US20030017619A1 - Test method for IgA nephropathy - Google Patents

Test method for IgA nephropathy Download PDF

Info

Publication number
US20030017619A1
US20030017619A1 US10/117,798 US11779802A US2003017619A1 US 20030017619 A1 US20030017619 A1 US 20030017619A1 US 11779802 A US11779802 A US 11779802A US 2003017619 A1 US2003017619 A1 US 2003017619A1
Authority
US
United States
Prior art keywords
iga1
hinge region
iga nephropathy
core peptide
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/117,798
Inventor
Tohru Rokubo
Kenji Arai
Kazunori Toma
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Asahi Kasei Corp
Original Assignee
Asahi Kasei Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Asahi Kasei Corp filed Critical Asahi Kasei Corp
Priority to US10/117,798 priority Critical patent/US20030017619A1/en
Publication of US20030017619A1 publication Critical patent/US20030017619A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/811Test for named disease, body condition or organ function

Definitions

  • This invention relates to a novel test method for IgA nephropathy. More particularly, the present invention pertains to a rapid and simple test method for IgA nephropathy and a determination method of antibody, which has low risks in emotional distress, peripheral hemorrhage in the kidney and financial burden to the patient, by determining antibody recognizing IgA1 hinge region core peptide in the specimen.
  • IgA (immunoglobulin A) nephropathy is a disease concept provided by Berger et al. (J. Urol., 74, pp. 694-695, 1968), and is a primary glomerular nephritis having features with clinically poor symptoms except for continuous proteinuria and hematuria, and histological features with precipitants consisting of mainly IgA in the mesangium.
  • the incidence of IgA nephropathy in Japan is high and accounts for 30% of the chronic nephritis.
  • the long term prognosis is not so favorable, and 10-15% of patients with 10 years progress and about 3 0% of patients with 20 years progress suffer from terminal renal failure. Consequently, IgA nephropathy is especially noticed as a causal disease for terminal renal failure.
  • the only known test method for IgA nephropathy is the renal biopsy.
  • This test method causes emotional distress for the patients, and her may cause peripheral hemorrhage in the kidney after the biopsy.
  • the patients must have absolute rest for more than 24 hours after the renal biopsy, and this requires the patients to stay in hospital for several days, and this causes a heavy financial burden for them.
  • the test method has disadvantages including requiring many kinds of test facilities together with long term testing time.
  • An object of the present invention is to provide a rapid and simple test method for IgA nephropathy.
  • Such a test method requires no renal biopsy, which has been essential in the conventional test method for IgA nephropathy.
  • the test method is comprised of detecting antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens. As a result, the patients' emotional distress, risk of peripheral hemorrhage of the kidney and the financial burden can be reduced.
  • the present invention relates to a test method for IgA nephropathy by detecting antibody recognizing the core peptide of the hinge region in IgA1 in specimens.
  • a test method comprising detecting antibody recognizing the core peptide of the hinge acid sequence consisting of region having at least a part of amino acid sequence consisting of Pro Val Pro Ser Thr Pro Pro Thr Pro (SEQ ID NO:1) Ser Pro Ser Thr Pro Pro Pro Thr Pro Ser Pro Ser Cys
  • the test method comprises using an immobilized solid phase core peptide of the hinge region, said immobilized solid phase of which is microtiter plate or latex particles.
  • Examples of detection methods for the antibody of the present invention are test method for IgA nephropathy which includes enzyme immunoassay, latex agglutination photometric immunoassay, nephelometric immunoassay, latex slide agglutination test, turbidimetric immunoassay, luminescent immunoassay, fluorescence polarization immunoassay, fluorescence immunoassay and radioimmunoassay.
  • FIG. 1 illustrates a schematic drawing showing the outline of a molecular structure of IgA1.
  • IgA which may be involved in a cause for IgA nephropathy, is a type of immunoglobulin.
  • the IgA molecule consists of two heavy chains and two light chains, and is classified into subtypes of IgA1 and IgA2 depending on the difference in heavy chain structures.
  • IgA nephropathy since the deposited IgA in the glomeruli is mainly IgA1 subtype (Conley et al., J. Clin. Invest. 66, pp. 1432-1436, 1980), the properties of IgA1 between those of patients with IgA nephropathy and those of healthy subjects or patients with non-IgA nepliropathy may differ from one another.
