11, a reference value acquisition unit 12, a measurement value comparison unit 13, and a prediction unit 14. These functional blocks are realized by, for example, installing the program according to the present invention in the recording unit 103 or the memory 102 of the arithmetic device 1 and executing the program by the CPU 101. The acquisition unit and the prediction unit described in the claims correspond to the measurement value acquisition unit 11 and the prediction unit 14 shown in FIG. 3, respectively. The reference value acquisition unit 12 and the measurement value comparison unit 13 may have any configuration.
 本実施態様では、バイオマーカータンパク質の測定値M11は、測定装置5aから演算装置1に取り込まれ、バイオマーカーmRNAの測定値M21は、測定装置5bから演算装置1に取り込まれる。バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22は、演算装置1の外部に記録されており、例えばインターネットを介して演算装置1に取り込まれる。 In this embodiment, the measured value M11 of the biomarker protein is taken into the arithmetic device 1 from the measuring device 5a, and the measured value M21 of the biomarker mRNA is taken into the arithmetic device 1 from the measuring device 5b. The reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are recorded outside the computing device 1, and are taken into the computing device 1 via the Internet, for example.
 なお、バイオマーカータンパク質の測定値M11、バイオマーカーmRNAの測定値M21は、ネットワークを介して医療機関(図示せず)から取り込まれてもよい。また、バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22は、演算装置1の記録部103又はメモリ102に予め記録されていてもよい。 The measured value M11 of the biomarker protein and the measured value M21 of the biomarker mRNA may be taken from a medical institution (not shown) via a network. Further, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA may be recorded in advance in the recording unit 103 or the memory 102 of the arithmetic device 1.
 また、測定値取得部11と、基準値取得部12と、測定値比較部13と、予測部14の各機能ブロックは、単一のCPUで実行されることは必ずしも必要なく、複数のCPUで分散して処理されてもよい。たとえば、測定値取得部11と、基準値取得部12と、測定値比較部13の機能は第1のコンピュータのCPUにより実行され、予測部14の機能は別の第2のコンピュータのCPUにより実行される、というような構成であってもよい。 In addition, each function block of the measurement value acquisition unit 11, the reference value acquisition unit 12, the measurement value comparison unit 13, and the prediction unit 14 does not necessarily need to be executed by a single CPU, and a plurality of CPUs It may be distributed and processed. For example, the functions of the measurement value acquisition unit 11, the reference value acquisition unit 12, and the measurement value comparison unit 13 are performed by the CPU of the first computer, and the function of the prediction unit 14 is performed by the CPU of another second computer It may be configured as such.
 また、演算装置1は、以下の図4で説明するステップS11~S17の処理を行うために、本発明に係るプログラムを、例えば実行形式(例えばプログラミング言語からコンパイラにより変換されて生成される)で記録部103に予め記録しており、演算装置1は、記録部103に記録したプログラムを使用して処理を行う。なお、上記プログラムは、CD-ROM等の、コンピュータ読み取り可能であって一時的でない有形の記録媒体109から、演算装置1にインストールしてもよいし、演算装置1をインターネット(図示せず)と接続し、インターネットを介してプログラムのプログラムコードをダウンロードしてもよい。 In addition, in order to perform the processes of steps S11 to S17 described in FIG. 4 below, the arithmetic device 1 executes the program according to the present invention in, for example, an executable format (for example, converted from a programming language by a compiler). The arithmetic unit 1 performs processing using the program recorded in the recording unit 103. The program may be installed in the arithmetic device 1 from a tangible computer readable non-temporary storage medium 109 such as a CD-ROM, or the arithmetic device 1 may be installed on the Internet (not shown). You may connect and download the program code of the program via the Internet.
 なお、本項で使用される用語の説明は、上記I.4-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 4-1. Use the description of.
4-3.演算装置の動作とプログラム
 第1の実施態様における腎疾患の予測方法は、第1の実施態様に係る演算装置1が、下記プログラムによって、本発明の下記の腎疾患の予測方法を実行してもよい。
4-3. Operation of Arithmetic Device and Program The kidney disease prediction method in the first embodiment is the same as the calculation device 1 according to the first embodiment in executing the following kidney disease prediction method of the present invention by the following program Good.
 図4は、本発明の第1の態様に係る演算装置1が動作を示すフローチャートである。なお、図3に示す測定値取得部11により図4のステップS11が、基準値取得部12により図4のステップS12が、測定値比較部13により図4のステップS13が、予測部14により図4に示すステップS15及びS16の処理がそれぞれ実行される。 FIG. 4 is a flowchart showing an operation of the arithmetic device 1 according to the first aspect of the present invention. 4 by the measured value acquisition unit 11 shown in FIG. 3, step S12 by FIG. 4 by the reference value acquisition unit 12, step S13 by FIG. 4 by the measured value comparison unit 13, and FIG. The processes of steps S15 and S16 shown in FIG.
 ステップS11では、初めに、検査者からのバイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、測定値取得部11は、前記測定値の取得を開始する。ステップS11は、特許請求の範囲に記載の取得処理に相当する。 In step S11, first, an input of start of acquisition of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started. The measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S11 corresponds to the acquisition process described in the claims.
 次に、ステップS12では、基準値取得部12は、検査者による入力部8からの測定値の取得開始の入力又は比較開始の入力に応じて、あるいは測定値取得部11による前記測定値の取得に応じて、バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22を取得する。 Next, in step S12, the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 or according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner. In accordance with, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
 次に、ステップS13では、測定値比較部13が、バイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21と、各バイオマーカーの測定値に対応するバイオマーカータンパク質の基準値R12、又はバイオマーカーmRNAの基準値R22とを比較する。 Next, in step S13, the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
 次に、ステップS14において、予測部14は、ステップS13で比較した結果が、各バイオマーカーの基準値の範囲内である場合には、被験者が腎疾患ではないと予測する(ステップS15)。また、予測部14は、ステップS13で比較した結果が、各バイオマーカーの基準値の範囲外である場合には、被験者が腎疾患であると予測する(ステップS16)。ステップS14、ステップS15、ステップS16は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S14, when the comparison result in step S13 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is not a renal disease (step S15). Further, when the comparison result in step S13 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is a renal disease (step S16). Steps S14, S15, and S16 correspond to the prediction processing described in the claims.
 得られた予測結果は、演算装置1の表示部9に表示されるか(ステップS17)、演算装置1内の記録部103に記録される。もしくは、インターネットを介して接続された、演算装置1の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained prediction result is displayed on the display unit 9 of the arithmetic device 1 (step S17) or recorded in the recording unit 103 in the arithmetic device 1. Alternatively, it may be displayed on a display unit of a computer terminal outside the arithmetic device 1 connected via the Internet, for example, in a medical institution.
 第1の実施態様に係る、腎疾患を予測するためのコンピュータプログラムは、演算装置1のCPU101に、前記ステップS11~S17を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記演算装置が前記プログラムを読み取り可能である限り制限されない。 The computer program for predicting kidney disease according to the first embodiment includes a program that causes the CPU 101 of the arithmetic unit 1 to execute the steps S11 to S17. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
 なお、本項で使用される用語の説明は、上記I.4-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 4-1. Use the description of.
5.リン代謝異常の予測
5-1.概要
 第2の実施態様においては、上記「2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、被験体におけるリン代謝異常を予測する。
5. Prediction of phosphorus metabolism disorder
5-1. Overview In the second embodiment, the measurement value obtained by performing the method of “2. Method for obtaining each measurement value” is used to predict abnormal phosphorus metabolism in a subject.
 より具体的には、本実施態様における被験体のリン代謝異常を予測する方法は、被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程と、前記取得工程で取得された測定値に基づいて、前記リン代謝異常を予測する予測工程とを含む。 More specifically, the method of predicting phosphorus metabolism abnormality in a subject in this embodiment comprises measuring at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from the subject Acquisition step of acquiring a measurement value of at least one mRNA selected from the group consisting of a value and / or a biomarker contained in a specimen collected from the subject, and the measurement value acquired in the acquisition step And a prediction step of predicting the phosphorus metabolism abnormality based on
 本実施態様は、具体的には、上記「2.各測定値の取得方法」によって取得された前記測定値を、測定値それぞれに対応する所定の基準値と比較し、該測定値が前記基準値範囲外である場合には、前記被験体がリン代謝異常であると決定することができる。 Specifically, the present embodiment compares the measured values obtained by the above-mentioned “2. Method for obtaining each measured value” with a predetermined reference value corresponding to each measured value, and the measured value is the reference. If out of the value range, the subject can be determined to be abnormal in phosphorus metabolism.
 また、1群のバイオマーカーの測定値が基準値の範囲外である場合には、前記リン代謝異常に、FGF23が関与していると決定してもよい。また、2群のバイオマーカーの測定値が基準値の範囲内である場合、前記リン代謝異常に、FGF23が関与していると決定してもよい。より好ましくは、1群のバイオマーカーの測定値が基準値の範囲外であり、2群のバイオマーカーの測定値が基準値の範囲内である場合に、前記リン代謝異常に、FGF23が関与していると決定してもよい。 In addition, when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is involved in the phosphorus metabolism abnormality. In addition, when the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the above-mentioned phosphorus metabolism disorder is related to FGF23. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, FGF23 is involved in the phosphorus metabolism abnormality. You may decide to
 前記工程は、後述するCPU101が行ってもよいが、検査者が行ってもよい。 The process may be performed by the CPU 101 described later, or may be performed by an examiner.
 さらに、前記リン代謝異常の予測方法において、リン代謝異常であると予測された場合には、リン代謝異常を改善するための治療(例えば、低リン食等の食事療法)を行う工程を、含めてもよい。 Furthermore, in the method for predicting phosphorus metabolism abnormality, when it is predicted that phosphorus metabolism abnormality is caused, a step of performing treatment (eg, diet therapy such as low phosphorus diet) for improving phosphorus metabolism abnormality is included. May be
5-2.リン代謝異常を予測する装置
 第2の実施態様は、CPU101が、後述するプログラムによって下記の演算機能を実行することによって制御される、被験体のリン代謝異常を予測する装置を含む:
 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記リン代謝異常を予測する予測手段。
5-2. Apparatus for predicting phosphorus metabolism abnormality The second embodiment includes an apparatus for predicting phosphorus metabolism abnormality of a subject, which is controlled by the CPU 101 by executing the following arithmetic function by a program described later:
From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the phosphorus metabolism abnormality based on the measurement value acquired by the acquisition means.
 本実施態様では、上記装置として演算装置1を備えたシステム100(図1及び図2)によって、リン代謝異常の予測を行うことができる。 In the present embodiment, it is possible to predict phosphorus metabolism abnormality by the system 100 (FIG. 1 and FIG. 2) provided with the arithmetic device 1 as the above-mentioned device.
 第2の実施態様に係る演算装置2の機能を説明するためのブロック図を図3に示す。演算装置2の構成は、上記4-2.で述べた演算装置1と同様である。 The block diagram for demonstrating the function of the calculating | arithmetic apparatus 2 which concerns on a 2nd embodiment is shown in FIG. The configuration of the arithmetic unit 2 is the same as the above 4-2. It is the same as the arithmetic unit 1 described above.
 なお、本項で使用される用語の説明は、上記I.5-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 5-1. Use the description of.
5-3.演算装置の動作とプログラム
 第2の実施態様におけるリン代謝異常の予測方法は、第2の実施態様に係る演算装置2が、下記プログラムによって、本発明の下記のリン代謝異常の予測方法を実行してもよい。
5-3. Operation of Arithmetic Device and Program The method for predicting phosphorus metabolism abnormality in the second embodiment is such that the arithmetic device 2 according to the second embodiment executes the following method for predicting phosphorus metabolism abnormality of the present invention by the following program May be
 図5は、本発明の第2の態様に係る演算装置2の動作を示すフローチャートである。なお、図3に示す測定値取得部11により図5のステップS21が、基準値取得部12により図5のステップS22が、測定値比較部13により図5のステップS23が、予測部14により図5に示すステップS25及びS26の処理がそれぞれ実行される。 FIG. 5 is a flowchart showing the operation of the arithmetic device 2 according to the second aspect of the present invention. The step S21 in FIG. 5 is performed by the measured value acquiring unit 11 shown in FIG. 3, the step S22 in FIG. 5 is performed by the reference value acquiring unit 12, the step S23 in FIG. The processes of steps S25 and S26 shown in FIG.
 ステップS21では、初めに、検査者からのバイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、測定値取得部11は、前記測定値の取得を開始する。ステップS21は、特許請求の範囲に記載の取得処理に相当する。 In step S21, first, an input of start of acquisition of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started. The measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S21 corresponds to the acquisition process described in the claims.
 次に、ステップS22では、基準値取得部12は、検査者による入力部8からの測定値の取得開始の入力又は比較開始の入力に応じて、あるいは測定値取得部11による前記測定値の取得に応じて、バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22を取得する。 Next, in step S22, the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner. In accordance with, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
 次に、ステップS23では、測定値比較部13が、バイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21と、各バイオマーカーの測定値に対応するバイオマーカータンパク質の基準値R12、又はバイオマーカーmRNAの基準値R22とを比較する。 Next, in step S23, the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
 次に、ステップS24において、予測部14は、ステップS23で比較した結果が、各バイオマーカーの基準値の範囲内である場合には、被験者がリン代謝異常ではないと予測する(ステップS25)。また、予測部14は、ステップS23で比較した結果が、各バイオマーカーの基準値の範囲外である場合には、被験者がリン代謝異常であると予測する(ステップS26)。ステップS24、ステップS25、ステップS26は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S24, when the comparison result in step S23 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is not abnormal in phosphorus metabolism (step S25). Further, when the comparison result in step S23 is outside the range of the reference value of each biomarker, the prediction unit 14 predicts that the subject is abnormal in phosphorus metabolism (step S26). Steps S24, S25, and S26 correspond to the prediction process described in the claims.
 得られた予測結果は、演算装置2の表示部9に表示されるか(ステップS27)、演算装置2内の記録部103に記録される。もしくは、インターネットを介して接続された、演算装置2の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained prediction result is displayed on the display unit 9 of the computing device 2 (step S 27) or recorded in the recording unit 103 in the computing device 2. Alternatively, it may be displayed on a display unit of a computer terminal outside the computing device 2 connected via the Internet, for example, in a medical institution.
 第2の実施態様に係る、リン代謝異常を予測するためのコンピュータプログラムは、演算装置2のCPU101に、前記ステップS21~S27を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記演算装置が前記プログラムを読み取り可能である限り制限されない。 A computer program for predicting phosphorus metabolism abnormality according to the second embodiment includes a program that causes the CPU 101 of the arithmetic unit 2 to execute the steps S21 to S27. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
 なお、本項で使用される用語の説明は、上記I.5-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 5-1. Use the description of.
6.FGF23の活性化の予測
6-1.概要
 第3の実施態様においては、上記「2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、被験体におけるFGF23の活性化を予測する。
6. Prediction of FGF23 activation
6-1. Overview In the third embodiment, the measurement values obtained by performing the method of “2. Acquisition method of each measurement value” are used to predict the activation of FGF23 in a subject.
 より具体的には、本実施態様における被験体のFGF23の活性化を予測する方法は、被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程と、前記取得工程で取得された測定値に基づいて、前記FGF23の活性化を予測する予測工程とを含む。 More specifically, the method of predicting FGF23 activation in a subject according to this embodiment is a method of at least one protein selected from the group consisting of biomarkers contained in a skin-derived sample collected from a subject An acquisition step of acquiring a measurement value of at least one type of mRNA selected from the group consisting of a measurement value and / or a biomarker included in a sample collected from the subject, and the measurement acquired in the acquisition step And V. predicting the activation of the FGF23 based on the value.
 本実施態様は、具体的には、上記「2.各測定値の取得方法」によって取得された前記測定値を、測定値それぞれに対応する所定の基準値と比較し、該測定値が前記基準値範囲外である場合には、前記被験体が腎疾患であると決定することができる。 Specifically, the present embodiment compares the measured values obtained by the above-mentioned “2. Method for obtaining each measured value” with a predetermined reference value corresponding to each measured value, and the measured value is the reference. If out of the value range, the subject can be determined to have renal disease.
 また、1群のバイオマーカーの測定値が基準値の範囲外である場合には、前記被験体においてFGF23の活性化していると決定してもよい。また、2群のバイオマーカーの測定値が基準値の範囲内である場合、前記被験体においてFGF23の活性化していると決定してもよい。より好ましくは、1群のバイオマーカーの測定値が基準値の範囲外であり、2群のバイオマーカーの測定値が基準値の範囲内である場合に、前記被験体においてFGF23の活性化していると決定してもよい。 In addition, when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that FGF23 is activated in the subject. In addition, when measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that FGF23 is activated in the subject. More preferably, when the measured value of one group of biomarkers is outside the range of the reference value and the measured value of two groups of biomarkers is within the range of the reference value, FGF23 is activated in the subject You may decide to
 前記工程は、後述するCPU101が行ってもよいが、検査者が行ってもよい。 The process may be performed by the CPU 101 described later, or may be performed by an examiner.
 さらに、前記FGF23の活性化の予測方法において、FGF23の活性化であると予測された場合には、FGF23の活性化を改善するための治療(例えば、抗FGF抗体の投与、高リン食等の食事療法)を行う工程を、含めてもよい。 Furthermore, in the method for predicting FGF23 activation, when it is predicted that FGF23 activation is to be achieved, a treatment for improving FGF23 activation (eg, administration of anti-FGF antibody, high phosphorus diet, etc.) Dietary treatment may be included.
6-2.FGF23の活性化を予測する装置
 第3の実施態様は、CPU101が、後述するプログラムによって下記の演算機能を実行することによって制御される、被験体のFGF23の活性化を予測する装置を含む: 前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
 前記取得手段が取得した測定値に基づいて、前記FGF23の活性化を予測する予測手段。
6-2. Device for predicting activation of FGF23 A third embodiment includes a device for predicting activation of FGF23 in a subject, which is controlled by the CPU 101 executing the following arithmetic function by a program described later: It consists of a measured value of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from subjects, and / or biomarkers contained in samples collected from said subjects Acquisition means for acquiring a measurement value of at least one type of mRNA selected from a group, and prediction means for predicting the activation of the FGF23 based on the measurement value acquired by the acquisition means.
 本実施態様では、上記装置として演算装置1を備えたシステム100(図1及び図2)によって、FGF23の活性化の予測を行うことができる。 In the present embodiment, it is possible to predict the activation of FGF 23 by the system 100 (FIGS. 1 and 2) provided with the arithmetic device 1 as the above-mentioned device.
 第3の実施態様に係る演算装置3の機能を説明するためのブロック図を図3に示す。演算装置3の構成は、上記4-2.で述べた演算装置1と同様である。 The block diagram for demonstrating the function of the arithmetic unit 3 which concerns on a 3rd embodiment is shown in FIG. The configuration of the arithmetic unit 3 is the same as the above 4-2. It is the same as the arithmetic unit 1 described above.
 なお、本項で使用される用語の説明は、上記I.6-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 6-1. Use the description of.
6-3.演算装置の動作とプログラム
 第3の実施態様におけるFGF23の活性化の予測方法は、第3の実施態様に係る演算装置3が、下記プログラムによって、本発明の下記のFGF23の活性化の予測方法を実行してもよい。
6-3. Operation of Arithmetic Device and Program The method of predicting FGF23 activation in the third embodiment is as follows: The arithmetic device 3 according to the third embodiment uses the following program to predict the following FGF23 activation method of the present invention It may be executed.
 図6は、本発明の第3の態様に係る演算装置3の動作を示すフローチャートである。なお、図3に示す測定値取得部11により図6のステップS31が、基準値取得部12により図6のステップS32が、測定値比較部13により図6のステップS33が、予測部14により図6に示すステップS35及びS36の処理がそれぞれ実行される。 FIG. 6 is a flowchart showing the operation of the arithmetic device 3 according to the third aspect of the present invention. 6 by the measured value acquisition unit 11 shown in FIG. 3, step S32 by FIG. 6 by the reference value acquisition unit 12, step S33 by FIG. 6 by the measured value comparison unit 13, and FIG. The processes of steps S35 and S36 shown in 6 are respectively executed.
 ステップS31では、初めに、検査者からのバイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、測定値取得部11は、前記測定値の取得を開始する。ステップS31は、特許請求の範囲に記載の取得処理に相当する。 In step S31, first, an acceptance start input of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started. The measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S31 corresponds to the acquisition process described in the claims.