  • IgA1 and IgA2 The largest structural difference between IgA1 and IgA2 exists in the hinge region of the heavy chain.
  • the hinge region of IgA2 is deficient in 13 amino acids (Frangione et al., Proc. Natl. Acad. Sci. USA, 69, pp. 3673-3676, 1972) as compared with the hinge region of IgA1 (SEQ ID NO: 13) [Basic immunology, Upper Vol., pp. 158-162, (1986), The University Tokyo Press].
  • the core peptide of the hinge region has structural similarity with mucin insofar as containing large amounts of proline, serine and threonine.
  • a rapid test for IgA nephropathy can be performed by determining antibody, which recognizes the core peptide of the hinge region of IgA1, in specimens such as body fluid of the patients.
  • the present invention relates to a test method for IgA nephropathy based on determining antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens.
  • the hinge region of IgA1 indicates the region between CH1 domain and CH2 domain in the heavy chains, which construct IgA1 molecule, and may optionally include a structure having added thereto a sequence of Pro-Va1- in the N-terminal region.
  • This region contains disulfide bond between heavy chains, and is rich in proline, serine and threonine.
  • the core peptide of the hinge region in IgA1 as an antigen can include the peptide constructing the hinge region in IgA1 or a partial structure thereof.
  • peptides having partial structure thereof consisting of at least three amino acids thereof are preferably used.
  • peptides are peptides comprising amino acid sequences of the formulas: Val Pro Ser, Ser Thr Pro, Thr Pro Pro, Pro Pro Thr, Pro Thr Pro, Thr Pro Ser, Pro Ser Pro, Ser Pro Ser or Pro Ser Thr.
  • peptides consisting of at least five amino acids, for example, formulas, ProValProSerThr (SEQ ID NO: 2), ProSerThrProPro (SEQ ID NO: 3), ThrProProThrPro (SEQ ID NO: 4), ProThrProSerPro (SEQ ID NO: 5), ProSerProSerThr (SEQ ID NO: 6) or ProSerProSerCys (SEQ ID NO: 7)
  • the core peptide of the hinge region in IgA1 modified by sugars, lipids, functional groups, protective groups or cross linking agents, and bound to carrier such as proteins, plastics or latex particles can be used.
  • any core peptide of the hinge region in IgA1, i.e. antigen, produced by any method, for example, a product by synthesis, a product derived from IgA1 originated from patients or subjects with non-IgA nephropathy, or a product by gene recombinant technology, can be used.
  • specimens in the present invention are biological fluid such as blood, plasma, serum, saliva or urine, biological tissue extracts and supernatant solutions of the tissue culture.
  • Specimens can be used without dilution or with aliquot dilution of buffer solution depending on the principle of detection method, detection sensitivity, detection range and other factors in the present invention.
  • Specimens can contain anticoagulants such as sodium EDTA, sodium citrate or heparin sodium, materials for stabilizing the specimen, or other substances.
  • anticoagulants such as sodium EDTA, sodium citrate or heparin sodium, materials for stabilizing the specimen, or other substances.
  • buffers which are customarily used in immunological determination methods and preferably have a pH value in the range of from 5 to 9 can be employed.
  • phosphate buffer borate buffer, carbonate buffer, Tris-HCl buffer, veronal buffer and Good's buffer such as HEPES buffer, PIPES buffer and MES buffer, can be used.
  • any type of additives for example, bovine serum albumin (BSA), protein such as gelatin or skimmed milk, surface active agents, sugars, chelating agents, reducing agents or salt can be added in the buffer solution.
  • BSA bovine serum albumin
  • the concentration of the additives can be selected from the concentration used in the immunological determination method of antibody, which is applied in known antigen-antibody reactions.
  • Examples of antibody to be determined in the present invention are all antibodies that react with the core peptide of the hinge region in IgA1 in specimens, and are preferably IgG class, IgM class or IgA class.
  • a method for immunologically determining the antibody, which recognizes the core peptide of the hinge region in IgA1 as the antigen, in specimens is characterized by reacting the above described core peptide of the hinge region in IgA1 with the specimen under the condition to form antigen-antibody complex and qualitating or quantitating the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen based on assaying the thus generated product of the antigen-antibody reaction.