 次に、ステップS32では、基準値取得部12は、検査者による入力部8からの測定値の取得開始の入力又は比較開始の入力に応じて、あるいは測定値取得部11による前記測定値の取得に応じて、バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22を取得する。 Next, in step S <b> 32, the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 or according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner. In accordance with, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
 次に、ステップS33では、測定値比較部13が、バイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21と、各バイオマーカーの測定値に対応するバイオマーカータンパク質の基準値R12、又はバイオマーカーmRNAの基準値R22とを比較する。 Next, in step S33, the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or the bio The marker mRNA is compared with the reference value R22.
 次に、ステップS34において、予測部14は、ステップS33で比較した結果が、各バイオマーカーの基準値の範囲内である場合には、被験者においてFGF23が活性化していないと予測する(ステップS35)。また、予測部14は、ステップS33で比較した結果が、各バイオマーカーの基準値の範囲外である場合には、被験者においてFGF23が活性化していると予測する(ステップS36)。ステップS34、ステップS35、ステップS36は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S34, when the comparison result in step S33 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is not activated in the subject (step S35). . Further, when the comparison result in step S33 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is activated in the subject (step S36). Steps S34, S35, and S36 correspond to the prediction process described in the claims.
 得られた予測結果は、演算装置3の表示部9に表示されるか(ステップS37)、演算装置3内の記録部103に記録される。もしくは、インターネットを介して接続された、演算装置3の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained prediction result is displayed on the display unit 9 of the arithmetic device 3 (step S37) or recorded in the recording unit 103 in the arithmetic device 3. Alternatively, it may be displayed on a display unit of a computer terminal outside the arithmetic device 3 connected via the Internet, for example, in a medical institution.
 第3の実施態様に係る、FGF23の活性化を予測するためのコンピュータプログラムは、演算装置3のCPU101に、前記ステップS31~S37を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記演算装置が前記プログラムを読み取り可能である限り制限されない。 The computer program for predicting the activation of FGF23 according to the third embodiment includes a program that causes the CPU 101 of the arithmetic unit 3 to execute the steps S31 to S37. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
 なお、本項で使用される用語の説明は、上記I.6-1.の説明を援用する。 For the explanation of terms used in this section, the above-mentioned I. 6-1. Use the description of.
II.第4及び第5の実施態様
1.用語の説明
 第4及び第5の実施態様の説明において、使用する用語について説明する。
II. Fourth and fifth embodiments
1. Description of Terms In the description of the fourth and fifth embodiments, the terms used will be described.
 「被験体」は、被験物質を投与する対象となる個体であり、好ましくはヒトが除かれる。ヒト以外の哺乳動物としては、例えばウシ、ウマ、ヒツジ、ヤギ、ブタ、イヌ、ネコ、ウサギ、サル等が挙げられる。好ましくは、ヒト、ネコ、イヌである。また、個体の年齢、性別は問わない。 A "subject" is an individual to whom a test substance is administered, preferably a human is excluded. Examples of mammals other than human include cows, horses, sheep, goats, pigs, dogs, cats, rabbits, monkeys and the like. Preferred are humans, cats and dogs. Also, the age and gender of the individual do not matter.
 被験体は、腎機能低下やその他の腎疾患の既往を有している個体であっても、有していない個体であってもよい。また、FGF23が関連する疾患を患っている被験体でも、患っていない被験体であってもよい。 The subject may or may not be an individual having a history of impaired renal function or other renal disease. Also, the subject may or may not be a subject suffering from a disease associated with FGF23.
 本実施態様において、検体には、皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つが含まれる。また、前記皮膚から採取された検体は、生体の皮膚に由来する限り制限されない。また、皮膚由来検体には、汗、皮膚からの分泌液等が含まれる。皮膚の採取方法も制限されず、生検材料、ピアスの穴を空けた際に穿孔針に付着する皮膚、皮膚表面の擦過等を上げることができる。 In this embodiment, the sample is at least one selected from the group consisting of a sample collected from the skin, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin included. Further, the sample collected from the skin is not limited as long as it originates from the skin of a living body. The skin-derived sample also includes sweat, secretions from the skin, and the like. The method of collecting the skin is also not limited, and it is possible to increase the biopsy material, the skin adhering to the piercing needle when the piercing hole is made, the abrasion of the skin surface, and the like.
 さらに、検体は、新鮮なものであっても、保存されていたものであってもよい。検体を保存する場合には、室温環境、冷蔵環境又は冷凍環境において保存することができるが、好ましくは冷凍保存である。 Furthermore, the sample may be fresh or stored. When the sample is stored, it can be stored in a room temperature environment, a refrigerated environment or a frozen environment, but is preferably frozen.
 本実施態様において、「被験組織」とは、被験物質を投与する対象となる組織であり、例えば、前記被験体となりうる個体から採取され、体外で培養される等して体外において生存している組織である。当該組織は、一器官全体であってもよく、一器官の一部であってもよい。 In the present embodiment, the “test tissue” is a tissue to which a test substance is to be administered, for example, it is collected from an individual who can be the subject, and survives outside the body by being cultured outside the body. It is an organization. The tissue may be an entire organ or a part of an organ.
 本実施態様において、「被験細胞」とは、被験物質を投与する対象となる細胞であり、例えば、前記被験体となりうる個体から採取され、体外で培養される等して体外において生存している細胞である。当該細胞は、初代培養等の継代性が限られている細胞であってもよく、継代性が維持されている、いわゆる培養細胞であってもよい。また、これらは、遺伝子操作によって作成された細胞であってもよい。 In the present embodiment, the “test cell” is a cell to which a test substance is to be administered, for example, it is collected from an individual who can be the subject, and survives outside the body by being cultured in vitro. It is a cell. The cell may be a cell having limited passage such as primary culture, or may be a so-called cultured cell whose passage is maintained. Also, these may be cells produced by genetic engineering.
 本明細書において、「被験物質」とは、有効成分の候補物質であるか否かを評価する対象となりうる物質であり、特に制限されない。例えば、化合物、タンパク質、ペプチド、核酸、脂質、糖質、糖脂質、糖タンパク、金属等を挙げることができる。被験物質の投与方法も特に制限されない。被験細胞や被験組織に被験物質を投与する場合には、例えばその培養培地に1pg/mlから1mg/mlとなるように被験物質を投与することができる。また、被験体個体の場合には、1日あたり1ng/kg~1g/kgとなるように被験物質を投与することができる。被験物質の投与から、検体の採取までの期間は、当該被験物質の効果が得られる限り、特に制限されない。 In the present specification, the “test substance” is a substance that can be a subject to be evaluated as to whether it is a candidate substance of the active ingredient and is not particularly limited. For example, compounds, proteins, peptides, nucleic acids, lipids, carbohydrates, glycolipids, glycoproteins, metals and the like can be mentioned. The administration method of the test substance is also not particularly limited. When a test substance is administered to a test cell or a test tissue, the test substance can be administered, for example, at 1 pg / ml to 1 mg / ml in the culture medium. In addition, in the case of a subject individual, the test substance can be administered at 1 ng / kg to 1 g / kg per day. The period from the administration of the test substance to the collection of the sample is not particularly limited as long as the effect of the test substance can be obtained.
 さらに、被験物質で処理された被験体、被験組織又は被験細胞から採取された検体は、本明細書において、「被験物質処理検体」と呼ばれることもある。また、被験物質で処理されていない被験体、被験組織又は被験細胞から採取された検体は、本明細書において、「未処理検体」と呼ばれることもある。 Furthermore, a subject that has been treated with a test substance, a test tissue or a sample collected from test cells may be referred to herein as a "test substance-treated sample". In addition, a subject that has not been treated with a test substance, a subject collected from a subject tissue or a subject cell may be referred to herein as a "naive subject".
 本実施態様において、「腎疾患」、「FGF23が関連する疾患」、「バイオマーカー」、「バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値」、「バイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値」、「所定の基準値」、「健常個体」、「複数の検体」、「複数の値」、「抗バイオマーカー抗体」、「バイオマーカー mRNA検出核酸」の説明は、前記I.1.の用語の説明をここに援用する。また、「バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値」、「バイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値」、の取得方法は、上記I.2.に準ずる。 In this embodiment, “kidney disease”, “fGF23 related disease”, “biomarker”, “measured value of at least one protein selected from the group consisting of biomarkers”, “selected from the group consisting of biomarkers” Value of at least one type of mRNA, “predetermined reference value”, “healthy individual”, “multiple samples”, “multiple values”, “anti-biomarker antibody”, “biomarker mRNA detection nucleic acid” The explanation is given in the above I.I. 1. A description of the terms of is incorporated herein. In addition, methods for obtaining “a measured value of at least one type of protein selected from the group consisting of biomarkers” and “a measured value of at least one type of mRNA selected from the group consisting of biomarkers” are described above in I. 2. It conforms to.
2.ハードウェアの構成
 第4及び第5の実施態様で使用されるハードウェアの構成を図7及び図8を用いて説明する。
2. Hardware Configuration The hardware configuration used in the fourth and fifth embodiments will be described with reference to FIGS. 7 and 8. FIG.
 図7は、本発明の第4及び第5の実施態様に係るシステム200の概観図であり、図8は、システム200のハードウェア構成を示すブロック図である。システム200は、スクリーニング装置(以下では、同様のハードウェア構成を有し、後に説明するような異なる機能構成を有するスクリーニング装置に言及することになるため、これら異なる機能構成を有するスクリーニング装置を総称するために、「スクリーニング装置4,5」と表記する)と、入力部8と、表示部9と、測定装置5aと、測定装置5bと、を備える。 FIG. 7 is a schematic view of a system 200 according to fourth and fifth embodiments of the present invention, and FIG. 8 is a block diagram showing a hardware configuration of the system 200. The system 200 refers to a screening apparatus (in the following, having the same hardware configuration and referring to screening apparatuses having different functional configurations as will be described later). For this purpose, the " screening devices 4 and 5" are provided, the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
 スクリーニング装置4,5は、例えば汎用のパーソナルコンピュータで構成されており、後述するデータ処理を行うCPU101と、データ処理の作業領域に使用するメモリ102と、処理データを記録する記録部103と、各部の間でデータを伝送するバス104と、外部機器とのデータの入出力を行うインタフェース部105(以下、I/F部と記す)とを備えている。入力部8及び表示部9は、スクリーニング装置4,5に接続されており、入力部8は、キーボード等で構成され、表示部9は、液晶ディスプレイ等で構成されている。入力部8と表示部9とは、一体化されてタッチパネル付き表示装置として実現されてもよい。なお、スクリーニング装置4,5は一体の装置である必要はなく、CPU101、メモリ102、記録部103等が別所に配置され、これらがネットワークで接続されていてもよい。また、入力部8や表示部9を省略した操作者を必要としない装置であってもよい。 The screening devices 4 and 5 are, for example, general-purpose personal computers, and include a CPU 101 that performs data processing to be described later, a memory 102 used for a data processing work area, a recording unit 103 that records processing data, And an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device. The input unit 8 and the display unit 9 are connected to the screening devices 4 and 5, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like. The input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel. Note that the screening devices 4 and 5 do not have to be integrated devices, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate | omitted the input part 8 or the display part 9 may be sufficient.
 また、スクリーニング装置4,5と、測定装置5aと、測定装置5bとについても、一カ所に配置される必要は必ずしもなく、別所に設けられた装置間をネットワークで通信可能に接続したシステムを構成でもよい。 Also, the screening devices 4 and 5, the measuring device 5a, and the measuring device 5b do not necessarily have to be disposed at one place, and a system in which devices provided at different places are communicably connected via a network is configured. May be.
 以下の説明においては、特に断らない限りスクリーニング装置4,5が行う処理は、記録部103又はメモリ102に格納されたプログラムに基づいて、実際にはスクリーニング装置4,5のCPU101が行う処理を意味する。CPU101はメモリ102を作業領域として必要なデータ(処理途中の中間データ等)を一時記憶し、記録部103に演算結果等の長期保存するデータを適宜記録する。 In the following description, unless otherwise stated, the processing performed by the screening devices 4 and 5 actually means the processing performed by the CPU 101 of the screening devices 4 and 5 based on the program stored in the recording unit 103 or the memory 102. Do. The CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
 測定装置5aは、タンパク質を測定するための装置であり、試料置き場51と、反応部52と、検出部53とを備える。試料置き場51にセットされた被験体から採取された検体は、反応部52に設置された抗体捕捉用抗バイオマーカー抗体が固相されたマイクロプレートに分注されインキュベーションされる。必要に応じて未反応の抗原を除去した後、検出抗体がマイクロプレートに分注され、インキュベーションされる。必要に応じて未反応の抗原を除去した後、検出用抗体を検出するための基質がマイクロプレートに分注され、マイクロプレートが検出部53に移動され、基質が反応して発生したシグナルが測定される。また、測定装置5aの別態様は、マイクロアレイ解析によるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロアレイチップ上に分注し、ハイブリダイゼーションを行い、洗浄した後、検出部53に移動させシグナルを検出する。 The measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53. The sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased. After removing the unreacted antigen, if necessary, the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done. Moreover, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
 さらに、測定装置5aの別態様は、RT-PCRによるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロチューブ内に分注し、続いて定量的PCR用試薬をマイクロチューブ内に分注する。反応部52でPCR反応を行いながら、検出部53でチューブ内のシグナルを検出する。 Furthermore, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
 測定装置5bは、mRNAを測定するための装置であり、配列解析部54を備える。RNA-Seq用の反応を行ったサンプルを配列解析部54にセットし、配列解析部54内で、塩基配列の解析をおこなう。 The measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54. The sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
 測定装置5a,5bは、有線又は無線によってスクリーニング装置4,5に接続されている。測定装置5aは、タンパク質の測定値をA/D変換して、デジタルデータとしてスクリーニング装置4,5に送信する。同様に、測定装置5bは、mRNAの測定値をA/D変換して、デジタルデータとしてスクリーニング装置4,5に送信する。これにより、スクリーニング装置4,5は、タンパク質の測定値及びmRNAの測定値を、演算処理可能なデジタルデータとして取得することができる。なお、腎疾患マーカーの測定値は、例えば医療機関(図示せず)からインターネットを介してデジタルデータとして送信される。これにより、スクリーニング装置4,5は、腎疾患マーカーの測定値を、デジタルデータとして取得することができる。 The measuring devices 5a and 5b are connected to the screening devices 4 and 5 by wire or wirelessly. The measuring device 5a performs A / D conversion of the measured value of the protein, and transmits it to the screening device 4, 5 as digital data. Similarly, the measuring device 5b converts the measured value of mRNA into a digital signal, and transmits it to the screening devices 4 and 5 as digital data. Thereby, the screening devices 4 and 5 can acquire the measurement value of the protein and the measurement value of the mRNA as digital data that can be processed. The measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the screening devices 4 and 5 can acquire the measurement value of the kidney disease marker as digital data.
3.FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング
3-1.概要
 第4の実施態様においては、上記「I.2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングを行う。
3. Screening of candidate active ingredients for suppressing functional expression of FGF23
3-1. Overview In the fourth embodiment, an active ingredient for suppressing the functional expression of FGF23 using the measurement values obtained by performing the method of “I. 2. Method for obtaining each measurement value” described above is used. Screening of candidate substances.
 より具体的には、本実施態様における被験体のFGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングする方法は、被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する取得工程と、第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程とを含む。 More specifically, the method of screening for a candidate substance of the active ingredient for suppressing the functional expression of FGF23 in the subject in the present embodiment is the method of collecting the substance from the skin of the subject (except human) treated with the test substance Of the biomarker protein of at least one test substance-treated sample selected from the group consisting of a tested sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin and And / or acquiring the measurement value of mRNA of the protein, and determining the test substance as a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquiring means. .
 本実施態様は、好ましくは、上記「I.2.各測定値の取得方法」によって、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程を含む。また、前記被験物質処理検体から取得された各バイオマーカーの測定値と、それぞれに対応する未検体処理中の測定値とを比較し、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が変化していれば、前記被験物質がFGF23の機能発現を抑制する有効成分であると決定することができる。 In this embodiment, preferably, a test sample obtained from the skin of a subject (except for human beings) not treated with the test substance, a test derived from the skin according to the above-mentioned "I. Measurement value of corresponding biomarker protein and / or mRNA of said protein in at least one unprocessed sample selected from the group consisting of a sample collected from a tissue and a sample collected from a test cell derived from skin Including the step of obtaining a measurement value. In addition, the measured value of each biomarker obtained from the test substance-treated sample is compared with the corresponding measured value in the unsampled process, and compared with the measured value of the biomarker of the untreated sample to be treated with the test substance If the measured value of the biomarker of the sample has changed, it can be determined that the test substance is an active ingredient that suppresses the functional expression of FGF23.
 ここで、FGF23の機能発現とは、FGF23の本来の機能が発揮されることをいう。FGF23の機能発現の例は、例えば、血中の無機リン濃度を下げることである。また、FGF23の機能発現には、FGF23の発現が上昇することを含む。 Here, the functional expression of FGF23 means that the original function of FGF23 is exhibited. An example of the functional expression of FGF23 is, for example, lowering the concentration of inorganic phosphorus in blood. Moreover, functional expression of FGF23 includes an increase in expression of FGF23.
 被験物質処理検体のバイオマーカーの測定値の変化が、正方向であるか負方向であるかは、バイオマーカーに依存する。 Whether the change in the measured value of the biomarker of the test substance-treated sample is positive or negative depends on the biomarker.
 例えば、上記I.1.で述べたバイオマーカーのうち、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、及びLrrc2よりなる群より選択される少なくとも一種(3群という)は、高リン食を摂取したマウスと比較してFGF23のノックアウトマウスで発現が上昇する。また、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種(4群という)は、高リン食を摂取したマウスと比較してFGF23のノックアウトマウスで発現が減少する。さらに、Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種(上述の2群)は、FGF23のノックアウトマウスにおいても発現が変化しない。 For example, above I. 1. Among the biomarkers mentioned above, at least one selected from the group consisting of Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1 and Lrrc2 (referred to as the third group) Is up-regulated in FGF23 knockout mice as compared to mice receiving a high phosphorus diet. In addition, at least one type (group 4) selected from the group consisting of Col15a1, Sparc, Col11a1, Clec11a, Serpinb6d, and Defb8 has a decreased expression in FGF23 knockout mice as compared to mice receiving a high phosphorus diet. . Furthermore, at least one type (the above-mentioned two groups) selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 does not change the expression in FGF23 knockout mice.
 したがって、3群のバイオマーカーについては、例えば、高リン食を摂取したマウスを被験体として、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が上昇していれば、当該被験物質は、FGF23の機能発現を抑制すると決定することができる。また、4群のバイオマーカーについては、高リン食を摂取したマウスを被験体として、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が低下していれば、当該被験物質は、FGF23の機能発現を抑制すると決定することができる。さらに、これらのバイオマーカーの検討に加え、2群のバイオマーカーについても同様に検討した場合に、未処理検体のバイオマーカー測定値と被験物質処理検体のバイオマーカーの測定値に差がなければ、被験物質はFGF23の機能に特異的に作用していると決定することができる。 Therefore, for the three groups of biomarkers, for example, with the mouse receiving a high phosphorus diet as a subject, the measured value of the biomarker of the test substance-treated sample is increased compared to the measured value of the biomarker of the untreated sample. Then, the test substance can be determined to suppress the functional expression of FGF23. Also, for the 4 groups of biomarkers, if the mouse receiving a high phosphorus diet is the subject, the measured value of the biomarker of the test substance-treated sample is lower than the measured value of the biomarker of the untreated sample. The test substance can be determined to suppress the functional expression of FGF23. Furthermore, in addition to the examination of these biomarkers, when the two groups of biomarkers are similarly examined, if there is no difference between the measured values of the biomarkers of the untreated sample and the measured values of the biomarkers of the test substance treated sample, The test substance can be determined to act specifically on the function of FGF23.
 被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が、未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値よりも上昇しているか否かは、例えば、被験物質処理検体の測定値が未処理検体の測定値の例えば115%以上、好ましくは130%以上、より好ましくは150%以上になっている場合に、当該被験物質により、バイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が上昇していると決定することがでる。 The measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of the protein is higher than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of the protein For example, when the measured value of the test substance-treated sample is, for example, 115% or more, preferably 130% or more, more preferably 150% or more of the measured value of the untreated sample, the test substance Thus, it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is rising.
 被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が、未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値よりも低下しているか否かは、例えば、被験物質処理検体の測定値が未処理検体の測定値の例えば85%以下、好ましくは70%以下、より好ましくは50%以下になっている場合に、当該被験物質により、バイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が低下していると決定することがでる。 The measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of said protein is lower than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of said protein For example, when the measured value of the test substance-treated sample is, for example, 85% or less, preferably 70% or less, more preferably 50% or less of the measured value of the untreated sample, the test substance Thus, it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is decreasing.
 さらに、スクリーニングは、前記工程(I)の前に、(i)被験体、被験組織又は被験細胞に、被験物質を投与する投与工程、(ii)前記工程(i)で被験物質を投与された被験体、被験組織又は被験細胞から検体を採取する工程、及び(iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程を含んでいてもよい。この場合にも、工程(ii)と工程(iii)は、必ずしも同一機関において連続して行われる必要はなく、例えば、工程(ii)で採取された検体を、第三者機関に送り工程(iii)以降を実施してもよい。また、工程(iii)と工程(I)も、必ずしも同一機関において連続して行われる必要はなく、例えば、工程(iii)で回収されたタンパク質及び/又はmRNAを、第三者機関に送り工程(iii)以降を実施してもよい。 Furthermore, in the screening, before the step (I), the test substance is administered in the administration step of (i) administering the test substance to the subject, the test tissue or the test cell, and (ii) the step (i) The method may include the steps of collecting the sample from the subject, the test tissue or the test cells, and (iii) recovering the protein and / or the mRNA from the sample obtained in the step (ii). Also in this case, the step (ii) and the step (iii) do not necessarily need to be performed continuously in the same organization, and for example, the sample collected in the step (ii) is sent to a third party organization iii) The following may be implemented. In addition, step (iii) and step (I) do not necessarily have to be performed continuously in the same organization, for example, the step of sending the protein and / or mRNA recovered in step (iii) to a third party organization (Iii) The following may be implemented.
3-2.スクリーニング装置
 第4の実施態様は、CPU101が、後述するプログラムによって下記の演算機能を実行することによって制御される、下記の演算手段を有する、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング装置4を含む:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、並びに
 前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
3-2. Screening Device A fourth embodiment is a candidate substance of an active ingredient for suppressing the functional expression of FGF23 having the following arithmetic means, which is controlled by the CPU 101 executing the following arithmetic function according to a program described later The screening device 4 includes:
Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Means for acquiring the measurement value of the biomarker protein of at least one test substance-treated sample and / or the measurement value of mRNA of the protein, and the measurement acquired by the first measurement value acquisition means A determining means for determining that the test substance is a candidate substance of the active ingredient based on a value.
 本実施態様では、上記装置としてスクリーニング装置4を備えたシステム200(図7及び図8)によって、スクリーニングを行うことができる。 In this embodiment, screening can be performed by the system 200 (FIGS. 7 and 8) provided with the screening device 4 as the above-mentioned device.
 図9は、本実施態様に係るスクリーニング装置4の機能を説明するためのブロック図である。スクリーニング装置4は、第1の測定値取得部31と、第2の測定値取得部32と、測定値比較部33と、候補物質決定部34とを備える。第2の測定値取得部32は任意の構成である。これらの機能ブロックは、本発明に係るスクリーニングプログラムを、図8に示すスクリーニング装置4の記録部103又はメモリ102にインストールし、このスクリーニングプログラムをCPU101が実行することにより実現される。特許請求の範囲に記載の第1の測定値取得手段、第2の測定値取得手段、測定値比較手段及び決定手段が、図9に示す第1の測定値取得部31、第2の測定値取得部32、測定値比較部33及び候補物質決定部34にそれぞれ対応する。 FIG. 9 is a block diagram for explaining the function of the screening device 4 according to this embodiment. The screening device 4 includes a first measurement value acquisition unit 31, a second measurement value acquisition unit 32, a measurement value comparison unit 33, and a candidate substance determination unit 34. The second measurement value acquisition unit 32 is an arbitrary configuration. These functional blocks are realized by installing the screening program according to the present invention in the recording unit 103 or the memory 102 of the screening device 4 shown in FIG. 8 and the CPU 101 executing this screening program. The first measured value acquiring unit, the second measured value acquiring unit, the measured value comparing unit, and the determining unit according to the claims are the first measured value acquiring unit 31 and the second measured value illustrated in FIG. 9. The acquisition unit 32, the measurement value comparison unit 33, and the candidate substance determination unit 34 correspond to each other.
 本実施態様では、被験物質処理検体のバイオマーカータンパク質の測定値M31は、測定装置5aからスクリーニング装置4に取り込まれ、被験物質処理検体のバイオマーカーmRNAの測定値M41は、測定装置5bからスクリーニング装置4に取り込まれる。未処理検体のバイオマーカータンパク質の測定値M32、未処理検体のバイオマーカーmRNAの測定値M42は、スクリーニング装置4の外部に記録されており、例えばインターネットを介してスクリーニング装置4に取り込まれる。 In this embodiment, the measured value M31 of the biomarker protein of the test substance-treated sample is taken from the measuring device 5a to the screening device 4, and the measured value M41 of the biomarker mRNA of the test substance-treated sample is measured from the measuring device 5b to the screening device Captured in 4. The measured value M32 of the biomarker protein of the untreated sample and the measured value M42 of the biomarker mRNA of the untreated sample are recorded outside the screening device 4, and are taken into the screening device 4 via the Internet, for example.
 なお、被験物質処理検体のバイオマーカータンパク質の測定値M31、被験物質処理検体のバイオマーカーmRNAの測定値M41は、ネットワークを介して医療機関(図示せず)から取り込まれてもよい。また、未処理検体のバイオマーカータンパク質の測定値M32、未処理検体のバイオマーカーmRNAの測定値M42は、クリーニング装置4の記録部103又はメモリ102に予め記録されていてもよい。 The measured value M31 of the biomarker protein of the test substance-treated sample and the measured value M41 of the biomarker mRNA of the test substance-treated sample may be taken from a medical institution (not shown) via a network. In addition, the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample may be recorded in advance in the recording unit 103 or the memory 102 of the cleaning device 4.
 また、第1の測定値取得部31と、第2の測定値取得部32と、測定値比較部33と、候補物質決定部34の各機能ブロックは、単一のCPUで実行されることは必ずしも必要なく、複数のCPUで分散して処理されてもよい。たとえば、第1の測定値取得部31と、第2の測定値取得部32と、測定値比較部33と、の機能は第1のコンピュータのCPUにより実行され、候補物質決定部34の機能は別の第2のコンピュータのCPUにより実行される、というような構成であってもよい。 In addition, each functional block of the first measurement value acquisition unit 31, the second measurement value acquisition unit 32, the measurement value comparison unit 33, and the candidate substance determination unit 34 may be executed by a single CPU. It is not necessarily necessary, and processing may be distributed and performed by a plurality of CPUs. For example, the functions of the first measurement value acquisition unit 31, the second measurement value acquisition unit 32, and the measurement value comparison unit 33 are executed by the CPU of the first computer, and the function of the candidate substance determination unit 34 is It may be configured to be executed by the CPU of another second computer.
 また、スクリーニング装置4は、以下の図10で説明するステップS41~S47の処理を行うために、本発明に係るプログラムを、例えば実行形式(例えばプログラミング言語からコンパイラにより変換されて生成される)で記録部103に予め記録しており、スクリーニング装置4は、記録部103に記録したプログラムを使用して処理を行う。なお、上記プログラムは、CD-ROM等の、コンピュータ読み取り可能であって一時的でない有形の記録媒体109から、スクリーニング装置4にインストールしてもよいし、スクリーニング装置4をインターネット(図示せず)と接続し、インターネットを介してプログラムのプログラムコードをダウンロードしてもよい。 In addition, the screening device 4 executes the program according to the present invention in, for example, an execution form (for example, converted from a programming language by a compiler and generated) in order to perform the processes of steps S41 to S47 described in FIG. The screening device 4 performs the process using the program recorded in the recording unit 103. The above program may be installed in the screening device 4 from a tangible computer readable non-temporary recording medium 109 such as a CD-ROM, or the screening device 4 may be connected to the Internet (not shown). You may connect and download the program code of the program via the Internet.
3-3.スクリーニング装置の動作とプログラム
 第4の実施態様におけるFGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングは、第4の実施態様に係るスクリーニング装置4が、下記プログラムによって、本発明の下記のFGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングを実行してもよい。
3-3. Operation of Screening Device and Program In the fourth embodiment, the screening of candidate substances for the active ingredient for suppressing the functional expression of FGF23 is carried out by the screening device 4 according to the fourth embodiment of Screening of candidate active ingredients for suppressing functional expression of FGF23 may be performed.
 図10は、本発明の第4の態様に係るスクリーニング装置4の動作を示すフローチャートである。なお、図9に示す第1の測定値取得部11により図10のステップS41が、第2の測定値取得部32により図10のステップS42が、測定値比較部33により図10のステップS43が、候補物質決定部34により図10に示すステップS45及びS46の処理がそれぞれ実行される。 FIG. 10 is a flowchart showing the operation of the screening device 4 according to the fourth aspect of the present invention. 10 is performed by the first measurement value acquisition unit 11 shown in FIG. 9, step S42 of FIG. 10 is performed by the second measurement value acquisition unit 32, and step S43 of FIG. 10 is performed by the measurement value comparison unit 33. The candidate substance determining unit 34 executes the processing of steps S45 and S46 shown in FIG.
 ステップS41では、初めに、検査者からの被験物質処理検体のバイオマーカータンパク質の測定値M31又は被験物質処理検体のバイオマーカーmRNAの測定値M41の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、第1の測定値取得部31は、前記測定値の取得を開始する。ステップS41は、特許請求の範囲に記載の取得処理に相当する。 In step S41, first, an input for starting acquisition of the measured value M31 of the biomarker protein of the test substance-treated sample or the measured value M41 of the biomarker mRNA of the test substance-treated sample from the examiner is received from the input unit 8 Alternatively, upon receiving a measurement start instruction from the measurement device 5a or 5b, the first measurement value acquisition unit 31 starts acquisition of the measurement value. Step S41 corresponds to the acquisition process described in the claims.
 次に、ステップS42では、第2の測定値取得部32は、未処理検体のバイオマーカータンパク質の測定値M32、未処理検体のバイオマーカーmRNAの測定値M42を取得する。 Next, in step S42, the second measurement value acquisition unit 32 acquires the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample.
 次に、ステップS43では、測定値比較部33が、被験物質処理検体のバイオマーカータンパク質の測定値M31とそれぞれに対応する未処理検体のバイオマーカータンパク質の測定値M32とを比較し、被験物質処理検体のバイオマーカーmRNAの測定値M41とそれぞれに対応する未処理検体のバイオマーカーmRNAの測定値M42とを比較する。 Next, in step S43, the measurement value comparison unit 33 compares the measurement value M31 of the biomarker protein of the test substance-treated sample with the measurement value M32 of the biomarker protein of the untreated sample respectively corresponding to the test substance treatment The measured value M41 of the biomarker mRNA of the sample is compared with the measured value M42 of the biomarker mRNA of the untreated sample corresponding to each.
 次に、ステップS44において、候補物質決定部34は、ステップS43で比較した結果が、変化している場合には、被験物質が有効成分の候補物質であると決定する(ステップS45)。また、候補物質決定部34は、ステップS43で比較した結果が、変化していない場合には、被験物質が有効成分の候補物質ではないと決定する(ステップS46)。ステップS44、ステップS45、ステップS46は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S44, if the comparison result in step S43 indicates a change, the candidate substance determination unit 34 determines that the test substance is a candidate substance of the active ingredient (step S45). In addition, when the result of the comparison in step S43 shows no change, the candidate substance determination unit 34 determines that the test substance is not a candidate substance of the active ingredient (step S46). Steps S44, S45, and S46 correspond to the prediction process described in the claims.
 得られた決定結果は、スクリーニング装置4の表示部9に表示されるか(ステップS47)、スクリーニング装置4内の記録部103に記録される。もしくは、インターネットを介して接続された、スクリーニング装置4の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained determination result is displayed on the display unit 9 of the screening device 4 (step S47) or recorded in the recording unit 103 in the screening device 4. Alternatively, it may be displayed on a display unit of a computer terminal outside the screening device 4 connected via the Internet, for example, at a medical institution.
 第4の実施態様に係る、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングのコンピュータプログラムは、スクリーニング装置4のCPU101に、前記ステップS41~S47を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記スクリーニング装置が前記プログラムを読み取り可能である限り制限されない。 The computer program for screening a candidate substance of an active ingredient for suppressing the functional expression of FGF23 according to the fourth embodiment includes a program for causing the CPU 101 of the screening device 4 to execute the steps S41 to S47. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the screening device can read the program.
4.FGF23の機能を活性化するための有効成分の候補物質のスクリーニング
4-1.概要
 第5の実施態様においては、上記「I.2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、FGF23の機能を活性化するための有効成分の候補物質のスクリーニングを行う。
4. Screening of candidate active ingredients for activating FGF23 function
4-1. Overview In a fifth embodiment, an active ingredient for activating the function of FGF23 using the measurement values obtained by performing the method of “I. 2. Method for obtaining each measurement value” described above is used. Screening of candidate substances.
 より具体的には、本実施態様における被験体のFGF23の機能を活性化するための有効成分の候補物質のスクリーニングする方法は、被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する取得工程と、第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程とを含む。 More specifically, the method of screening for a candidate substance of an active ingredient for activating the function of FGF23 in a subject according to this embodiment is a method for collecting a substance from the skin of a subject (excluding human) treated with a test substance Of the biomarker protein of at least one test substance-treated sample selected from the group consisting of a tested sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin and And / or acquiring the measurement value of mRNA of the protein, and determining the test substance as a candidate substance of the active ingredient based on the measurement value acquired by the first measurement value acquiring means. .
 本実施態様は、好ましくは、上記「I.2.各測定値の取得方法」によって、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程を含む。また、前記被験物質処理検体から取得された各バイオマーカーの測定値と、それぞれに対応する未検体処理中の測定値とを比較し、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が変化していれば、前記被験物質がFGF23の機能を活性化する有効成分であると決定することができる。 In this embodiment, preferably, a test sample obtained from the skin of a subject (except for human beings) not treated with the test substance, a test derived from the skin according to the above-mentioned "I. Measurement value of corresponding biomarker protein and / or mRNA of said protein in at least one unprocessed sample selected from the group consisting of a sample collected from a tissue and a sample collected from a test cell derived from skin Including the step of obtaining a measurement value. In addition, the measured value of each biomarker obtained from the test substance-treated sample is compared with the corresponding measured value in the unsampled process, and compared with the measured value of the biomarker of the untreated sample to be treated with the test substance If the measured value of the biomarker of the sample has changed, it can be determined that the test substance is an active ingredient that activates the function of FGF23.
 ここで、FGF23の機能の活性化とは、FGF23の本来の機能が活性されることをいう。FGF23の機能の活性化の例は、例えば、血中の無機リン濃度を下げることである。また、FGF23の機能の活性化には、FGF23の発現が上昇することを含む。 Here, the activation of the function of FGF23 means that the original function of FGF23 is activated. An example of functional activation of FGF23 is, for example, lowering the concentration of inorganic phosphorus in blood. Moreover, activation of the function of FGF23 includes an increase in the expression of FGF23.
 被験物質処理検体のバイオマーカーの測定値の変化が、正方向であるか負方向であるかは、バイオマーカーに依存する。 Whether the change in the measured value of the biomarker of the test substance-treated sample is positive or negative depends on the biomarker.
 例えば、上記II.4-1で述べた3群のバイオマーカーについては、例えば正常マウスを被験体として、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が減少していれば、当該被験物質は、FGF23の機能を活性化していると決定することができる。また、4群のバイオマーカーについては、正常マウスを被験体として、未処理検体のバイオマーカー測定値と比較して被験物質処理検体のバイオマーカーの測定値が上昇していれば、当該被験物質は、FGF23の機能を活性化と決定することができる。さらに、これらのバイオマーカーの検討に加え、2群のバイオマーカーについても同様に検討した場合に、未処理検体のバイオマーカー測定値と被験物質処理検体のバイオマーカーの測定値に差がなければ、被験物質はFGF23の機能に特異的に作用していると決定することができる。 For example, II. For the three groups of biomarkers described in 4-1, for example, if the normal mouse is a subject, the measured value of the biomarker of the test substance-treated sample is reduced compared to the measured value of the biomarker of the untreated sample. The test substance can be determined to activate the function of FGF23. Also, for the 4 groups of biomarkers, if the measured value of the biomarker of the test substance-treated sample is increased compared to the biomarker measured value of the untreated sample in a normal mouse as a subject, the test substance is The function of FGF23 can be determined as activation. Furthermore, in addition to the examination of these biomarkers, when the two groups of biomarkers are similarly examined, if there is no difference between the measured values of the biomarkers of the untreated sample and the measured values of the biomarkers of the test substance treated sample, The test substance can be determined to act specifically on the function of FGF23.
 被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が、未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値よりも上昇しているか否かは、例えば、被験物質処理検体の測定値が未処理検体の測定値の例えば115%以上、好ましくは130%以上、より好ましくは150%以上になっている場合に、当該被験物質により、バイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が上昇していると決定することがでる。 The measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of the protein is higher than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of the protein For example, when the measured value of the test substance-treated sample is, for example, 115% or more, preferably 130% or more, more preferably 150% or more of the measured value of the untreated sample, the test substance Thus, it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is rising.
 被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が、未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値よりも低下しているか否かは、例えば、被験物質処理検体の測定値が未処理検体の測定値の例えば85%以下、好ましくは70%以下、より好ましくは50%以下になっている場合に、当該被験物質により、バイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値が低下していると決定することがでる。 The measured value of the biomarker protein in the test substance-treated sample and / or the measured value of the mRNA of said protein is lower than the measured value of the corresponding biomarker protein of the untreated sample and / or the measured value of the mRNA of said protein For example, when the measured value of the test substance-treated sample is, for example, 85% or less, preferably 70% or less, more preferably 50% or less of the measured value of the untreated sample, the test substance Thus, it can be determined that the measured value of the biomarker protein and / or the measured value of the mRNA of said protein is decreasing.
 さらに、スクリーニングは、前記工程(I)の前に、(i)被験体、被験組織又は被験細胞に、被験物質を投与する投与工程、(ii)前記工程(i)で被験物質を投与された被験体、被験組織又は被験細胞から検体を採取する工程、及び(iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程を含んでいてもよい。この場合にも、工程(ii)と工程(iii)は、必ずしも同一機関において連続して行われる必要はなく、例えば、工程(ii)で採取された検体を、第三者機関に送り工程(iii)以降を実施してもよい。また、工程(iii)と工程(I)も、必ずしも同一機関において連続して行われる必要はなく、例えば、工程(iii)で回収されたタンパク質及び/又はmRNAを、第三者機関に送り工程(iii)以降を実施してもよい。 Furthermore, in the screening, before the step (I), the test substance is administered in the administration step of (i) administering the test substance to the subject, the test tissue or the test cell, and (ii) the step (i) The method may include the steps of collecting the sample from the subject, the test tissue or the test cells, and (iii) recovering the protein and / or the mRNA from the sample obtained in the step (ii). Also in this case, the step (ii) and the step (iii) do not necessarily need to be performed continuously in the same organization, and for example, the sample collected in the step (ii) is sent to a third party organization iii) The following may be implemented. In addition, step (iii) and step (I) do not necessarily have to be performed continuously in the same organization, for example, the step of sending the protein and / or mRNA recovered in step (iii) to a third party organization (Iii) The following may be implemented.
4-2.スクリーニング装置
 第5の実施態様は、CPU101が、後述するプログラムによって下記の演算機能を実行することによって制御される、下記の演算手段を有する、FGF23の機能を活性化するための有効成分の候補物質のスクリーニング装置5を含む:
 被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、並びに
 前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