  • EIA enzyme immunoassay
  • a carrier having immobilized thereon core peptide of the hinge region in IgA1 and a specimen are reacted, and a labeled antibody (a complex of anti-human IgG antibody, anti-human IgM antibody, anti-human IgA antibody or fragments thereof having binding activity to antigen and labeled enzyme) is reacted therewith, then the coloring agent (substrate) for developing color by applying the reaction with labeled enzyme bound with the carrier through the antigen-antibody complex, is added and subjected to reaction.
  • a labeled antibody a complex of anti-human IgG antibody, anti-human IgM antibody, anti-human IgA antibody or fragments thereof having binding activity to antigen and labeled enzyme
  • reaction is terminated, if necessary, by adding enzyme reaction terminator, and optical absorption of the thus generated coloration is measured at a suitable wavelength to quantitatively or qualitatively assay the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen.
  • a complex bound with the synthetic core peptide of the hinge region in IgA1 consisting of peptide of at least three amino acids, preferably at least five amino acids, selected from the amino acid sequence of the core peptide of the hinge region in IgA1 such as amino acid sequence of the formula, Pro Val Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Cys
  • bovine serum albumin at a binding ratio of 1:1-15:1 (peptide: BSA), preferably, 1:1-10:1 (peptide: BSA)
  • BSA bovine serum albumin
  • the antigen adjusted within a concentration range generally from 0.01 ⁇ g/ml-100 ⁇ g/ml is added to the plastic microtiter plate, generally at 25 ⁇ l -200 ⁇ l/well, allowed to stand at 4° C.-40° C., for 10 min.-48 hours and is washed with suitable buffer to immobilize the antigen.
  • specimens such as serum of the patients
  • suitable buffer generally 25 ⁇ l-200 ⁇ l
  • the self-known labeled antibody (a complex of anti-human IgG antibody, anti-human IgM antibody, anti-human IgA antibody or fragments thereof having binding activity to antigen, and the labeling enzyme, for example generally used in the enzyme immunoassay, such as peroxidase, alkaline phosphatase, ⁇ -galactosidase or glucose oxidase) diluted with suitable buffer, generally 25 ⁇ l-200 ⁇ l, is added and allowed to react generally at 4° C.-40° C., for 10 min.-48 hours then are washed with suitable buffer.
  • suitable buffer generally 25 ⁇ l-200 ⁇ l
  • coloring agent (substrate) for developing color with a reaction of the labeled enzyme is added generally 25 ⁇ l-200 ⁇ l, and is reacted generally within a range at 4° C. -40° C., for 1 min.-48 hours.
  • reaction is terminated, if necessary, by adding enzyme reaction terminator.
  • Optical absorb any of the supernatant of the enzymatic reaction mixture is colorimetrically measured and the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen is qualitatively or quantitatively measured.
  • orthophenylenediamine and the like can be used as a substrate, and the resultant coloring of the enzyme reaction is measured by absorbancy at 490 nm.
  • paranitrophenyl phosphate can be used as a substrate, and the resultant color development of the enzyme reaction is measured by absorbancy at 405 nm.
  • the antibody titer which recognizes the core peptide of the hinge region in IgA1 (IgG class antibody titer, IgM class antibody titer or IgA class antibody titer), is high, the above absorbancy values are also high.
  • the examination for IgA nephropathy can be performed as follows: the absorbancy of the specimens obtained from the healthy subjects or patients with non-IgA nephropathy, or the concentration values in these specimens, which are quantitatively determined by comparing the obtained absorbancy with the calibration curve calculated from the standard substance, are obtained. Measurement above the average of these values +2 ⁇ standard deviation are deemed to be positive, and those below or not above the value thereof are considered to be negative. Using these criteria, the examination of subjects, whether or not the patient is suffering from IgA nephropathy, can be determined by detecting the absorbancy of the patients or the quantitative value of concentration obtained from the same manner as above.
  • the determination method is not limited within the enzyme immunoassay, and all methods, which can determine antibody recognizing the core peptide of the hinge region in IgA1, can be used in the present test method. Examples thereof are as follows:
  • Latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed. Turbidity generated from the result of antigen-antibody reaction is measured as absorbancy.
  • the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen is determined quantitatively or qualitatively based on measurement of changes of the absorbancies.
  • Latex slide agglutination (Shibata, S. et al., Equipment and Reagents, Vol. 11, pp. 338 -342, 1988). Latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed on the plate, and agglutination of latex particles generated as a result of antigen-anitibody reaction is confirmed macroscopically to qualitate the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen.