4-2. Screening Device A fifth embodiment is a candidate substance of an active ingredient for activating the function of FGF23, having the following calculation means, which is controlled by the CPU 101 executing the following calculation function by a program described later The screening device 5 of:
Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Means for acquiring the measurement value of the biomarker protein of at least one test substance-treated sample and / or the measurement value of mRNA of the protein, and the measurement acquired by the first measurement value acquisition means A determining means for determining that the test substance is a candidate substance of the active ingredient based on a value.
 本実施態様では、上記装置としてスクリーニング装置5を備えたシステム200(図7及び図8)によって、スクリーニングを行うことができる。 In the present embodiment, screening can be performed by the system 200 (FIGS. 7 and 8) provided with the screening device 5 as the above-mentioned device.
 本実施態様に係るスクリーニング装置5の機能を説明するためのブロック図を図9に示す。スクリーニング装置5の構成は、上記II.3-2で述べたスクリーニング装置4と同様である。 The block diagram for demonstrating the function of the screening apparatus 5 which concerns on this embodiment is shown in FIG. The configuration of the screening device 5 is the same as that of the screening device 4 described in the above II.3-2.
4-3.スクリーニング装置の動作とプログラム
 第5の実施態様におけるFGF23の機能を活性化するための有効成分の候補物質のスクリーニングは、第5の実施態様に係るスクリーニング装置5が、下記プログラムによって、本発明の下記のFGF23の機能を活性化するための有効成分の候補物質のスクリーニングを実行してもよい。
4-3. Operation of Screening Device and Program In the fifth embodiment of the present invention, the screening device 5 according to the fifth embodiment uses the following program to screen the candidate substances of the active ingredient for activating the function of FGF23. Screening of candidate active ingredients for activating the function of FGF23 may be performed.
 図11は、本発明の第5の実施態様に係るスクリーニング装置5の動作を示すフローチャートである。なお、図9に示す第1の測定値取得部31により図11のステップS51が、第2の測定値取得部32により図11のステップS52が、測定値比較部33により図11のステップS53が、候補物質決定部34により図11に示すステップS55及びS56の処理がそれぞれ実行される。 FIG. 11 is a flowchart showing the operation of the screening device 5 according to the fifth embodiment of the present invention. Note that step S51 in FIG. 11 is performed by the first measurement value acquisition unit 31 shown in FIG. 9, step S52 in FIG. 11 is performed by the second measurement value acquisition unit 32, and step S53 in FIG. The candidate substance determining unit 34 executes the processing of steps S55 and S56 shown in FIG.
 ステップS51では、初めに、検査者からの被験物質処理検体のバイオマーカータンパク質の測定値M31又は被験物質処理検体のバイオマーカーmRNAの測定値M41の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、第1の測定値取得部31は、前記測定値の取得を開始する。ステップS51は、特許請求の範囲に記載の取得処理に相当する。 In step S51, first, an input for starting acquisition of the measured value M31 of the biomarker protein of the test substance-treated sample or the measured value M41 of the biomarker mRNA of the test substance-treated sample from the examiner is received from the input unit 8 Alternatively, upon receiving a measurement start instruction from the measurement device 5a or 5b, the first measurement value acquisition unit 31 starts acquisition of the measurement value. Step S51 corresponds to the acquisition process described in the claims.
 次に、ステップS52では、第2の測定値取得部32は、未処理検体のバイオマーカータンパク質の測定値M32、未処理検体のバイオマーカーmRNAの測定値M42を取得する。 Next, in step S52, the second measurement value acquisition unit 32 acquires the measurement value M32 of the biomarker protein of the untreated sample and the measurement value M42 of the biomarker mRNA of the untreated sample.
 次に、ステップS53では、測定値比較部33が、被験物質処理検体のバイオマーカータンパク質の測定値M31とそれぞれに対応する未処理検体のバイオマーカータンパク質の測定値M32とを比較し、被験物質処理検体のバイオマーカーmRNAの測定値M41とそれぞれに対応する未処理検体のバイオマーカーmRNAの測定値M42とを比較する。 Next, in step S53, the measurement value comparison unit 33 compares the measurement value M31 of the biomarker protein of the test substance-treated sample with the measurement value M32 of the biomarker protein of the untreated sample respectively corresponding to the test substance treatment The measured value M41 of the biomarker mRNA of the sample is compared with the measured value M42 of the biomarker mRNA of the untreated sample corresponding to each.
 次に、ステップS54において、候補物質決定部34は、ステップS53で比較した結果が、変化している場合には、被験物質が有効成分の候補物質であると決定する(ステップS55)。また、候補物質決定部34は、ステップS53で比較した結果が、変化していない場合には、被験物質が有効成分の候補物質ではないと決定する(ステップS56)。ステップS54、ステップS55、ステップS56は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S54, when the result of comparison in step S53 is changing, the candidate substance determination unit 34 determines that the test substance is a candidate substance of the active ingredient (step S55). In addition, when the result of the comparison in step S53 is not changed, the candidate substance determination unit 34 determines that the test substance is not a candidate substance of the active ingredient (step S56). Steps S54, S55, and S56 correspond to the prediction process described in the claims.
 得られた決定結果は、スクリーニング装置4の表示部9に表示されるか(ステップS57)、スクリーニング装置4内の記録部103に記録される。もしくは、インターネットを介して接続された、スクリーニング装置4の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained determination result is displayed on the display unit 9 of the screening device 4 (step S57) or recorded in the recording unit 103 in the screening device 4. Alternatively, it may be displayed on a display unit of a computer terminal outside the screening device 4 connected via the Internet, for example, at a medical institution.
 第4の実施態様に係る、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニングのコンピュータプログラムは、スクリーニング装置4のCPU101に、前記ステップS51~S57を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記スクリーニング装置が前記プログラムを読み取り可能である限り制限されない。 The computer program of the screening of the candidate substance of the active ingredient for suppressing the functional expression of FGF23 according to the fourth embodiment includes a program that causes the CPU 101 of the screening device 4 to execute the steps S51 to S57. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the screening device can read the program.
III.第6の実施態様
1.用語の説明
 第6の実施態様の説明において、使用する用語について説明する。
III. Sixth embodiment
1. Description of Terms In the description of the sixth embodiment, the terms used will be described.
 本実施態様において使用される用語の説明は、上記I.1.の記載をここに援用する。また、「バイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値」、「バイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値」、の取得方法は、上記I.2.に準ずる。 The explanation of terms used in this embodiment is the same as that described in I. 1. Is incorporated herein by reference. In addition, methods for obtaining “a measured value of at least one type of protein selected from the group consisting of biomarkers” and “a measured value of at least one type of mRNA selected from the group consisting of biomarkers” are described above in I. 2. It conforms to.
 本実施態様において、「疾患」は制限されない。例えば、前記疾患は、前記個体の各器官で発症しうるあらゆる疾患、異常を含みうる。また、当該疾患に至る前に起こる当該特定疾患特有の異常(「前病変」ともいう)を含む。疾患として、好ましくは、血栓症、塞栓症、狭窄症等の虚血性疾患(特に心臓、脳、肺、大腸等);動脈瘤、静脈瘤、うっ血、出血等の循環障害(大動脈、静脈、肺、肝臓、脾臓、網膜等);アレルギー性気管支炎、糸球体腎炎等のアレルギー性疾患;認知症、パーキンソン病、筋萎縮性側索硬化症、重症筋無力症、等の変性疾患(神経、骨格筋等);腫瘍(良性上皮性腫瘍、良性非上皮性腫瘍、悪性上皮性腫瘍、悪性非上皮性腫瘍);代謝性疾患(糖質代謝異常、脂質代謝異常、電解質異常);感染症(細菌、ウイルス、リケッチア、クラミジア、真菌等、原虫、寄生虫等)等が挙げられる。また、前記疾患には、全身症状を引き起こす疾患を含む。全身症状を引き起こす疾患としては、例えば、全身性エリテマトーデス、多発性硬化症等の自己免疫疾患;遺伝性ムコ多糖症等の代謝異常等を挙げることができる。 In the present embodiment, the "disease" is not limited. For example, the disease may include any disease or disorder that may develop in each organ of the individual. In addition, it includes abnormalities specific to the specific disease (also referred to as “pre-lesion”) that occur before the disease is reached. Preferred diseases include ischemic diseases such as thrombosis, embolism, and stenosis (in particular, heart, brain, lung, large intestine, etc.); circulatory disorders such as aneurysm, varicose, congestion, hemorrhage, etc. (aorta, vein, lung Liver, spleen, retina etc.); allergic diseases such as allergic bronchitis, glomerulonephritis; degenerative diseases such as dementia, Parkinson's disease, amyotrophic lateral sclerosis, myasthenia gravis, etc. Muscles, etc .; Tumors (benign epithelial tumors, benign non-epithelial tumors, malignant epithelial tumors, malignant non-epithelial tumors); metabolic diseases (glycometabolic disorders, dyslipidemia, electrolytic abnormalities); infections (bacterial) , Viruses, rickettsial, chlamydia, fungi etc., protozoa, parasites etc. The diseases also include diseases causing systemic symptoms. Examples of diseases that cause systemic symptoms include autoimmune diseases such as systemic lupus erythematosus and multiple sclerosis; metabolic abnormalities such as hereditary mucopolysaccharidosis and the like.
2.ハードウェアの構成
 第6の実施態様で使用されるハードウェアの構成を図12及び図13を用いて説明する。
2. Hardware Configuration The hardware configuration used in the sixth embodiment will be described using FIG. 12 and FIG.
 図12は、本発明の第6の実施態様に係るシステム300の概観図である。システム300は、演算装置(以下では、同様のハードウェア構成を有し、後に説明するような異なる機能構成を有する演算装置に言及することになるため、これら異なる機能構成を有する演算装置を総称するために、「演算装置6」と表記する)と、入力部8と、表示部9と、測定装置5aと、測定装置5bと、を備える。 FIG. 12 is a schematic view of a system 300 according to a sixth embodiment of the present invention. The system 300 refers to arithmetic devices having the same hardware configuration and different functional configurations as described later, and therefore, the system 300 collectively refers to the arithmetic devices having these different functional configurations. For this purpose, it is provided with “the arithmetic unit 6”, the input unit 8, the display unit 9, the measuring device 5a, and the measuring device 5b.
 演算装置6は、例えば汎用のパーソナルコンピュータで構成されており、後述するデータ処理を行うCPU101と、データ処理の作業領域に使用するメモリ102と、処理データを記録する記録部103と、各部の間でデータを伝送するバス104と、外部機器とのデータの入出力を行うインタフェース部105(以下、I/F部と記す)とを備えている。入力部8及び表示部9は、演算装置6に接続されており、入力部8は、キーボード等で構成され、表示部9は、液晶ディスプレイ等で構成されている。入力部8と表示部9とは、一体化されてタッチパネル付き表示装置として実現されてもよい。なお、演算装置6は一体の装置である必要はなく、CPU101、メモリ102、記録部103等が別所に配置され、これらがネットワークで接続されていてもよい。また、入力部8や表示部9を省略した操作者を必要としない装置であってもよい。 The arithmetic device 6 is constituted by, for example, a general-purpose personal computer, and is located between a CPU 101 for performing data processing described later, a memory 102 used for a data processing work area, a recording unit 103 for recording processing data, And an interface unit 105 (hereinafter referred to as an I / F unit) for inputting and outputting data with an external device. The input unit 8 and the display unit 9 are connected to the arithmetic device 6, the input unit 8 is configured by a keyboard or the like, and the display unit 9 is configured by a liquid crystal display or the like. The input unit 8 and the display unit 9 may be integrated and realized as a display device with a touch panel. The arithmetic device 6 does not have to be an integrated device, and the CPU 101, the memory 102, the recording unit 103, and the like may be separately provided and connected via a network. Moreover, the apparatus which does not require the operator which abbreviate | omitted the input part 8 or the display part 9 may be sufficient.
 また、演算装置6と、測定装置5aと、測定装置5bとについても、一カ所に配置される必要は必ずしもなく、別所に設けられた装置間をネットワークで通信可能に接続したシステムを構成でもよい。 Further, the arithmetic device 6, the measuring device 5a, and the measuring device 5b are not necessarily arranged at one place, and a system in which devices provided at different places are communicably connected by a network may be configured. .
 以下の説明においては、特に断らない限り演算装置6が行う処理は、記録部103又はメモリ102に格納されたプログラムに基づいて、実際には演算装置6のCPU101が行う処理を意味する。CPU101はメモリ102を作業領域として必要なデータ(処理途中の中間データ等)を一時記憶し、記録部103に演算結果等の長期保存するデータを適宜記録する。 In the following description, unless otherwise stated, the processing performed by the arithmetic device 6 actually means the processing performed by the CPU 101 of the arithmetic device 6 based on the program stored in the recording unit 103 or the memory 102. The CPU 101 temporarily stores necessary data (intermediate data in the middle of processing, etc.) using the memory 102 as a work area, and records data to be stored for a long time, such as calculation results, in the recording unit 103 as appropriate.
 測定装置5aは、タンパク質を測定するための装置であり、試料置き場51と、反応部52と、検出部53とを備える。試料置き場51にセットされた被験体から採取された検体は、反応部52に設置された抗体捕捉用抗バイオマーカー抗体が固相されたマイクロプレートに分注されインキュベーションされる。必要に応じて未反応の抗原を除去した後、検出抗体がマイクロプレートに分注され、インキュベーションされる。必要に応じて未反応の抗原を除去した後、検出用抗体を検出するための基質がマイクロプレートに分注され、マイクロプレートが検出部53に移動され、基質が反応して発生したシグナルが測定される。また、測定装置5aの別態様は、マイクロアレイ解析によるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロアレイチップ上に分注し、ハイブリダイゼーションを行い、洗浄した後、検出部53に移動させシグナルを検出する。 The measuring device 5a is a device for measuring a protein, and includes a sample storage place 51, a reaction unit 52, and a detection unit 53. The sample collected from the subject set in the sample storage area 51 is aliquoted and incubated in a microplate on which the anti-biomarker antibody for antibody capture placed in the reaction unit 52 is solid phased. After removing the unreacted antigen, if necessary, the detection antibody is aliquoted into a microplate and incubated. If necessary, after removing the unreacted antigen, the substrate for detecting the detection antibody is dispensed to the microplate, the microplate is moved to the detection unit 53, and the signal generated by the reaction of the substrate is measured Be done. Moreover, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by microarray analysis, and the reverse transcription reaction set in the sample storage place 51 is divided on the microarray chip set in the reaction unit 52. After injection, hybridization and washing, the sample is transferred to the detection unit 53 to detect a signal.
 さらに、測定装置5aの別態様は、RT-PCRによるmRNAの測定値の測定するための装置であり、試料置き場51にセットされた逆転写反応物を反応部52にセットされたマイクロチューブ内に分注し、続いて定量的PCR用試薬をマイクロチューブ内に分注する。反応部52でPCR反応を行いながら、検出部53でチューブ内のシグナルを検出する。 Furthermore, another embodiment of the measuring device 5a is a device for measuring the measurement value of mRNA by RT-PCR, in which the reverse transcription reaction set in the sample storage place 51 is placed in the microtube set in the reaction unit 52. Dispense, then dispense quantitative PCR reagents into microtubes. While performing the PCR reaction in the reaction unit 52, the detection unit 53 detects the signal in the tube.
 測定装置5bは、mRNAを測定するための装置であり、配列解析部54を備える。RNA-Seq用の反応を行ったサンプルを配列解析部54にセットし、配列解析部54内で、塩基配列の解析をおこなう。 The measuring device 5b is a device for measuring mRNA, and includes a sequence analysis unit 54. The sample subjected to the reaction for RNA-Seq is set in the sequence analysis unit 54, and the sequence analysis is performed in the sequence analysis unit 54.
 測定装置5a,5bは、有線又は無線によって演算装置6に接続されている。測定装置5aは、タンパク質の測定値をA/D変換して、デジタルデータとして演算装置6に送信する。同様に、測定装置5bは、mRNAの測定値をA/D変換して、デジタルデータとして演算装置6に送信する。これにより、演算装置6は、タンパク質の測定値及びmRNAの測定値を、演算処理可能なデジタルデータとして取得することができる。なお、腎疾患マーカーの測定値は、例えば医療機関(図示せず)からインターネットを介してデジタルデータとして送信される。これにより、演算装置6は、腎疾患マーカーの測定値を、デジタルデータとして取得することができる。 The measuring devices 5a and 5b are connected to the computing device 6 by wire or wirelessly. The measuring device 5a performs A / D conversion of the measured value of the protein, and transmits the converted value as digital data to the computing device 6. Similarly, the measuring device 5b A / D converts the measured value of mRNA, and transmits it to the computing device 6 as digital data. Thereby, the arithmetic unit 6 can acquire the measurement value of the protein and the measurement value of the mRNA as digital data that can be processed. The measured value of the kidney disease marker is transmitted as digital data from, for example, a medical institution (not shown) via the Internet. Thereby, the arithmetic unit 6 can acquire the measurement value of the kidney disease marker as digital data.
3.疾患におけるFGF23の関与を予測する方法
3-1.概要
 第6の実施態様においては、上記「I.2.各測定値の取得方法」の方法を実施することによって取得される測定値を用いて、被験体が有する疾患においてFGF23が関与しているか否かを予測する。
3. Method of predicting the involvement of FGF23 in diseases
3-1. Overview In the sixth embodiment, whether FGF23 is involved in a disease that the subject has, using the measurement values obtained by performing the method of “I.2. Acquisition method of each measurement value”. Predict whether or not.
 より具体的には、疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程と、前記取得工程で取得された測定値に基づいて、前記疾患にFGF23が関与していると決定する工程とを含む。 More specifically, a process for obtaining a measured value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measured value of mRNA of the protein, and the measurement obtained in the obtaining process Determining based on the value that FGF23 is involved in the disease.
 本実施態様は、具体的には、上記「I.2.各測定値の取得方法」によって取得された前記測定値を、測定値それぞれに対応する所定の基準値と比較し、該測定値が前記基準値範囲外である場合には、前記疾患にFGF23が関与していると決定することができる。 Specifically, the present embodiment compares the measured values obtained by the above-mentioned “I.2. Acquisition method of each measured value” with a predetermined reference value corresponding to each measured value, and the measured value When it is outside the reference value range, it can be determined that the disease is related to FGF23.
 また、1群のバイオマーカーの測定値が基準値の範囲外である場合には、前記疾患にFGF23が関与していると決定してもよい。さらに、2群のバイオマーカーの測定値が基準値の範囲内である場合、前記疾患にFGF23が関与していると決定してもよい。より好ましくは、1群のバイオマーカーの測定値が基準値の範囲外であり、2群のバイオマーカーの測定値が基準値の範囲内である場合に、前記リン代謝異常に、前記疾患にFGF23が関与していると決定してもよい。 In addition, when the measured value of one group of biomarkers is out of the range of the reference value, it may be determined that the disease is related to FGF23. Furthermore, if the measured values of the two groups of biomarkers fall within the range of the reference value, it may be determined that the disease is related to FGF23. More preferably, when the measured value of one group of biomarkers is out of the range of the reference value and the measured value of the two groups of biomarkers is within the range of the reference value, May be determined to be involved.
 例えば、上記II.3-1.述べた3群のバイオマーカーは、高リン食を接種したマウスと比較してFGF23のノックアウトマウスで発現が上昇する。また、4群のバイオマーカーは、高リン食を接種したマウスと比較してFGF23のノックアウトマウスで発現が減少する。さらに、Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種(上述の2群)は、FGF23のノックアウトマウスにおいても発現が変化しない。 For example, II. 3-1. The three groups of biomarkers mentioned are up-regulated in FGF23 knockout mice as compared to mice inoculated with a high phosphorus diet. Also, the four groups of biomarkers have reduced expression in FGF23 knockout mice as compared to mice inoculated with a high phosphorus diet. Furthermore, at least one type (the above-mentioned two groups) selected from the group consisting of Aldhl2, Col5a1, Col3a1, C1qtnf6, and Col1a1 does not change the expression in FGF23 knockout mice.
 したがって、3群のバイオマーカーについては、例えば、疾患を有する被験体においてそれぞれのバイオマーカーの基準値と比較して測定値が上昇していれば、当該疾患において、FGF23の機能発現が抑制されていると決定することができる。また、4群のバイオマーカーについては、疾患を有する被験体において、それぞれのバイオマーカーの基準値と比較して測定値が低下していれば、当該疾患において、FGF23の機能発現が抑制されていると決定することができる。さらに、これらのバイオマーカーの検討に加え、2群のバイオマーカーについても同様に検討した場合に、前記被験体において測定値が基準値と比較して変化がなければ、前記疾患においてFGF23の機能が特異的に作用していると決定することができる。 Therefore, for the 3 groups of biomarkers, for example, if the measured value is increased in the subject having the disease as compared to the reference value of each biomarker, the functional expression of FGF23 is suppressed in the disease. Can be determined. In addition, for the 4 groups of biomarkers, if the measured value is lower in the subject having the disease as compared to the reference value of each biomarker, the functional expression of FGF23 is suppressed in the disease. It can be decided. Furthermore, in addition to the examination of these biomarkers, when the two groups of biomarkers are examined in the same manner, if the measured value in the subject does not change compared to the reference value, the function of FGF23 in the disease is It can be determined that it is acting specifically.
 前記工程は、後述するCPU101が行ってもよいが、検査者が行ってもよい。 The process may be performed by the CPU 101 described later, or may be performed by an examiner.
 さらに、前記疾患にFGF23が関与しているか否かの予測方法において、疾患にFGF23が関与していると予測された場合には、FGF23の機能発現を改善するための治療(例えば、低リン食等の食事療法)を行う工程を、含めてもよい。 Furthermore, in a method of predicting whether or not FGF23 is involved in the disease, when it is predicted that FGF23 is involved in the disease, a treatment for improving the functional expression of FGF23 (for example, a low phosphorus diet) Etc.) may be included.
3-2.疾患にFGF23が関与しているか否かを予測する装置
 第6の実施態様は、CPU101が、後述するプログラムによって下記の演算機能を実行することによって下記手段が制御される、疾患におけるFGF23の関与を予測する予測装置を含む:
 疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、並びに 前記第1の測定値取得手段が取得した測定値に基づいて、前記疾患にFGF23が関与していると決定する決定手段。