  • the enzyme immunoassay in case of determining antibody recognizing the core peptide of the hinge region in IgA1 in specimens using microtiter plate immobilized with the core peptide of the hinge region in IgA1, for example, BECKMAN Biomek 1000 Automated Laboratory Workstation (Beckman Inc.) or CELL ASSAY-2000 ROBOTIC ASSAY SYSTEM (Moritex Inc.), both of which are automated system equipment with supplying specimens, supplying reagents, washing operation, measurement of absorbancy and data processing, can be used.
  • a specimen, enzyme labeled antibody and substrate solution are added, in this order, together with washing operation among each addition, in the microtiter plate immobilized with the core peptide of the hinge region in IgA1, and the reaction is terminated, if necessary, by adding a terminator for enzyme reaction, then absorbancy of the color development as a result of the enzyme reaction between the labeled enzyme and substrate used is measured at appropriate wave length selected from the customary measurement conditions to determine the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen.
  • the latex agglutination photometric immunoassay in order to determine antibody, which recognizes the core peptide of the hinge region in IgA1 in specimen by using latex particles having immobilized thereon the core peptide of the hinge region in IgA1, for example, a Hitachi 705, 7050, 7150, 736 or 7070 automatic analyzer, which are automatic equipment systems for supplying specimens, supplying reagents, measurement of absorbance and data processing, can be used.
  • latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed in a reaction cell, and the blank is corrected by measuring with suitable wave length selected from the known measurement conditions, preferably at 400 nm-950 nm, then changes of absorbancy after a constant time are measured to determine antibody recognizing the core peptide of the hinge region in IgA1 in the specimen.
  • measurement operations may be facilitated if the necessary reagents for practicing the present invention are prepared in a kit. Further, it is convenient to practice the present invention using automated analytical equipment.
  • Example of reagent kits used in the present invention is a kit comprising a combination of a carrier (such as microtiter plate or latex particles) immobilized with the core peptide of the hinge region in IgA1 and reagents necessary for practice in the present invention such as buffer, standard substance, labeled antibody, substrate, solubilizing agent for substrate or reaction terminator.
  • a carrier such as microtiter plate or latex particles
  • reagents necessary for practice in the present invention such as buffer, standard substance, labeled antibody, substrate, solubilizing agent for substrate or reaction terminator.
  • FIG. 1 is a schematic drawing showing the outline of a molecular structure of IgA1.
  • FIG. 2 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy and a group of non-IgA nephropathy in case of adding anti-human IgG.
  • FIG. 3 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy and a group of non-IgA nephropathy in case of adding anti-human IgM.
  • FIG. 4 is a graph plotting absorbancies at 490 nm showing the reactivity of the serum of patient with IgA nephropathy, previously reacted with various synthetic peptide or blank solution, and the core peptide of the hinge region in IgA1.
  • BSA bovine serum albumin, purchased from SIGMA Inc.
  • PBS 0.01 M phosphate buffer, pH 7.2, containing 0.15 M NaCl
  • EMCS N-( ⁇ -maleimidocaproyloxy) succinimide, purchased from Dojin Chemical Lab. Inc.
  • DMSO dimethyl sulfoxide purchased from Wako Pure Chemicals Co.
  • the incubation mixture was charged on an Econo-Pac IODG column (purchased from Bio-Rad Inc.) equilibrated with PBS, and eluated with PBS. Each 1 ml of the eluates was fractionated into 15 ml plastic centrifugal tube (purchased from Sumitomo Bakelite Co.), and absorbancy was measured at 280 nm to collect the protein fractions.
  • the incubation mixture was charged on the Econo-Pac 10DG column equilibrated with PBS and eluated with PBS. Each 1 ml of the eluates was fractionated in the 15 ml plastic centrifugal tube (purchased from Sumitomo Bakelite Co.), and was measured absorbancy at 280 nm and the protein fractions were collected to obtain a protein solution (19.3 mg/ml, 3 ml). The obtained solution was used as an antigen in the following experiments.
  • Coloring agent 100 ⁇ l
  • sodium dihydrogen phosphate 12H 2 O 1.795 g, citric acid 0.525 g, and hydrogen peroxide 15 ⁇ l were dissolved in water 50 ml) was added in each well, and incubated at room temperature for 1 hour.