3-2. Device for predicting whether FGF23 is involved in a disease The sixth embodiment relates to the involvement of FGF23 in a disease in which the following means are controlled by the CPU 101 executing the following arithmetic function by a program described later Including a predictor to predict:
First measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition means A determination means for determining that FGF23 is involved in the disease based on the measurement value obtained by
 本実施態様では、上記装置として演算装置6を備えたシステム300(図12及び図13)によって、疾患におけるFGF23の関与を予測することができる。 In this embodiment, the involvement of FGF23 in a disease can be predicted by the system 300 (FIGS. 12 and 13) provided with the arithmetic device 6 as the above-mentioned device.
 図14は、第6の実施態様に係る演算装置6の機能を説明するためのブロック図である。演算装置6の構成は、上記I.4-2.で述べた演算装置1と同様である。 FIG. 14 is a block diagram for explaining the function of the arithmetic device 6 according to the sixth embodiment. The configuration of the arithmetic unit 6 is the same as that of the above-described I. 4-2. It is the same as the arithmetic unit 1 described above.
3-3.演算装置の動作とプログラム
 第6の実施態様における疾患におけるFGF23の関与を予測する方法は、第6の実施態様に係る演算装置6が、下記プログラムによって、本発明の下記の疾患にFGF23が関与しているか否かの予測方法を実行してもよい。
3-3. Operation of Arithmetic Device and Program The method for predicting the involvement of FGF23 in a disease according to the sixth embodiment is that the arithmetic device 6 according to the sixth embodiment relates FGF23 to the following disease of the present invention by the following program. The prediction method may be performed.
 図15は、本発明の第6の態様に係る演算装置6が動作を示すフローチャートである。なお、図14に示す測定値取得部11により図15のステップS61が、基準値取得部12により図15のステップS62が、測定値比較部13により図15のステップS63が、予測部14により図15に示すステップS65及びS66の処理がそれぞれ実行される。 FIG. 15 is a flowchart showing the operation of the arithmetic device 6 according to the sixth aspect of the present invention. The step S61 in FIG. 15 is performed by the measured value acquiring unit 11 shown in FIG. 14, the step S62 in FIG. 15 is performed by the reference value acquiring unit 12, the step S63 in FIG. The processes of steps S65 and S66 shown in 15 are respectively executed.
 ステップS61では、初めに、検査者からのバイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21の取得開始の入力を入力部8から受け付けることにより、又は測定装置5a又は5bからの測定開始の指令を受け付けることにより、測定値取得部11は、前記測定値の取得を開始する。ステップS61は、特許請求の範囲に記載の取得処理に相当する。 In step S61, first, an acceptance start input of measurement value M11 of biomarker protein or measurement value M21 of biomarker mRNA from the examiner is received from input unit 8, or measurement from measurement device 5a or 5b is started. The measurement value acquisition unit 11 starts acquisition of the measurement value by receiving the command of Step S61 corresponds to the acquisition process described in the claims.
 次に、ステップS62では、基準値取得部12は、検査者による入力部8からの測定値の取得開始の入力又は比較開始の入力に応じて、あるいは測定値取得部11による前記測定値の取得に応じて、バイオマーカータンパク質の基準値R12、バイオマーカーmRNAの基準値R22を取得する。 Next, in step S62, the reference value acquisition unit 12 acquires the measurement value by the measurement value acquisition unit 11 according to an input of acquisition start of measurement value or input of comparison start from the input unit 8 by the examiner. In accordance with, the reference value R12 of the biomarker protein and the reference value R22 of the biomarker mRNA are obtained.
 次に、ステップS63では、測定値比較部13が、バイオマーカータンパク質の測定値M11又はバイオマーカーmRNAの測定値M21と、各バイオマーカーの測定値に対応するバイオマーカータンパク質の基準値R12、又はバイオマーカーmRNAの基準値R22とを比較する。 Next, in step S63, the measurement value comparison unit 13 measures the measurement value M11 of the biomarker protein or the measurement value M21 of the biomarker mRNA, and the reference value R12 of the biomarker protein corresponding to the measurement value of each biomarker, or bio The marker mRNA is compared with the reference value R22.
 次に、ステップS64において、予測部14は、ステップS63で比較した結果が、各バイオマーカーの基準値の範囲内である場合には、疾患においてFGF23が関与していないと予測する(ステップS65)。また、予測部14は、ステップS63で比較した結果が、各バイオマーカーの基準値の範囲外である場合には、疾患においてFGF23が関与していると予測する(ステップS66)。ステップS64、ステップS65、ステップS66は、特許請求の範囲に記載の予測処理に相当する。 Next, in step S64, when the comparison result in step S63 is within the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is not involved in the disease (step S65). . Further, when the comparison result in step S63 is out of the range of the reference value of each biomarker, the prediction unit 14 predicts that FGF23 is involved in the disease (step S66). Steps S64, S65, and S66 correspond to the prediction process described in the claims.
 得られた予測結果は、演算装置6の表示部9に表示されるか(ステップS67)、演算装置6内の記録部103に記録される。もしくは、インターネットを介して接続された、演算装置6の外部の例えば医療機関におけるコンピュータ端末の表示部に表示されてもよい。 The obtained prediction result is displayed on the display unit 9 of the arithmetic device 6 (step S67) or recorded in the recording unit 103 in the arithmetic device 6. Alternatively, it may be displayed on a display unit of a computer terminal, for example, in a medical institution, which is connected via the Internet and which is external to the arithmetic device 6.
 第6の実施態様に係る、疾患にFGF23が関与しているか否かを予測するためのコンピュータプログラムは、演算装置6のCPU101に、前記ステップS61~S67を実行させるプログラムを含む。前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記録媒体に記憶されていてもよい。前記記録媒体へのプログラムの記憶形式は、前記演算装置が前記プログラムを読み取り可能である限り制限されない。 A computer program according to the sixth embodiment for predicting whether or not FGF23 is involved in a disease includes a program that causes the CPU 101 of the arithmetic unit 6 to execute the steps S61 to S67. The computer program may be stored in a recording medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program on the recording medium is not limited as long as the arithmetic device can read the program.
 なお、本項で使用される用語の説明は、上記III.3-1.の説明を援用する。 In addition, the explanation of the term used in this section is the above-mentioned III. 3-1. Use the description of.
IV.検査試薬及び検査キット
 本発明の第7の実施態様の一例は、上記「I.2.測定値の取得方法」に使用される抗バイオマーカー抗体を含む、検査試薬に関する。
IV. Test Reagent and Test Kit An example of the seventh embodiment of the present invention relates to a test reagent comprising the anti-biomarker antibody used in the above-mentioned "I.2.
 抗バイオマーカー抗体は、上記「I.1.用語の説明」に記載したものを使用することができる。 As the anti-biomarker antibody, those described above in "I.1. Explanation of terms" can be used.
 本実施態様の検査試薬には、少なくとも抗バイオマーカー抗体が1種以上含まれていればよい。抗バイオマーカー抗体がポリクローナル抗体である場合には、1種の抗原で免役して得られたポリクローナル抗体であってもよく、また2種以上の抗原で並行して同一個体に免役して得られたポリクローナル抗体であってもよい。さらに、2種以上の抗原をそれぞれ別の動物に接種して得られたそれぞれのポリクローナル抗体を混合してもよい。抗バイオマーカー抗体がモノクローナル抗体である場合には、1種のハイブリドーマから産生されるモノクローナル抗体であってもよいが、2種以上のハイブリドーマから産生されたモノクローナル抗体であって、それぞれのモノクローナル抗体が同一又は異なるエピトープを認識する複数のモノクローナル抗体が2種以上含まれていてもよい。また、ポリクローナル抗体とモノクローナル抗体を1種以上ずつ混合して含んでいてもよい。 The test reagent of this embodiment may contain at least one or more anti-biomarker antibodies. When the anti-biomarker antibody is a polyclonal antibody, it may be a polyclonal antibody obtained by immunizing with one type of antigen, or may be obtained by immunizing the same individual with two or more types of antigens in parallel. It may be a polyclonal antibody. Furthermore, each polyclonal antibody obtained by inoculating different animals with two or more kinds of antigens may be mixed. When the anti-biomarker antibody is a monoclonal antibody, it may be a monoclonal antibody produced from one type of hybridoma, but is a monoclonal antibody produced from two or more types of hybridomas, and each monoclonal antibody is Two or more types of multiple monoclonal antibodies that recognize the same or different epitopes may be included. In addition, one or more kinds of polyclonal antibodies and monoclonal antibodies may be mixed and contained.
 当該検査試薬に含まれる抗バイオマーカー抗体の形態は、特に制限されず、抗バイオマーカー抗体を含む抗血清若しくは腹水等の乾燥状態又は液体状態であってもよい。また、抗バイオマーカー抗体の形態は、精製抗バイオマーカー抗体、抗バイオマーカー抗体を含む免疫グロブリン画分若しくは抗バイオマーカー抗体を含むIgG画分の乾燥状態又は水溶液であってもよい。 The form of the anti-biomarker antibody contained in the test reagent is not particularly limited, and may be in a dry state or liquid state such as an antiserum containing anti-biomarker antibody or ascites fluid. In addition, the form of the anti-biomarker antibody may be a dried state or an aqueous solution of a purified anti-biomarker antibody, an immunoglobulin fraction containing the anti-biomarker antibody, or an IgG fraction containing the anti-biomarker antibody.
 前記形態が、抗バイオマーカー抗体を含む抗血清若しくは腹水の乾燥状態又は液体状態である場合、さらにβ-メルカプトエタノール、DTT等の安定化剤;アルブミン等の保護剤;ポリオキシエチレン(20)ソルビタンモノラウレート、ポリオキシエチレン(10)オクチルフェニルエーテル等の界面活性剤、アジ化ナトリウム等の防腐剤等の少なくとも一つを含んでいてもよい。また、抗バイオマーカー抗体の形態が、精製抗バイオマーカー抗体、抗バイオマーカー抗体を含む免疫グロブリン画分若しくは抗バイオマーカー抗体を含むIgG画分の乾燥状態又は水溶液である場合、さらに、リン酸緩衝液等のバッファー成分;β-メルカプトエタノール、DTT等の安定化剤;アルブミン等の保護剤;塩化ナトリウム等の塩;ポリオキシエチレン(20)ソルビタンモノラウレート、ポリオキシエチレン(10)オクチルフェニルエーテル等の界面活性剤等、アジ化ナトリウム等の防腐剤の少なくとも一つを含んでいてもよい。 When the form is a dry state or liquid state of antiserum or ascites fluid containing an anti-biomarker antibody, a stabilizer such as β-mercaptoethanol and DTT; a protective agent such as albumin; polyoxyethylene (20) sorbitan It may contain at least one of surfactants such as monolaurate, polyoxyethylene (10) octyl phenyl ether, and preservatives such as sodium azide. Furthermore, when the form of the anti-biomarker antibody is a purified anti-biomarker antibody, an immunoglobulin fraction containing the anti-biomarker antibody, or a dried state or an aqueous solution of an IgG fraction containing the anti-biomarker antibody, phosphate buffer Buffer component such as liquid; Stabilizing agent such as β-mercaptoethanol and DTT; Protective agent such as albumin; Salt such as sodium chloride; polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (10) octyl phenyl ether And the like, and may contain at least one preservative such as sodium azide.
 本発明においては、当該抗バイオマーカー抗体は未標識であっても、上述の標識物質で標識されていてもよいが、上述の標識物質で標識されていることが好ましい。標識物質は、上記「I.2.測定値の取得方法」の項に例示されたものを使用することができる。また、本発明においては、抗原捕捉用の抗バイオマーカー抗体が、固相表面等に固定化された状態で提供されるものであってもよい。固相及び固定化については、上記「I.2.測定値の取得方法」の項で例示したとおりである。固相として好ましくは、マイクロプレートである。 In the present invention, the anti-biomarker antibody may be unlabeled or labeled with the above-mentioned labeling substance, but is preferably labeled with the above-mentioned labeling substance. As the labeling substance, those exemplified in the above-mentioned section “I.2. Measurement value acquisition method” can be used. In the present invention, the anti-biomarker antibody for antigen capture may be provided in a state of being immobilized on a solid phase surface or the like. The solid phase and the immobilization are as described above in the section “I.2. Method of obtaining measured value”. Preferably, the solid phase is a microplate.
 さらに、当該検査試薬は、キットとして提供されてもよい。 Furthermore, the test reagent may be provided as a kit.
 抗原捕捉用抗バイオマーカー抗体が予めマイクロプレート等の固相に結合している場合は、本実施態様の検査キットは、抗原捕捉用抗バイオマーカー抗体を固定化した固相と、検出用抗バイオマーカー抗体とを含むことができる。さらに、標識物質が酵素である場合、その基質液を含んでいてもよい。 When the antigen-capture anti-biomarker antibody is previously bound to a solid phase such as a microplate, the test kit of this embodiment comprises a solid phase on which the antigen-capture anti-biomarker antibody is immobilized, and a detection anti-bio And a marker antibody. Furthermore, when the labeling substance is an enzyme, it may contain the substrate solution.
 上述の検査キットは、例えば、図16に示されるようなキットである。検査キット9は、外装箱94と、抗原捕捉用抗体を固相したマイクロプレート92と、標識物質で標識された検出用抗バイオマーカー抗体を含む第1容器91aと、酵素と反応する基質液を含む第2容器91bと、検査キットの添付文書93とを含む。添付文書93には、検査キットの取り扱い方法、保管条件等を記載しておくことができる。洗浄用の水性媒体を含む容器などを外装箱94に同梱してもよい。 The aforementioned test kit is, for example, a kit as shown in FIG. The test kit 9 includes an external box 94, a microplate 92 on which an antibody for capturing an antigen is immobilized, a first container 91a containing an anti-biomarker antibody for detection labeled with a labeling substance, and a substrate solution that reacts with an enzyme. And the package insert 93 of the test kit. In the attached document 93, the handling method of the test kit, storage conditions and the like can be described. A container or the like containing an aqueous medium for washing may be packaged in the packaging box 94.
 第7の実施態様の別の例は、上記「I.2.測定値の取得方法」に使用されるバイオマーカーmRNA検出核酸を含む検査試薬に関する。 Another example of the seventh embodiment relates to a test reagent containing the biomarker mRNA detection nucleic acid used in the above-mentioned "I.2. Method for obtaining measurement value".
 バイオマーカーmRNA検出核酸は、上記「I.1.用語の説明」に記載したものを使用することができる。 As the biomarker mRNA detection nucleic acid, those described above in “I.1. Explanation of terms” can be used.
 マイクロアレイに用いられるバイオマーカーmRNA検出核酸を含む検査試薬は、凍結乾燥状態、又はTris-HCl等のバッファー、EDTA、塩等が含まれる溶液に溶解した状態であってもよい。標的となるバイオマーカーmRNAが複数ある時は、それぞれの検出核酸を別の容器に入れることが好ましい。また、バイオマーカーmRNA検出核酸を基板上に固定して、マイクロアレイチップとして提供してもよい。マイクロアレイの基板は、検出核酸を固相化できるものであれば特に制限はないが、例えばガラス、ポリプロピレン等のポリマー、ナイロン膜等である。検出核酸を基板上に固定する方法も、公知の方法にしたがって行うことができ、例えば検出核酸を固定するための反応性基を含むスペーサーやクロスリンカーを使用することができる。 The test reagent containing the biomarker mRNA detection nucleic acid used for the microarray may be in a lyophilized state or in a solution containing a buffer such as Tris-HCl, EDTA, salt and the like. When there are a plurality of target biomarker mRNAs, it is preferable to place each detection nucleic acid in a separate container. Alternatively, the biomarker mRNA detection nucleic acid may be immobilized on a substrate and provided as a microarray chip. The substrate of the microarray is not particularly limited as long as it can immobilize the detection nucleic acid, and examples thereof include glass, polymers such as polypropylene, and nylon membranes. The method for immobilizing the detection nucleic acid on the substrate can also be performed according to a known method, and for example, a spacer or crosslinker containing a reactive group for immobilizing the detection nucleic acid can be used.
 さらに、バイオマーカーmRNA検出核酸を含む検査試薬は、当該試薬の他、当該核酸の情報、又はこれらの情報にアクセスするための情報を記録した紙、コンパクトディスク等の媒体と共に、キットとして提供されてもよい。 Furthermore, a test reagent containing a biomarker mRNA detection nucleic acid is provided as a kit together with the reagent, a medium such as paper, compact disc or the like on which information of the nucleic acid or information for accessing such information is recorded. It is also good.
 RT-PCRに用いられるバイオマーカーmRNA検出核酸を含む検査試薬は、凍結乾燥状態、又はTris-HCl等のバッファー、EDTA、塩等が含まれる溶液に溶解した状態であってもよい。またプライマーは、フォワードプライマー及びリバースプライマーが別々の容器に入れられた状態で提供されてもよく、混合した状態で提供されてもよい。さらに定量プローブを含む場合でも、定量プローブは、各プライマーと別の容器に入れられた状態で提供されてもよく、また、各プライマーと定量プローブは全て混合された状態で提供されてもよい。 The test reagent containing a biomarker mRNA detection nucleic acid used for RT-PCR may be in a lyophilised state or in a solution containing a buffer such as Tris-HCl, EDTA, salts and the like. Also, the primers may be provided in separate containers provided with the forward primer and the reverse primer, or may be provided in a mixed state. Furthermore, even when the quantitative probe is included, the quantitative probe may be provided in a separate container with each primer, and each primer and the quantitative probe may be provided in a mixed state.
 さらにバイオマーカーmRNA検出核酸を含む検査試薬は、フォワードプライマー及びリバースプライマーを含む、又はフォワードプライマー、リバースプライマー及び定量用プローブ、必要に応じて添付書類を含む、キットの形態で提供されてもよい。さらにキットには、定量的PCR用の試薬を同梱してもよい。 Furthermore, a test reagent containing a biomarker mRNA detection nucleic acid may be provided in the form of a kit, which comprises a forward primer and a reverse primer, or a forward primer, a reverse primer and a quantitative probe, and optionally a package insert. Furthermore, the kit may include reagents for quantitative PCR.
 バイオマーカーmRNA検出核酸を含む検査試薬はキットとして提供されてもよい。 The test reagent containing the biomarker mRNA detection nucleic acid may be provided as a kit.
 第7の実施態様に係る検査試薬及び検査キットは、第1~第6の実施態様において各測定値を取得するために使用することができる。 The test reagent and the test kit according to the seventh embodiment can be used to obtain each measurement value in the first to sixth embodiments.
V.バイオマーカー
 本発明の第8の実施態様は、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種を含む、腎機能、リン代謝異常、又はFGF23の関与を予測するための皮膚のバイオマーカーとして使用する方法に関する。
V. Biomarker The eighth embodiment of the present invention is Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, Il22ra2, Rbfox1, Lrrc2, Col15a1, Aldh1l, Elc1 The present invention relates to a method for use as a biomarker of skin for predicting involvement of renal function, abnormal phosphorus metabolism, or FGF23, comprising at least one selected from the group consisting of Col11a1, Clec11a, Col3a1, Serpinb6d, Col1a1, and Defb8.
 これらのバイオマーカーは、各検体に含まれる。 These biomarkers are included in each sample.
 以下に実施例を示して本発明の概要をより詳しく説明するが、本発明は実施例に限定して解釈されるものではない EXAMPLES The following will explain the summary of the present invention in more detail by way of examples, but the present invention is not construed as being limited to the examples.
実験例I.Fgf23遺伝子変異マウスの作製
I-1.gRNA及びCas9発現ベクターの構築
 Optimized CRISPR design tool [Massachusetts Institute of Technology, Zhang LabのHP (http://crispr.mit.edu/) において公開] を用いて、gRNA配列 (Fgf23g-RNA1, Fgf23-gRNA2)を設計し、gRNA配列をコードするオリゴDNAを合成した。これらの配列に含まれるFgf23遺伝子標的配列は、Fgf23-gRNA1がGCACGCCCACCAGGAGTCTA(配列番号1)及びFgf23-gRNA2がGCCCACCAGGAGTCTAAGGC(配列番号2)に示される配列であり、Fgf23遺伝子のエクソン1に存在する配列である。これらを、Cas9発現ベクターであるpX330-U6-Chimeric_BB-CBh-hSpCas9に挿入した(pX330-Fgf23-gRNA1, pX330-Fgf23-gRNA2) 。得られたベクターについて、gRNA挿入部位の塩基配列を決定し、設計通りにgRNAが挿入されたことを確認した。また、それぞれのFgf23遺伝子標的配列をはさむようにsingle-stranded oligodeoxynuleotides (ssODNs) を合成した。ドナーオリゴDNAは、Fgf23-gRNA1及びFgf23-gRNA2切断予定部位のLeucineコード配列直下に終止コドンを置くデザインとした(図17A)。ssODNs for Fgf23-gRNA1, 2:
CGGATAGGCTCTAGCAGTGCCCAAGCTGCAGACAGTGCAGAGCACGCCCACCAGGAGTCTcctcagaagaactcgtcaagaagCTAAAGGCAGGTCCCTAGCATTGCACAGCACTGAGTGGCTAATGCTGAGTTTGAAATCTGACA(配列番号3)
Experimental Example I. Preparation of Fgf23 gene mutant mice
I-1. Construction of gRNA and Cas9 Expression Vectors Using the Optimized CRISPR design tool [published in HP of Massachusetts Institute of Technology, Zhang Lab (http://crispr.mit.edu/)], gRNA sequences (Fgf23g-RNA1, Fgf23-gRNA2 Was designed to synthesize an oligo DNA encoding a gRNA sequence. The target sequences of the Fgf23 gene contained in these sequences are the sequences in which Fgf23-gRNA1 is indicated by GCACGCCCACCAGGA GTCTA (SEQ ID NO: 1) and Fgf23-gRNA2 is indicated by GCCCACCAGGAGTCTAAGGC (SEQ ID No. 2), and the sequence present in exon 1 of the Fgf23 gene is there. These were inserted into the Cas9 expression vector pX330-U6-Chimeric_BB-CBh-hSpCas9 (pX330-Fgf23-gRNA1, pX330-Fgf23-gRNA2). The base sequence of the gRNA insertion site was determined for the obtained vector, and it was confirmed that the gRNA was inserted as designed. In addition, single-stranded oligodeoxynucleotides (ssODNs) were synthesized so as to sandwich each Fgf23 gene target sequence. The donor oligo DNA was designed to have a stop codon immediately below the Leucine coding sequence at the planned cleavage site for Fgf23-gRNA1 and Fgf23-gRNA2 (FIG. 17A). ssODNs for Fgf23-gRNA1, 2:
CGGATAGGCTCTAGCAGTGCCCAAGCTGCAGACAGTGCAAGAGCAGCGCCCAGGAGTCTcctcagaagaactcgtcaagaagCAGGTCCAGTCATGGCACAGCACTGAGTGGCTAATGCTGAGTTTGAATCTGACAA (SEQ ID NO: 3)
I-2.Fgf23遺伝子変異マウスの作製
 pX330-Fgf23-gRNA1とpX330-Fgf23-gRNA2を対応するssODNsと共にC57BL/6N Slc受精卵にそれぞれインジェクションした。インジェクションした受精卵は、偽妊娠ICRメスマウスの卵管に注入した。F0マウスをPCRとダイレクトシークェンス(プライマーは表5参照)でジェノタイプを確認した。F1ヘテロ接合体マウスをF0マウスと野生型マウスの交配により作製した。得られたF1ヘテロ接合体マウスオスと野生型マウスメスの体外受精により、F2ヘテロ接合体マウスを作製し、得られたF2ヘテロ接合体マウスメスとF1ヘテロ接合体マウスオスの体外受精により、F3マウスを作製した。また、F3ホモ接合体マウスメス及びF3ヘテロ接合体マウスメスとF1ヘテロ接合体マウスオスの体外受精により、F4マウスを作製した。実験には、F3世代以降のホモ接合体を用いた。
I-2. Preparation of Fgf23 Gene Mutant Mice pX330-Fgf23-gRNA1 and pX330-Fgf23-gRNA2 were injected into C57BL / 6N Slc fertilized eggs together with the corresponding ssODNs. The injected fertilized eggs were injected into the fallopian tube of a pseudopregnant ICR female mouse. Genotype was confirmed by PCR and direct sequencing (see Table 5 for primers) of F0 mice. F1 heterozygous mice were generated by mating F0 mice with wild-type mice. F2 heterozygous mice were produced by in vitro fertilization of the obtained F1 heterozygous male mice and wild type mouse females, and F3 mice were produced by in vitro fertilization of the obtained F2 heterozygous female mice and F1 heterozygous male mice. . In addition, F4 mice were prepared by in vitro fertilization of F3 homozygous female mice and F3 heterozygous female mice and F1 heterozygous male mice. For the experiments, homozygotes from the F3 generation onward were used.
 それぞれのマウスのジェノタイプは、ダイレクトシークェンスで決定した。 Genotype of each mouse was determined by direct sequence.
 得られた変異マウスのジェノタイプの配列を図17Bに示す。 The sequence of the resulting mutant mouse genotype is shown in FIG. 17B.
実験例II.疾患モデルマウスの作製
II-1.UNx/HPiモデルマウスの作成と組織の摘出
 UNx/HPiマウス(片腎切除-高リン食マウス)は、片腎摘出後に高リン食で飼育して作製した。対照として、偽手術後に低リン食で飼育したマウスを作成した。
Experimental Example II. Production of disease model mice
II-1. Preparation of UNx / HPi Model Mouse and Extraction of Tissue UNx / HPi mouse (one nephrectomy-high phosphorus diet mouse) was prepared by rearing on a high phosphorus diet after single nephrectomy. As a control, mice were raised on a low phosphorus diet after a sham operation.
1-1.片腎摘出
 マウス(C57BL/6J、8週齢オス)を、Avertin (250 mg/kg)の腹腔内投与で麻酔後、背部より皮膚を切開、右腎動静脈と尿管を結紮した。結紮部より遠位側で切断し、右腎を摘出した後閉腹した。対照については、偽手術を施した。偽手術においては、右腎動静脈と尿管を露出した後、結紮せずにそのまま閉腹した。手術侵襲から完全に回復するのを待つ意味で、0.54%無機リン含有普通食 (CE-2, 日本クレア)で4週間飼育した。
1-1. After anesthetizing a single nephrectomized mouse (C57BL / 6J, 8-week-old male) with Avertin (250 mg / kg) intraperitoneally, the skin was incised from the back and the right renal arteriovenous vein and ureteral were ligated. It was cut distal to the ligature, and after removing the right kidney, it was closed. For controls, sham surgery was performed. In the sham operation, after exposing the right renal arteriovenous and ureteral tubes, they were closed without ligation. The animals were bred for 4 weeks on a normal diet containing 0.54% inorganic phosphorus (CE-2, CLEA Japan, Inc.) in the sense of waiting for complete recovery from surgical invasion.
1-2.リン負荷と組織の摘出
 手術終了4週間後(12週齢)から、片腎摘出したマウスには2%無機リン含有高リン食(TD.10662, オリエンタルバイオサービス)を与えた(以下、腎疾患群ともいう)。偽手術したマウスには0.35%無機リン含有低リン食(TD.10662変型, オリエンタルバイオサービス)を与えた(以下、Sham群ともいう)。
1-2. Four weeks after the end of the phosphorus load and tissue removal surgery (12 weeks of age), mice that had one-sided nephrectomy were given a 2% inorganic phosphorus-containing high phosphorus diet (TD. 10662, Oriental Bioservice) (following, renal disease) Also called a group). Sham-operated mice were given a low phosphorus diet (TD. 10662 variant, Oriental Bioservice) containing 0.35% inorganic phosphorus (hereinafter also referred to as Sham group).
 この慢性腎疾患モデルは、Hu MC et al.(J Am Soc Nephrol 22, 124-136, 2011)に記載の方法の変法である。Hu MC et al.は、上記(1)の片腎摘出時、残存腎(左腎)に虚血再灌流障害を加えるが、今回の変法では、虚血再灌流は行わなかった。
高リン食開始後(Sham群においては低リン食)、1週間後(E)、4週間後(M)及び8週間後(L)に組織を摘出した。
This chronic kidney disease model is a modification of the method described in Hu MC et al. (J Am Soc Nephrol 22, 124-136, 2011). Hu MC et al. Added ischemia reperfusion injury to the remaining kidney (left kidney) at the time of half nephrectomy described in (1) above, but in this modification, ischemia reperfusion was not performed.
Tissues were removed after 1 week (E), 4 weeks (M) and 8 weeks (L) after initiation of the high phosphorus diet (low phosphorus diet in the Sham group).
 組織を摘出する動物は、Avertin (250 mg/kg) 腹腔内投与による麻酔下で、眼窩よりEDTA添加チューブへの採血を行い、頸椎脱臼して安楽死させ、器官、及び組織(骨髄、脳、皮膚、心臓、腎臓、肝臓、肺、膵臓、骨格筋、脾臓、精巣、胸腺、脂肪、大腸、胃、副腎、大動脈、目、回腸、空腸、下垂体、頭蓋骨、唾液腺及び甲状腺)を摘出した。摘出した器官、及び組織は湿重量を測定した後、液体窒素で速やかに凍結後-80℃にて保存した。採取した血液は、室温にて10分間1200gで遠心した。遠心後に上清の血漿を回収し、-80℃にて保存した。 The animals from which the tissue is removed are bled from the orbital to the EDTA-added tube under anesthesia by intraperitoneal administration of Avertin (250 mg / kg), and cervical dislocation is euthanized, and organs and tissues (bone marrow, brain, The skin, heart, kidney, liver, lung, pancreas, skeletal muscle, spleen, testis, thymus, fat, large intestine, stomach, adrenal gland, aorta, eye, ileum, jejunum, pituitary, skull, salivary gland and thyroid) were removed. The excised organs and tissues were measured for wet weight and immediately frozen with liquid nitrogen and stored at -80 ° C. The collected blood was centrifuged at 1200 g for 10 minutes at room temperature. The supernatant plasma was collected after centrifugation and stored at -80.degree.
II-2. リン負荷マウスモデルの作成と組織の摘出
 Fgf23遺伝子変異マウスはFgf23機能の喪失により、生後から高リン状態をきたし、早期に死亡する。今回、4週齢のFgf23遺伝子変異マウス(オス及びメス)を実験に用いて、この対象群としてリン負荷マウスモデルを、WTマウス(C57BL/6N、3週齢オス)に高リン食を1週間与えて作成した。
II-2. Preparation of phosphorus-loaded mouse model and excision of tissue Fgf23 gene mutant mice develop hyperphosphate from postnatal age due to loss of Fgf23 function and die early. In this experiment, a 4-week old Fgf23 mutant mouse (male and female) is used for experiments, and a phosphorus-loaded mouse model is used as a target group, and a WT mouse (C57BL / 6N, 3-week-old male) has a high phosphorus diet for 1 week. I gave it and made it.
2-1.リン負荷
 WTマウス (C57BL/6N、2週齢オス)を授乳メスマウスとともに1週間飼育した後に離乳して、特別リン含有食として2%無機リン含有高リン食(TD.10662, オリエンタルバイオサービス) 又は、0.35%無機リン含有低リン食 (TD.10662変型, オリエンタルバイオサービス) 又は、0.54%無機リン含有普通食 (CE-2, 日本クレア)を1週間与えた。上記の各モデルを以下、WT3W/HP1W, WT3W/LP1W, WT3W/ND1Wとする
2-1. Phosphorus-loaded WT mice (C57BL / 6N, 2 weeks old males) are bred with lactating female mice for 1 week and then weaned to obtain a special phosphorus-containing diet and a 2% inorganic phosphorus-rich high phosphorus diet (TD. 10662, Oriental Bioservice) or A 0.35% inorganic phosphorus-containing low phosphorus diet (TD. 10662 variant, Oriental Bioservice) or a 0.54% inorganic phosphorus-containing normal diet (CE-2, CLEA Japan, Inc.) was given for one week. Each of the above models is hereinafter referred to as WT3W / HP1W, WT3W / LP1W, WT3W / ND1W
2-2.組織の摘出
 WTマウスへの特別リン含有食の開始後1週間 (4週齢)に頭蓋骨と皮膚を摘出した。Fgf23遺伝子変異マウス(オス及びメス)は4週齢で、頭蓋骨と皮膚を摘出した。組織を摘出する動物は、Avertin (250 mg/kg) の腹腔内投与で麻酔後、眼窩よりEDTA添加チューブへの採血を行い、頸椎脱臼による安楽死の後に、頭蓋骨と皮膚を摘出した。摘出した組織は重量を測定した後、液体窒素で速やかに凍結後、-80℃にて保存した。採取した血液は、室温にて10分間1200gで遠心した。遠心後に上清の血漿を回収し、-80℃にて保存した。
2-2. Tissue Extraction The skull and skin were excised one week (4 weeks old) after the start of the special phosphorus-containing diet in WT mice. Fgf23 gene mutant mice (male and female) were 4 weeks old and the skull and skin were removed. The animals from which the tissue was removed were anesthetized with intraperitoneal injection of Avertin (250 mg / kg), blood was collected from the orbital side into a tube added with EDTA, and the skull and skin were removed after euthanasia by cervical dislocation. The excised tissue was weighed and immediately frozen with liquid nitrogen and then stored at -80 ° C. The collected blood was centrifuged at 1200 g for 10 minutes at room temperature. The supernatant plasma was collected after centrifugation and stored at -80.degree.
実験例III:血漿中の無機リン測定
 E-/M-/L-UNx/HPi, E-/M-/L-Sham, 4週齢のFgf23遺伝子変異マウス, WT3W/HP1W, WT3W/LP1W, WT3W/ND1Wの各マウスで、凍結保存していた血漿サンプルを各々100μlずつ用いて血漿中無機リン値を、酵素法により測定した。
Experiment III: Measurement of inorganic phosphorus in plasma E- / M- / L-UNx / HPi, E- / M- / L-Sham, 4-week-old Fgf23 mutant mouse, WT3W / HP1W, WT3W / LP1W, WT3W In each mouse of / ND1W, 100 μl each of the cryopreserved plasma sample was used to measure the inorganic phosphorus level in plasma by the enzyme method.
実験例IV.各組織における遺伝子発現解析
IV-1.各組織からのRNA抽出
 凍結保存された各組織を、TRIzol Reagent (Thermo Fisher Scientific, MA, USA)中で、PT10-35 GT Polytron homogenizer (KINEMATICA, Luzern, Switzerland) で15,000 rpm で10秒間ホモジナイズするか、乳鉢と乳棒を使い液体窒素中で粉砕、乾燥させた後、TRIzol Reagent中で、PT10-35 GT Polytron homogenizerで15,000 rpm で10秒間ホモジナイズするか、Cell Destroyer PS1000又はPS2000 (Bio Medical Science Inc., Tokyo, Japan) で異なるサイズのジルコニアビーズ(1.5 mm diameter × 50, 3 mm diameter ×5, 5 mm diameter× 2)を用いて4,260 rpmで45秒間、4℃でホモジナイズした。その後タンパク質を分離するため室温にて5分インキュベート後、1 ml のTRIzolに対して0.2mlのクロロホルムを加え、チューブの蓋をした後に15秒間激しくボルテックスした。撹拌後3分室温でインキュベートし、4℃で15分間12,000 gで遠心し、RNAを含む水相を新しいチューブに回収した。回収した水相に等量の70%エタノールを加え撹拌後、RNeasy mini column (Qiagen)に700μlずつアプライしRNeasy mini kit(Qiagen)標準プロトコルに従って精製RNAを回収した。回収したRNAはNanodrop (Thermo Fisher Scientific, MA, USA)にて品質及び濃度の確認を行った。
Experimental Example IV. Gene expression analysis in each tissue
IV-1. Extraction of RNA from each tissue Cryopreserved tissue is homogenized in TRIzol Reagent (Thermo Fisher Scientific, MA, USA) for 10 seconds at 15,000 rpm with PT10-35 GT Polytron homogenizer (KINEMATICA, Luzern, Switzerland) After grinding and drying in liquid nitrogen using a mortar and pestle, homogenize for 10 seconds at 15,000 rpm with PT10-35 GT Polytron homogenizer in TRIzol Reagent, or use Cell Destroyer PS1000 or PS2000 (Bio Medical Science Inc., Homogenize at 4 ° C. for 45 seconds at 4,260 rpm using zirconia beads (1.5 mm diameter × 50, 3 mm diameter × 5, 5 mm diameter × 2) of different sizes in Tokyo, Japan). After incubation for 5 minutes at room temperature to separate proteins, 0.2 ml of chloroform was added to 1 ml of TRIzol, and the tube was capped and vortexed vigorously for 15 seconds. After stirring, the mixture was incubated at room temperature for 3 minutes, centrifuged at 12,000 g for 15 minutes at 4 ° C., and the aqueous phase containing RNA was collected in a new tube. An equal volume of 70% ethanol was added to the collected aqueous phase, and after stirring, 700 μl each was applied to an RNeasy mini column (Qiagen), and purified RNA was recovered according to the RNeasy mini kit (Qiagen) standard protocol. The recovered RNA was subjected to quality and concentration confirmation with Nanodrop (Thermo Fisher Scientific, MA, USA).
IV-2.RNAの発現解析(RNASeq)
(1)RNAseqデータの取得
 上記試料を使用してRNAseqのデータを以下の手順で取得した。
a.品質検査
 下記項目にて、受入サンプルの品質検定を行った。
  ・Nanodrop(分光光度計)を用いた濃度測定
  ・Agilent 2100 Bioanalyzerによる濃度測定・品質の確認
b.サンプル調製
 SureSelect Strand-Specific RNA ライブラリ調製キットを用いて次世代シークエンサー HiSeq 用ライブラリを以下の工程に従い調製した。
i. Total RNA から、オリゴ(dT)磁性ビーズを用いて、poly (A)+RNA (mRNA) を回収
ii. RNA の断片化
iii. cDNA 合成
iv. 2 本鎖 cDNA 合成
v. 末端修復、リン酸化、A テイル付加
vi. インデックス付アダプターのライゲーション
vii. 13 サイクルPCR
viii. 磁性ビーズによる精製
c.次世代シーケンサによるデータ取得
次世代シーケンサHiSeq 2500又は4000 (illumina 社)を使用し、以下の工程に従いシーケンスを行った。
i. シーケンス試薬の添加
  試薬:TruSeq PE Cluster Kit v3 - cBot - HS (1 flowcell) <PE-401-3001> (illumina 社)
  試薬:TruSeq SBS Kit v3 - HS (200 cycle) <FC-401-3001> (illumina 社)
ii. 1塩基伸長反応
iii. 未反応塩基の除去
iv. 蛍光シグナルの取り込み
v. 保護基と蛍光の除去
    2Cycle…3Cycle…とサイクルを繰り返し、100 cycle まで実施。
vi. 逆鎖(Read2)について、i~viを100 cycle まで実施
IV-2. RNA expression analysis (RNASeq)
(1) Acquisition of RNAseq Data Using the above sample, data of RNAseq were acquired according to the following procedure.
a. Quality inspection The quality inspection of the receiving sample was performed in the following items.
-Measurement of concentration using Nanodrop (spectrophotometer)-Measurement of concentration using Agilent 2100 Bioanalyzer-Confirmation of quality
b. Sample Preparation A library for the next generation sequencer HiSeq was prepared according to the following steps using SureSelect Strand-Specific RNA Library Preparation Kit.
i. Recover poly (A) + RNA (mRNA) from Total RNA using oligo (dT) magnetic beads
ii. Fragmentation of RNA
iii. cDNA synthesis
iv. Double-stranded cDNA synthesis
v. End repair, phosphorylation, A tail addition
vi. Indexed Adapter Ligation
vii. 13 cycle PCR
viii. Purification by magnetic beads
c. Data acquisition by next-generation sequencer Using the next-generation sequencer HiSeq 2500 or 4000 (illumina), sequencing was performed according to the following steps.
i. Addition of sequencing reagent Reagent: TruSeq PE Cluster Kit v3-cBot-HS (1 flowcell) <PE-401-3001> (illumina)
Reagents: TruSeq SBS Kit v3-HS (200 cycle) <FC-401-3001> (illumina)
ii. Single base extension reaction
iii. Removal of unreacted base
iv. Incorporation of fluorescent signal
v. Removal of protecting group and fluorescence 2 Cycle ... 3 Cycle ... Repeat the cycle up to 100 cycles.
vi. Perform i to vi up to 100 cycles for reverse chain (Read 2)
(2)RNAseqデータの解析
(2)-1.次世代シーケンサ出力データの解析
上記出力データについて、下記に挙げる情報処理を実施した。
 i. ベースコール:出力された解析生データ(画像データ)より、塩基配列のテキストデータを取得した。
 ii.フィルタリング:所定のフィルタリングによるリードデータの選別を行った。 iii.Index配列による振り分け:Index情報による各サンプルデータの振り分けを行った。
(2) Analysis of RNAseq data
(2) -1. Analysis of Next-Generation Sequencer Output Data The following information processing was performed on the output data.
i. Base call: Text data of the base sequence was acquired from the output analysis raw data (image data).
ii. Filtering: Screening of lead data by predetermined filtering was performed. iii. Sorting by Index arrangement: Each sample data was sorted by Index information.
(2)-2.出力されたデータの2次解析
 Illumina Hiseq2500又は4000にて得られたデータファイル(Fastq形式)をローカルサーバーにダウンロードしたGalaxy (https://usegalaxy.org/) 上にアップロードした。その後マウスゲノムマップ情報mm10に各配列をマッピングするためにBowtie2(http://bowtie-bio.sourceforge.net/bowtie2/index.shtml) を用いて解析した。Bowtie2で得られたBAMファイルをCufflinks (http://cole-trapnell-lab.github.io/cufflinks/) にて解析することで遺伝子のFPKM(RPKM)を算出した。すなわちこの解析では、各組織に発現している全てのRNAを解析対象とした。
(2) -2. Secondary Analysis of Output Data The data file (Fastq format) obtained by Illumina Hiseq 2500 or 4000 was uploaded onto Galaxy (https://usegalaxy.org/) downloaded to the local server. Then, in order to map each sequence to mouse genome map information mm10, analysis was performed using Bowtie 2 (http://bowtie-bio.sourceforge.net/bowtie2/index.shtml). The gene for FPKM (RPKM) was calculated by analyzing the BAM file obtained by Bowtie 2 with Cufflinks (http://cole-trapnell-lab.github.io/cufflinks/). That is, in this analysis, all RNAs expressed in each tissue were analyzed.
IV-3.qRT-PCR
 頭蓋骨及び皮膚から得られたTotal RNA 0.5~1 μgをcDNA合成のテンプレートとしOligo dT20プライマーを用いてSuperscrtipt III First-Strand Synthesis Supermix (Thermo Fisher Scientific, MA, USA) の標準プロトコルに従いcDNAを合成した。合成されたcDNAをTEバッファー(10mM Tris-HCl pH8.0、0.1 mM EDTA)にて20倍希釈した後、LightCycler 480 SYBR Green I Master (Roche, Basel, Switzerland)の標準プロトコルに従い、LightCycler480II (Roche)にてリアルタイムPCRを行いCp値を測定した。各遺伝子で得られたCp値はreference geneとしてMaeaのCp値と比較することでreference geneに対する各遺伝子の相対的発現量を定量した。リアルタイムPCRで使用したプライマーペアは表5の通りである。全てのプライマーはPrimer-BLAST(NCBI)で設計した。
IV-3. qRT-PCR
CDNA was synthesized according to the standard protocol of Superscrtipt III First-Strand Synthesis Supermix (Thermo Fisher Scientific, MA, USA) using Oligo dT20 primers using 0.5-1 μg of total RNA obtained from skull and skin as a template for cDNA synthesis. The synthesized cDNA is diluted 20-fold with TE buffer (10 mM Tris-HCl pH 8.0, 0.1 mM EDTA), and then LightCycler 480 II (Roche) according to the standard protocol of LightCycler 480 SYBR Green I Master (Roche, Basel, Switzerland) Real-time PCR was performed to measure Cp value. The Cp value obtained for each gene was compared with the Cp value of Maea as a reference gene to quantify the relative expression level of each gene relative to the reference gene. The primer pairs used in real time PCR are as shown in Table 5. All primers were designed by Primer-BLAST (NCBI).
Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-I000009
Figure JPOXMLDOC01-appb-T000008
Figure JPOXMLDOC01-appb-I000009
IV-4.発現差のある遺伝子の解析
 発現差のある遺伝子を抽出するため、Bowtie2によってマッピングされた配列データについてHTSeq-count(パラメーターは -rがpos、及び -sがno)を使用してそれぞれの転写物のアノテーションリードナンバーをカウントした。得られた結果は、DESeq2(Love, M. I., Huber, W. & Anders, S.; Genome biology 15, 550, doi:10.1186/s13059-014-0550-8 (2014))解析により、デフォルト設定で行った。発現差は、E-UNx/HPi vs. E-/M-/L-Sham (n=3)、M-UNx/HPi vs. E-/M-/L-Sham (n=3)、L-UNx/HPi vs. E-/M-/L-Sham (n=3)で比較した。
IV-4. Analysis of differentially expressed genes To extract differentially expressed genes, HTSeq-count (parameters -r is pos and -s is no) on the sequence data mapped by Bowtie 2 for each transcript The annotation lead number of was counted. The obtained result is performed with default settings by DESeq 2 (Love, MI, Huber, W. & Anders, S .; Genome biology 15, 550, doi: 10.1186 / s13059-014-0550-8 (2014)) analysis. The E-UNx / HPi vs. E- / M- / L-Sham (n = 3), M-UNx / HPi vs. E- / M- / L-Sham (n = 3), L- The comparison was made with UNx / HPi vs. E- / M- / L-Sham (n = 3).
IV-5.統計解析
 統計解析は、ステューデントのt検定を行い、有意差を求めた。そしてp値が0.05より小さい場合に有意差有りと定義した。
IV-5. Statistical Analysis Statistical analysis was performed by Student's t-test to obtain a significant difference. And when p value was smaller than 0.05, it defined as significant difference.
IV-6.結果
 E-UNx/HPi vs. E-/M-/L-Sham (n=3) の比較で、皮膚において発現差がある遺伝子リスト(|log(fold-change)| > 1, p-value < 0.05)がDESeq2解析により得られた。UNx/HPi及びWT3W/HP1W, WT3W/LP1W, WT3W/ND1Wマウスモデルの皮膚における、これらの遺伝子の発現をqRT-PCRにより検証し(n=6-9)、図18-1~図18-7及び表6に示す25遺伝子で発現に差がある事が示された。WT3W/HP1Wについては発現差をWT3W/LP1Wと比較した。
IV-6. Results List of genes with differential expression in skin compared to E-UNx / HPi vs. E- / M- / L-Sham (n = 3) (| log 2 (fold-change) |> 1, p-value <0.05) was obtained by DESeq2 analysis. The expression of these genes was verified by qRT-PCR in the skin of UNx / HPi and WT3W / HP1W, WT3W / LP1W, and WT3W / ND1W mouse models (n = 6-9), as shown in FIGS. And it was shown that there is a difference in expression in 25 genes shown in Table 6. The differential expression was compared to WT3W / LP1W for WT3W / HP1W.
 この25遺伝子について、Fgf23遺伝子の機能発現との関連を評価するために、4週齢のFgf23遺伝子変異マウス(オス及びメス)の皮膚における発現をqRT-PCRで検証した(n=7)。尚、E-/M-/L-UNx/HPi, WT3W/HP1Wの骨 (頭蓋骨) において、Fgf23の発現が上昇している事は、qRT-PCRにより確認しており(図19A及びB)、またFgf23遺伝子変異マウスでは、図20に示すように、血中無機リン濃度が上昇しているため、Fgf23遺伝子変異マウスの対照は、同週齢(4週齢)の高リン負荷マウス(WT3W/HP1W, n=3)とした。 The expression of these 25 genes in the skin of 4-week-old Fgf23 gene mutant mice (male and female) was verified by qRT-PCR (n = 7) in order to evaluate the relationship with functional expression of the Fgf23 gene. In addition, it was confirmed by qRT-PCR that expression of Fgf23 is increased in bones (skulls) of E- / M- / L-UNx / HPi and WT3W / HP1W (FIGS. 19A and B), In addition, as shown in FIG. 20, in the Fgf23 gene mutant mice, the blood inorganic phosphorus concentration is increased, and therefore the control of the Fgf23 gene mutant mice is a high phosphorus load mouse (WT 3 W / w) at the same age (4 weeks old). HP1W, n = 3).
 Fgf23遺伝子変異マウスとWT3W/HP1Wの比較においては、図21-1~図21-7及び表6に示すようにいくつかの遺伝子において、統計学的に有意な発現の上昇及び低下が認められた。Fgf23遺伝子変異マウスでは、性差による発現の差も認められた。以上より、高リン状態の皮膚で発現変動する遺伝子は、図22に示すFgf23遺伝子機能発現に依存又は非依存性の、4タイプのモデルに分別できる事が示された。 In comparison of Fgf23 gene mutant mice and WT3W / HP1W, statistically significant increase and decrease in expression were observed in several genes as shown in FIGS. 21-1 to 21-7 and Table 6. . In Fgf23 mutant mice, differences in expression due to sex differences were also observed. From the above, it was shown that the genes whose expression is fluctuated in the high phosphorus state skin can be classified into four types of models depending on or independent of Fgf23 gene functional expression shown in FIG.
 具体的には、4タイプは以下の様に説明される。
モデルIのメカニズムにより発現が制御されている遺伝子群:
腎疾患及び/又は高リン状態により皮膚において発現が上昇する。これらの遺伝子の発現は、腎疾患及び/又は 高リン状態により骨におけるFGF23の発現上昇により抑制される経路と、FGF23の発現上昇に影響を受けない二つの経路がある。
Specifically, the four types are described as follows.
Genes whose expression is controlled by Model I mechanism:
Expression is elevated in the skin due to renal disease and / or hyperphosphatemia. There are two pathways in which the expression of these genes is suppressed by renal disease and / or hyperphosphorus condition due to the elevated expression of FGF23 in bone and the pathway not affected by the elevated expression of FGF23.
モデルIIのメカニズムにより発現が制御されている遺伝子群:
腎疾患及び/又は 高リン状態により皮膚において発現が抑制される。これらの遺伝子の発現抑制は、腎疾患及び/又は高リン状態により骨におけるFGF23の発現上昇により抑制される経路と、FGF23の発現上昇に影響を受けない二つの経路がある。
Genes whose expression is regulated by the Model II mechanism:
Renal disease and / or high phosphorus status suppresses expression in the skin. There are two pathways in which the suppression of the expression of these genes is suppressed by renal disease and / or hyperphosphorylation, which is suppressed by the increase in the expression of FGF23 in bone, and the two pathways which are not affected by the expression of FGF23.
モデルIIIのメカニズムにより発現が制御されている遺伝子群:
腎疾患及び/又は高リン状態により皮膚において発現が抑制される。これらの遺伝子の発現は、腎疾患及び/又は 高リン状態により骨におけるFGF23の発現上昇により抑制される。
Genes whose expression is controlled by the mechanism of Model III:
Renal disease and / or hyperphosphatemia suppresses expression in the skin. The expression of these genes is suppressed by the upregulation of FGF23 in bone due to renal disease and / or hyperphosphatemia.
モデルIVのメカニズムにより発現が制御されている遺伝子群:
腎疾患及び/又は高リン状態により皮膚において発現が抑制される。これらの遺伝子の発現は、腎疾患及び/又は高リン状態により骨におけるFGF23の発現上昇には影響されない。
Genes whose expression is controlled by the mechanism of Model IV:
Renal disease and / or hyperphosphatemia suppresses expression in the skin. The expression of these genes is not affected by the elevated expression of FGF23 in bone due to renal disease and / or hyperphosphate status.
Figure JPOXMLDOC01-appb-T000010
Figure JPOXMLDOC01-appb-T000010
1 演算装置1
2 演算装置2
3 演算装置3
4 スクリーニング装置4
5 スクリーニング装置5
6 演算装置6
11 測定値取得部
12 基準値取得部
13 測定値比較部
14 予測部
31 第1の測定値取得部
32 第2の測定値取得部
33 測定値比較部
34 候補物質決定部
1 Arithmetic unit 1
2 Arithmetic unit 2
3 Arithmetic unit 3
4 Screening device 4
5 Screening device 5
6 Arithmetic device 6
11 measurement value acquisition unit 12 reference value acquisition unit 13 measurement value comparison unit 14 prediction unit 31 first measurement value acquisition unit 32 second measurement value acquisition unit 33 measurement value comparison unit 34 candidate substance determination unit