  • FIG. 2 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy (in FIG. 2, IgAN group) and a group of non-IgA nephlopathy (in FIG. 2, non-IgAN group) in case of adding peroxidase labeled anti-human IgG.
  • FIG. 3 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy (in FIG. 3, IgAN group) and a group of non-IgA nephropathy (in FIG. 3, non-IgAN group) in case of adding peroxidase labeled anti-human IgM.
  • Antigen was prepared according to the method described in the above example 1 (1). (2) Assay of inhibitory activities of various synthetic peptides The above antigen solution was dissolved to 10 ⁇ g/ml in 0.015 M carbonate buffer (pH 9.6) and each 100 ⁇ l was added in each well of the 96 well microtiter plate (A Flow General Co., Linbro/Titeltek EIA Microtitration plate, Cat. No. 76-381-04), then incubated at 4° C. overnight (standing culture).
  • PBS (0.01M phosphate buffer, pH 7.5 containing 0.15M NaCl) solution (200 ⁇ l) containing 1% bovine serum albumin (fraction V, purchased from SIGMA Inc.) was added in each well, and incubated at 4° C. overnight. Each well was washed with 200 ⁇ l of PBS. The antigen was immobilized.
  • PVPST Pro Val Pro Ser Thr
  • PSTPP Pro Ser Thr Pro Pro
  • Thr Pro Pro Thr Pro (SEQ ID NO: 4) (hereinafter designated as TPPTP)
  • PTPSP Pro Thr Pro Ser Pro
  • PSPST Pro Ser Pro Ser Thr
  • PSPSC Pro Ser Pro Ser Cys
  • SPDLP Ser Pro Asp Leu Pro
  • LKPDP Leu Lys Pro Asp Pro
  • LPPLT Leu Pro Pro Leu Thr
  • TPVES Thr Pro Val Glu Ser
  • TPDET Thr Pro Asp Glu Ser
  • [0115] was dissolved to 1 ⁇ g/ml with PBS.
  • the above diluted serum of patient with IgA nephropathy and the solution of synthetic peptide were mixed in equal volume (1:1).
  • the above diluted serum of patient with IgA nephropathy and PBS without containing synthetic peptide (hereinafter designated as blank solution) were mixed in equal volume (1:1). These mixtures were incubated at room temperature for 3 hours.
  • each solution 100 ⁇ l was added in each well, which was provided with the antigen immobilized therein, of the 96 well microtiter plate and incubated at room temperature for 3 hours.
  • PS/BSA/Tween 0.05% Tween-20
  • Coloring agent 100 ⁇ l
  • sodium dihydrogen phosphate. 12H 2 O 1.795 g, citric acid 0.525 g, and hydrogen peroxide 15 ⁇ l were dissolved in water 50 ml) was added in each well, and incubated at room temperature for 1 hour.
  • FIG. 4 is a graph plotting absorbancies at 490 nm showing the reactivity of the serum of patient with IgA nepliropathy, previously reacted with various synthetic peptide or blank solution, and the core peptide of the hinge region in IgA1.
  • test method of the present invention could specifically measure the antibody recognizing the core peptide of the hinge region in IgA1, which was represented by the amino acid sequence of the formula Pro Val Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Cys
  • test in specimens, and when the peptide, which constructed a partial structure of the core peptide of the hinge region in IgA1 and consisting of at least 5 amino acids, was used, the test can be performed.