Claims (63)

  1.  下記の手段を有する、被験体の腎疾患を予測する装置:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
     前記取得手段が取得した測定値に基づいて、前記腎疾患を予測する予測手段。
    Apparatus for predicting renal disease in a subject having the following means:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the renal disease based on the measurement value acquired by the acquisition means.
  2.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項1に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to claim 1, wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
  3.  前記予測手段は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、請求項1又は2に記載の装置。 The prediction means compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is out of the range of the reference value. The device described in 2.
  4.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
    には、前記腎疾患に、FGF23が関与していると決定する、請求項3に記載の装置。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value The device according to claim 3, wherein, if it is, it is determined that FGF23 is involved in the renal disease.
  5.  コンピュータに実行させたときに、被験体の腎疾患を予測するための下記の処理を当該コンピュータに実施させるプログラム:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
     前記取得処理で取得された測定値に基づいて、前記腎疾患を予測する予測処理。
    A program that, when executed on a computer, causes the computer to perform the following processing to predict renal disease in a subject:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring a measurement value of at least one type of mRNA selected from the group consisting of and prediction processing for predicting the renal disease based on the measurement value acquired in the acquisition processing.
  6.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項5に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to claim 5, which is at least one selected from the group consisting of, Col1a1 and Defb8.
  7.  前記予測処理では、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、請求項5又は6に記載のプログラム。 The prediction process compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is out of the range of the reference value. The program described in 6.
  8.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
    には、前記腎疾患に、FGF23が関与していると決定する、請求項7に記載のプログラム。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value The program according to claim 7, wherein, if it is, it is determined that FGF23 is involved in the renal disease.
  9.  下記の工程を有する、被験体の腎疾患を予測する方法:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
     前記取得工程で取得された測定値に基づいて、前記腎疾患を予測する予測工程。
    A method of predicting renal disease in a subject comprising the following steps:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining the measurement value of at least one type of mRNA selected from the group consisting of: and predicting the kidney disease based on the measurement value obtained in the acquisition step.
  10.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項9に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The method according to claim 9, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  11.  前記予測工程は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体は腎疾患であると決定する、請求項9又は10に記載の方法。 10. The method according to claim 9, wherein the predicting step compares the measured value with a predetermined reference value, and determines that the subject has a renal disease if the measured value is out of the range of the reference value. The method according to 10.
  12.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
    には、前記腎疾患に、FGF23が関与していると決定する、請求項11に記載の方法。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value The method according to claim 11, wherein if it is determined that FGF23 is involved in the renal disease.
  13.  下記の手段を有する、被験体におけるリン代謝異常を予測する装置:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
     前記取得手段が取得した測定値に基づいて、前記リン代謝異常を予測する予測手段。
    Apparatus for predicting abnormal phosphorus metabolism in a subject, having the following means:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting the phosphorus metabolism abnormality based on the measurement value acquired by the acquisition means.
  14. 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項13に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The device according to claim 13, wherein the device is at least one selected from the group consisting of: Col1a1, and Defb8.
  15.  前記予測手段は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、請求項13又は14に記載の装置。 The prediction means compares the measured value with a predetermined reference value, and when the measured value is out of the range of the reference value, predicts that the subject has a phosphorus metabolism abnormality. Or the apparatus as described in 14.
  16.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
    には、前記リン代謝異常に、FGF23が関与していると決定する、請求項15に記載の装置。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value The apparatus according to claim 15, wherein, if it is, it is determined that the phosphorus metabolism disorder is related to FGF23.
  17.  コンピュータに実行させたときに、被験体におけるリン代謝異常を予測するための下記の処理を当該コンピュータに実施させるプログラム:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
     前記取得処理で取得された測定値に基づいて、前記リン代謝異常を予測する予測処理。
    A program that, when run on a computer, causes the computer to perform the following processing for predicting phosphorus metabolism abnormality in a subject:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition processing for acquiring a measurement value of at least one type of mRNA selected from the group consisting of and prediction processing for predicting the phosphorus metabolism abnormality based on the measurement value acquired in the acquisition processing.
  18. 前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項17に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to claim 17, which is at least one selected from the group consisting of, Col1a1 and Defb8.
  19.  前記予測処理では、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、請求項17又は18に記載のプログラム。 The prediction processing compares the measured value with a predetermined reference value, and when the measured value is out of the range of the reference value, predicts that the subject has a phosphorus metabolism abnormality. Or the program described in 18.
  20.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合
    には、前記リン代謝異常に、FGF23が関与していると決定する、請求項19に記載のプログラム。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value 20. The program according to claim 19, wherein, if it is, it is determined that FGF23 is involved in the phosphorus metabolism disorder.
  21.  下記の工程を有する、被験体におけるリン代謝異常を予測する方法:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
     前記取得工程で取得された測定値に基づいて、前記リン代謝異常を予測する予測工程。
    A method of predicting abnormal phosphorus metabolism in a subject comprising the following steps:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining the measured value of at least one type of mRNA selected from the group consisting of: and predicting the phosphorus metabolism abnormality based on the measured value obtained in the obtaining step.
  22.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項21に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 22. The method according to claim 21, which is at least one selected from the group consisting of, Col1a1 and Defb8.
  23.  前記予測工程は、前記測定値と所定の基準値とを比較し、該測定値が前記基準値の範囲外である場合には、前記被験体にリン代謝異常があると予測する、請求項21又は22に記載の方法。 22. The method according to claim 21, wherein the prediction step compares the measured value with a predetermined reference value, and predicts that the subject has a phosphorus metabolism abnormality if the measured value is out of the range of the reference value. Or the method according to 22.
  24.  さらに、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲内である場合には、前記リン代謝異常に、FGF23が関与していると決定する、請求項23に記載の方法。
    Furthermore, Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I122ra2, Rbfox1, Lrrc2, Col15a1, Sparc, Col11a1, Clec11a, Serpinb7, When the measured value of one type of biomarker is out of the range of the reference value, and / or the measured value of at least one biomarker selected from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 is in the range of the reference value The method according to claim 23, wherein, if it is, it is determined that the phosphorus metabolism disorder involves FGF23.
  25.  下記の手段を有する、被験体におけるFGF23の活性化を予測する装置:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得手段、並びに
     前記取得手段が取得した測定値に基づいて、前記FGF23の活性化を予測する予測手段。
    Device for predicting FGF23 activation in a subject, having the following means:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Acquisition means for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: prediction means for predicting activation of the FGF23 based on the measurement value acquired by the acquisition means.
  26.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項25に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to claim 25, which is at least one selected from the group consisting of, Col1a1 and Defb8.
  27.  前記予測手段は、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、請求項25又は26に記載の装置。
    The prediction means compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, When the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 26. The method according to claim 25, wherein FGF23 is predicted to be activated in said subject if at least one biomarker selected is within the range of the reference value. Is the device described in 26.
  28.  コンピュータに実行させたときに、被験体におけるFGF23の活性化を予測するための下記の処理を当該コンピュータに実施させるプログラム:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得処理、並びに
     前記取得処理で取得された測定値に基づいて、前記FGF23の活性化を予測する予測処理。
    A program that, when run on a computer, causes the computer to perform the following processing to predict FGF23 activation in a subject:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject An acquisition process for acquiring a measurement value of at least one type of mRNA selected from the group consisting of: and a prediction process for predicting activation of the FGF23 based on the measurement value acquired in the acquisition process.
  29.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項28に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The program according to claim 28, wherein the program is at least one selected from the group consisting of: Col1a1 and Defb8.
  30.  前記予測処理では、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、請求項28又は29に記載のプログラム。
    In the prediction processing, the measured value is compared with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, When the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 28. The method according to claim 28, wherein FGF23 is predicted to be activated in said subject if at least one biomarker selected is within the range of reference values. Or the program described in 29.
  31.  下記の工程を有する、被験体におけるFGF23の活性化を予測する方法:
     前記被験体から採取された皮膚由来検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のタンパク質の測定値、及び/又は、前記被験体から採取された検体中に含まれるバイオマーカーからなる群から選択される少なくとも一種のmRNAの測定値を取得する取得工程、並びに
     前記取得工程で取得された測定値に基づいて、前記FGF23の活性化を予測する予測工程。
    A method of predicting FGF23 activation in a subject comprising the steps of:
    From measured values of at least one protein selected from the group consisting of biomarkers contained in skin-derived samples collected from the subject and / or from biomarkers contained in samples collected from the subject Obtaining a measurement value of at least one type of mRNA selected from the group consisting of: and predicting the activation of the FGF23 based on the measurement value obtained in the acquisition step.
  32.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項31に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The method according to claim 31, wherein the method is at least one selected from the group consisting of: Col1a1 and Defb8.
  33.  前記予測工程は、前記測定値と所定の基準値とを比較し、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Sparc、Col11a1、Clec11a、Serpinb6d、及びDefb8よりなる群から選択される少なくとも一種のバイオマーカーの測定値が基準値の範囲外である場合、及び/又は
     Aldh1l2、Col5a1、Col3a1、C1qtnf6、及びCol1a1よりなる群から選択される少なくとも一種のバイオマーカーが基準値の範囲内である場合には、前記被験体においてFGF23が活性化していると予測する、請求項31又は32に記載の方法。
    The prediction step compares the measured value with a predetermined reference value, and Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, Il22ra2, Rbfox1, Lrrc2, Col15a1 Sparc, When the measured value of at least one biomarker selected from the group consisting of Col11a1, Clec11a, Serpinb6d, and Defb8 is out of the range of the reference value, and / or from the group consisting of Aldh11, Col5a1, Col3a1, C1qtnf6, and Col1a1 34. The method according to claim 31, wherein FGF23 is predicted to be activated in said subject if at least one biomarker selected is within the range of reference values. The method described in 32.
  34.  下記の手段を有する、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング装置:
     被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び
     前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
    Screening apparatus for candidate active ingredients for suppressing the functional expression of FGF23, having the following means:
    Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Means for acquiring the measurement value of the biomarker protein of at least one test substance-treated sample and / or the measurement value of mRNA of the protein, and the measurement acquired by the first measurement value acquisition means A determining means for determining that the test substance is a candidate substance of the active ingredient based on a value.
  35.  被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
     被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較手段、
    をさらに有し、
     前記決定手段は、前記測定値比較手段の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、請求項34に記載の装置。
    From a group consisting of a sample collected from the skin of a subject (except for human beings) not treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Second measurement value acquiring means for acquiring the measurement value of the corresponding biomarker protein in at least one untreated sample to be selected and / or the measurement value of mRNA of said protein, and the measurement value of the test substance-treated sample and Measured value comparison means for comparing the measured values of an untreated sample,
    And have
    The apparatus according to claim 34, wherein the determination means determines that the test substance is a candidate substance of the active ingredient based on the comparison result of the measurement value comparison means.
  36.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項35に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The device according to claim 35, wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
  37.  コンピュータに実行させたときに、FGF23の機能発現を抑制するための有効成分の候補物質をスクリーニングするための下記の処理を当該コンピュータに実施させるスクリーニングプログラム:
     被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一つの被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び
     前記第1の測定値取得処理で取得された測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定処理。
    A screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for suppressing the functional expression of FGF23:
    Selected from the group consisting of a sample collected from the skin of a subject (except for humans) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A first measurement value acquisition process for acquiring a measurement value of a biomarker protein in at least one test substance-treated sample and / or a measurement value of mRNA of the protein, and the first measurement value acquisition process A determination process of determining that the test substance is a candidate substance of an active ingredient based on measured values.
  38.  さらに、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
     被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較処理、
    を前記コンピュータに実施させ、
     前記決定処理では、前記測定値比較処理の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、請求項37に記載のプログラム。
    Furthermore, it comprises a sample collected from the skin of a subject not treated with the test substance (except for human beings), a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin A second measurement value acquisition process for acquiring a measurement value of a corresponding biomarker protein of at least one untreated sample selected from a group and / or a measurement value of mRNA of the protein, and the measurement value of the test substance-treated sample And a measured value comparison process comparing the measured values of the untreated sample,
    On the computer,
    The program according to claim 37, wherein in the determination process, the test substance is determined to be a candidate substance of the active ingredient based on a comparison result of the measurement value comparison process.
  39.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項37又は38に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to claim 37 or 38, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  40.  以下の工程を含む、FGF23の機能発現を抑制するための有効成分の候補物質のスクリーニング方法:
     (I)被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
     (II)前記工程(I)で得られた測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程。
    The screening method of the candidate substance of the active ingredient for suppressing the functional expression of FGF23 including the following processes:
    (I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein,
    (II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I).
  41.  前記工程(I)と(II)の間に、前記工程(I)で取得された被験物質処理検体の測定値と、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程をさらに含み、
     工程(II)が、前記比較結果に基づいて、前記被験物質が有効成分の候補物質であると決定する工程である、
    をさらに含む、請求項40に記載の方法。
    Between the steps (I) and (II), the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with
    Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
    41. The method of claim 40, further comprising
  42.  前記工程(I)の前に、
    (i)被験体(ヒトを除く)、皮膚に由来する被験組織又は被験細胞を、被験物質で処理する工程、
    (ii)前記工程(i)において被験物質で処理された前記被験体、被験組織又は被験細胞から検体を採取する工程、及び
    (iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程
    を含む、請求項40又は41に記載の方法。
    Before the step (I),
    (I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance,
    (Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in step (i), and (iii) proteins and / or proteins from the sample obtained in step (ii) 42. The method of claim 40 or 41, comprising the step of recovering mRNA.
  43.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項40~42のいずれか一項に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The method according to any one of claims 40 to 42, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  44.  下記の手段を有する、FGF23の機能を活性化するための有効成分の候補物質のスクリーニング装置:
     被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(被験物質処理検体)中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び
     前記第1の測定値取得手段が取得した測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定手段。
    Screening apparatus for candidate active ingredients for activating the function of FGF23, having the following means:
    Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample) The test substance is an active ingredient based on a first measurement value acquisition unit that acquires a measurement value of a protein and / or a measurement value of mRNA of the protein, and the measurement value acquired by the first measurement value acquisition unit. Determining means to determine a candidate substance.
  45.  被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(未処理検体)のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
     被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較手段、
    をさらに有し、
     前記決定手段は、前記測定値比較手段の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、請求項44に記載のスクリーニング装置。
    Biomarker protein of a sample collected from the skin of a subject (except for human beings) not treated with the test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (untreated sample) A second measured value acquiring means for acquiring a measured value of the protein and / or a measured value of mRNA of the protein, and a measured value comparing means for comparing the measured value of the test substance-treated sample with the measured value of the untreated sample;
    And have
    45. The screening device according to claim 44, wherein the determination means determines that the test substance is a candidate substance of the active ingredient based on the comparison result of the measurement value comparison means.
  46.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項45に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The device according to claim 45, wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
  47.  コンピュータに実行させたときに、FGF23の機能を活性化するための有効成分の候補物質をスクリーニングするための下記の処理を当該コンピュータに実施させるスクリーニングプログラム:
     被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(被験物質処理検体)中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び
     前記第1の測定値取得処理で取得された測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する決定処理。
    A screening program which, when executed on a computer, causes the computer to carry out the following processing for screening active substance candidate substances for activating the function of FGF23:
    Biomarker in a sample collected from the skin of a subject (except for human beings) treated with a test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (test substance treated sample) The test substance is an active ingredient based on a first measurement value acquisition process for acquiring a measurement value of a protein and / or a measurement value of mRNA of the protein, and a measurement value acquired in the first measurement value acquisition process. Decision processing to determine that it is a candidate substance of
  48.  さらに、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、又は皮膚に由来する被験組織若しくは皮膚に由来する被験細胞から採取された検体(未処理検体)のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
     被験物質処理検体の前記測定値及び未処理検体の前記測定値を比較する測定値比較処理、
    を前記コンピュータに実施させ、
     前記決定処理では、前記測定値比較処理の比較結果に基づいて前記被験物質が有効成分の候補物質であると決定する、請求項47に記載のプログラム。
    Furthermore, the bio of a sample collected from the skin of a subject (except for human beings) not treated with the test substance, or a sample collected from a test tissue derived from the skin or a test cell derived from the skin (untreated sample) A second measurement value acquisition process for acquiring a measurement value of a marker protein and / or a measurement value of mRNA of the protein, and a measurement value comparison process for comparing the measurement value of a test substance-treated sample with the measurement value of an untreated sample ,
    On the computer,
    The program according to claim 47, wherein in the determination process, the test substance is determined to be a candidate substance of the active ingredient based on a comparison result of the measurement value comparison process.
  49.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項47又は48に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, Bic7 The program according to claim 47 or 48, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  50.  以下の工程を含む、FGF23の機能を活性化するための有効成分の候補物質のスクリーニング方法:
     (I)被験物質で処理された被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の被験物質処理検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
     (II)前記工程(I)で得られた測定値に基づいて、前記被験物質が有効成分の候補物質であると決定する工程。
    The screening method of the active substance candidate substance for activating the function of FGF23 including the following processes:
    (I) A sample collected from the skin of a subject (except for human beings) treated with a test substance, a sample collected from a test tissue derived from the skin, and a sample collected from a test cell derived from the skin Obtaining a measured value of a biomarker protein in at least one test substance-treated sample selected from the group and / or a measured value of mRNA of said protein,
    (II) A step of determining that the test substance is a candidate substance of the active ingredient based on the measurement value obtained in the step (I).
  51.  前記工程(I)と(II)の間に、前記工程(I)で取得された被験物質処理検体の測定値と、被験物質で処理されていない被験体(ヒトを除く)の皮膚から採取された検体、皮膚に由来する被験組織から採取された検体、及び皮膚に由来する被験細胞から採取された検体よりなる群から選択される少なくとも一種の未処理検体中の対応するバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程をさらに含み、
     工程(II)が、前記比較結果に基づいて、前記被験物質が有効成分の候補物質であると決定する工程である、
    請求項50に記載の方法。
    Between the steps (I) and (II), the measured value of the test substance-treated sample obtained in the step (I) and the skin of a subject (except for human beings) not treated with the test substance Of the corresponding biomarker protein in at least one untreated sample selected from the group consisting of a selected sample, a sample collected from a test tissue derived from skin, and a sample collected from a test cell derived from skin And / or comparing the measured value of mRNA of said protein with
    Step (II) is a step of determining that the test substance is a candidate substance of the active ingredient based on the comparison result.
    51. The method of claim 50.
  52.  前記工程(I)の前に、
    (i)被験体(ヒトを除く)、皮膚に由来する被験組織又は被験細胞を、被験物質で処理する工程、
    (ii)前記工程(i)において被験物質で処理された前記被験体、被験組織又は被験細胞から検体を採取する工程、及び
    (iii)前記工程(ii)で得られた検体からタンパク質及び/又はmRNAを回収する工程
    を含む、請求項50又は51に記載の方法。
    Before the step (I),
    (I) treating a subject (excluding human), a test tissue or test cell derived from skin with a test substance,
    (Ii) collecting a sample from the subject, test tissue or test cells treated with the test substance in the step (i), and (iii) proteins and / or proteins from the sample obtained in the step (ii) 52. The method of claim 50 or 51, comprising the step of recovering mRNA.
  53.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項40~42のいずれか一項に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The method according to any one of claims 40 to 42, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  54.  下記の手段を有する、疾患におけるFGF23の関与を予測する予測装置:
     疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得手段、及び 前記第1の測定値取得手段が取得した測定値に基づいて、前記疾患にFGF23が関与していると決定する決定手段。
    Predictor for predicting the involvement of FGF23 in disease comprising the following means:
    First measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition means A determination means for determining that FGF23 is involved in the disease based on the measurement value obtained by
  55.  前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得手段、及び
     前記第1の測定値取得手段が取得した測定値及び前記第2の測定値取得手段が取得した測定値を比較する測定値比較手段、
    をさらに有し、
     前記決定手段は、前記比較の結果に基づいて前記疾患にFGF23が関与していると決定する、請求項54に記載の装置。
    A second measurement value acquiring means for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein, and the first Measurement value comparison means for comparing the measurement value acquired by the measurement value acquisition means with the measurement value acquired by the second measurement value acquisition means;
    And have
    55. The apparatus according to claim 54, wherein the determination means determines that FGF23 is involved in the disease based on the result of the comparison.
  56.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項55に記載の装置。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 56. The device according to claim 55, wherein the device is at least one selected from the group consisting of: Col1a1 and Defb8.
  57.  コンピュータに実行させたときに、疾患におけるFGF23の関与を予測するための下記の処理を当該コンピュータに実施させる予測プログラム:
     疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第1の測定値取得処理、及び 前記第1の測定値取得処理で取得された測定値に基づいて、前記疾患にFGF23が関与していると決定する決定処理。
    A prediction program which, when executed on a computer, causes the computer to carry out the following processing for predicting the involvement of FGF23 in a disease:
    A first measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measurement value of mRNA of the protein, and the first measurement value acquisition process The decision processing which determines that FGF23 is concerned in the said disease based on the measured value acquired by b.
  58.  さらに、前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する第2の測定値取得処理、及び
     第1の測定値取得処理で取得された測定値及び第2の測定値取得処理で取得された測定値を比較する測定値比較処理、
    を前記コンピュータに実施させ、
     前記決定処理では、前記比較の結果に基づいて前記疾患にFGF23が関与していると決定する、請求項57に記載のプログラム。
    Furthermore, a second measurement value acquisition process for acquiring a measurement value of a biomarker protein in a sample collected from the skin of a subject not having the disease and / or a measurement value of mRNA of the protein, A measurement value comparison process of comparing the measurement value acquired in the measurement value acquisition process of the first measurement value and the measurement value acquired in the second measurement value acquisition process;
    On the computer,
    58. The program according to claim 57, wherein said determination processing determines that FGF23 is involved in said disease based on the result of said comparison.
  59.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項57又は58に記載のプログラム。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The program according to claim 57 or 58, wherein the program is at least one selected from the group consisting of: Col1a1 and Defb8.
  60.  以下の工程を含む、疾患におけるFGF23の関与を予測する予測方法:
     (I)疾患を有する被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値を取得する工程、
     (II)前記工程(I)で得られた測定値に基づいて、前記疾患にFGF23が関与していると決定する工程。
    Methods of predicting FGF23's involvement in disease comprising the following steps:
    (I) obtaining a measured value of a biomarker protein in a sample collected from the skin of a subject having a disease and / or a measured value of mRNA of said protein,
    (II) A step of determining that FGF23 is involved in the disease based on the measurement value obtained in the step (I).
  61.  前記工程(I)と(II)の間に、前記工程(I)で取得された測定値と、前記疾患を有していない被験体の皮膚から採取された検体中のバイオマーカータンパク質の測定値及び/又は前記タンパク質のmRNAの測定値とを比較する工程
    をさらに含み、
     工程(II)が、前記比較の結果に基づいて、前記疾患にFGF23が関与していると決定する工程である、
    請求項60に記載の方法。
    Between the steps (I) and (II), the measured value obtained in the step (I) and the measured value of the biomarker protein in a sample collected from the skin of a subject not having the disease And / or comparing the measured value of mRNA of said protein with
    Step (II) is a step of determining that FGF23 is involved in the disease based on the result of the comparison.
    61. The method of claim 60.
  62.  前記バイオマーカーが、Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種である、請求項60~61のいずれか一項に記載の方法。 The biomarkers include Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4dip, I22ra2, Rbfox1, Ldrc2, Aldhl2, Col5a11, Col5a11, Col5a11, C5a11, C5a11, C5a1 The method according to any one of claims 60 to 61, which is at least one selected from the group consisting of: Col1a1 and Defb8.
  63.  Hamp2、Actn3、Asb11、Cxcl13、Ptp4a3、Klhl38、Serpinb6e、Atp2a1、Myot、Nrap、Pde4dip、Il22ra2、Rbfox1、Lrrc2、Col15a1、Aldh1l2、Col5a1、C1qtnf6、Sparc、Col11a1、Clec11a、Col3a1、Serpinb6d、Col1a1、及びDefb8よりなる群から選択される少なくとも一種を含む、腎機能、リン代謝異常、又はFGF23の関与を予測するための皮膚のバイオマーカー。 Hamp2, Actn3, Asb11, Cxcl13, Ptp4a3, Klhl38, Serpinb6e, Atp2a1, Myot, Nrap, Pde4 dip, I22ra2, Rbfox1, Lrcc2, Col1a1, Col5a1, C1qtnChIh, a C1qtnChIh, a C1qtnCh, a backgrip A skin biomarker for predicting involvement of renal function, abnormal phosphorus metabolism, or FGF23, comprising at least one selected from the group consisting of
PCT/JP2018/028702 2017-08-01 2018-07-31 Biomarkers from skin WO2019026918A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2019534538A JP7445288B2 (en) 2017-08-01 2018-07-31 skin biomarkers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2017-149487 2017-08-01
JP2017149487 2017-08-01

Publications (1)

Publication Number Publication Date
WO2019026918A1 true WO2019026918A1 (en) 2019-02-07

Family

ID=65232818

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/028702 WO2019026918A1 (en) 2017-08-01 2018-07-31 Biomarkers from skin

Country Status (2)

Country Link
JP (1) JP7445288B2 (en)
WO (1) WO2019026918A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003057733A1 (en) * 2001-12-28 2003-07-17 Kirin Beer Kabushiki Kaisha Antibodies against fibroblast growth factor 23
JP2013174608A (en) * 2005-08-23 2013-09-05 Fancl Corp Skin aging marker and technology for using the same
WO2017130985A1 (en) * 2016-01-29 2017-08-03 株式会社国際電気通信基礎技術研究所 Renal function evaluating device, device for predicting onset of kidney disease complications, and phosphorous ingestion amount estimating device
WO2017170610A1 (en) * 2016-03-29 2017-10-05 株式会社国際電気通信基礎技術研究所 Screening method for candidate substances for active component to prevent or treat at least one disease selected from the group consisting of renal hypofunction, chronic kidney disease and kidney failure

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003057733A1 (en) * 2001-12-28 2003-07-17 Kirin Beer Kabushiki Kaisha Antibodies against fibroblast growth factor 23
JP2013174608A (en) * 2005-08-23 2013-09-05 Fancl Corp Skin aging marker and technology for using the same
WO2017130985A1 (en) * 2016-01-29 2017-08-03 株式会社国際電気通信基礎技術研究所 Renal function evaluating device, device for predicting onset of kidney disease complications, and phosphorous ingestion amount estimating device
WO2017170610A1 (en) * 2016-03-29 2017-10-05 株式会社国際電気通信基礎技術研究所 Screening method for candidate substances for active component to prevent or treat at least one disease selected from the group consisting of renal hypofunction, chronic kidney disease and kidney failure

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KOUNO, KEIJI ET AL.: "Cross- talk with CKD and abnormal bone metabolism", THE MEDICAL FRONTLINE, vol. 71, no. 12, December 2016 (2016-12-01), pages 62 - 68 *
UGARTE, F. ET AL.: "Impaired phosphorylation of JAK2-STAT5b singnaling in fibroblasts from uremic children", PEDIATRIC NEPHROLOGY, vol. 31, no. 6, June 2016 (2016-06-01), pages 965 - 974, XP035670984 *

Also Published As

Publication number Publication date
JPWO2019026918A1 (en) 2020-06-11
JP7445288B2 (en) 2024-03-07

Similar Documents

Publication Publication Date Title
JP6432962B2 (en) Method for screening candidate substance of active ingredient for preventing or treating at least one disease selected from the group consisting of decreased renal function, chronic kidney disease and renal failure
CN102713632B (en) For the diagnosis and treatment of composition and the method for dog kidney condition
BR112013000745B1 (en) in vitro method to diagnose or detect colorectal cancer in an individual
Goulielmos et al. Endometriosis research in the-omics era
JP2023118802A (en) Non-invasive diagnostic of non-alcoholic fatty liver diseases, non-alcoholic steatohepatitis and/or liver fibrosis
CN111065748A (en) Non-invasive diagnosis of non-alcoholic steatoliver disease, non-alcoholic steatohepatitis and/or liver fibrosis
EP3497451A1 (en) Histones and/or proadm as markers indicating an adverse event
CN103384832A (en) Compositions and methods for diagnosing and treating kidney disorders in a feline
JP6414869B2 (en) Device for evaluating renal function, device for predicting the onset of complications of kidney disease, and device for estimating phosphorus intake
CN107208159A (en) Host DNA as Crohn&#39;s disease biomarker
CN101688247A (en) methods and compositions for diagnosing osteoarthritis in a feline
EP3334498B1 (en) Methods of treating volume depletion and kidney injury
JP7445288B2 (en) skin biomarkers
EP3203240B1 (en) Method for determining acute myocardial infarction
JP6829888B2 (en) Non-alcoholic steatohepatitis diagnostic kit and test method
JP2018105726A (en) Apparatus, program, and method for screening for active ingredient candidate substances for prevention or treatment of one disorder selected from group consisting of myocardial infarction, dementia, and tumors
US9638699B2 (en) Biomarkers of oxidative stress
US11180539B2 (en) Pharmaceutical composition or food composition, and method for assessing effect of active ingredient in vivo
Kato et al. Smad1 as a biomarker for diabetic nephropathy
JPWO2004040301A1 (en) Diagnosis method for type 2 diabetes
JP2021051085A (en) Apparatus, program, and method for screening for active ingredient candidate substances for prevention or treatment of one disorder selected from group consisting of myocardial infarction, dementia, and tumors
JP2021047198A (en) Apparatus, program, and method for screening for active ingredient candidate substances for prevention or treatment of one disorder selected from group consisting of myocardial infarction, dementia, and tumors
WO2023107473A1 (en) Identifying monoclonal gammopathies in a high-risk population for hematological malignancies
KR20210099858A (en) Biomarker for diagnosing pulmonary edema and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18842389

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019534538

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18842389

Country of ref document: EP

Kind code of ref document: A1