  • the test method of the present invention is advantageous as a rapid and simple test method for IgA nephropathy, since the test method has low risk of emotional distress for the patients, reduced risk of peripheral hemorrhage in the kidney and less financial burden to the patients, and operability with simple operation within short time.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Steroid Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

A test method for IgA nephropathy involves determining antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens. The method is a rapid and sample test method for IgA nephropathy having less emotional distress for the patients, low risk for peripheral hemorrhage of the kidney and reduced financial burden for the patients.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0001]
  • This invention relates to a novel test method for IgA nephropathy. More particularly, the present invention pertains to a rapid and simple test method for IgA nephropathy and a determination method of antibody, which has low risks in emotional distress, peripheral hemorrhage in the kidney and financial burden to the patient, by determining antibody recognizing IgA1 hinge region core peptide in the specimen. [0002]
  • 2. Description of Related Art [0003]
  • IgA (immunoglobulin A) nephropathy is a disease concept provided by Berger et al. (J. Urol., 74, pp. 694-695, 1968), and is a primary glomerular nephritis having features with clinically poor symptoms except for continuous proteinuria and hematuria, and histological features with precipitants consisting of mainly IgA in the mesangium. The incidence of IgA nephropathy in Japan is high and accounts for 30% of the chronic nephritis. The long term prognosis is not so favorable, and 10-15% of patients with 10 years progress and about 3 0% of patients with 20 years progress suffer from terminal renal failure. Consequently, IgA nephropathy is especially noticed as a causal disease for terminal renal failure. [0004]
  • At present, the only known test method for IgA nephropathy is the renal biopsy. This test method, however, causes emotional distress for the patients, and her may cause peripheral hemorrhage in the kidney after the biopsy. In addition, the patients must have absolute rest for more than 24 hours after the renal biopsy, and this requires the patients to stay in hospital for several days, and this causes a heavy financial burden for them. Furthermore, the test method has disadvantages including requiring many kinds of test facilities together with long term testing time. [0005]
  • Consequently, a test method for IgA nephropathy is desired, which is simple and operable within a short time as well as giving less emotional distress, without risk of peripheral hemorrhage of kidney and with less financial burden. [0006]
  • PROBLEMS TO BE SOLVED BY THE INVENTION
  • An object of the present invention is to provide a rapid and simple test method for IgA nephropathy. Such a test method requires no renal biopsy, which has been essential in the conventional test method for IgA nephropathy. The test method is comprised of detecting antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens. As a result, the patients' emotional distress, risk of peripheral hemorrhage of the kidney and the financial burden can be reduced. [0007]
  • MEANS FOR SOLVING THE PROBLEMS
  • We have made various trials for solving the above problems and have found a rapid and simple test method for IgA nephropathy and a determination method for antibody, which recognizes the core peptide of hinge region in IgA1, and completed the present invention. Such a test method is comprised of detecting antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens. As a result, the patients' emotional distress, risk of peripheral hemorrhage of the kidney and the financial burden can be reduced. [0008]
  • The present invention relates to a test method for IgA nephropathy by detecting antibody recognizing the core peptide of the hinge region in IgA1 in specimens. According to preferred embodiments of the present invention, a test method comprising detecting antibody recognizing the core peptide of the hinge acid sequence consisting of region having at least a part of amino acid sequence consisting of [0009]
    Pro Val Pro Ser Thr Pro Pro Thr Pro (SEQ ID NO:1)
    Ser Pro Ser Thr Pro Pro Thr Pro Ser
    Pro Ser Cys
  • or at least three amino acids thereof. According to a further preferred embodiment of the present invention, the test method comprises using an immobilized solid phase core peptide of the hinge region, said immobilized solid phase of which is microtiter plate or latex particles. [0010]
  • Examples of detection methods for the antibody of the present invention are test method for IgA nephropathy which includes enzyme immunoassay, latex agglutination photometric immunoassay, nephelometric immunoassay, latex slide agglutination test, turbidimetric immunoassay, luminescent immunoassay, fluorescence polarization immunoassay, fluorescence immunoassay and radioimmunoassay. [0011]
  • The present invention is explained in detail with reference to the drawings as follows. [0012]
  • FIG. 1 illustrates a schematic drawing showing the outline of a molecular structure of IgA1. [0013]
  • IgA, which may be involved in a cause for IgA nephropathy, is a type of immunoglobulin. The IgA molecule consists of two heavy chains and two light chains, and is classified into subtypes of IgA1 and IgA2 depending on the difference in heavy chain structures. [0014]
  • In IgA nephropathy, since the deposited IgA in the glomeruli is mainly IgA1 subtype (Conley et al., J. Clin. Invest. 66, pp. 1432-1436, 1980), the properties of IgA1 between those of patients with IgA nephropathy and those of healthy subjects or patients with non-IgA nepliropathy may differ from one another. [0015]
  • The largest structural difference between IgA1 and IgA2 exists in the hinge region of the heavy chain. The hinge region of IgA2 is deficient in 13 amino acids (Frangione et al., Proc. Natl. Acad. Sci. USA, 69, pp. 3673-3676, 1972) as compared with the hinge region of IgA1 (SEQ ID NO: 13) [Basic immunology, Upper Vol., pp. 158-162, (1986), The University Tokyo Press]. [0016]
  • Baenziger et al. reported that an 0-linked sugar chain is bonded to five serine residues of the hinge region in IgA1 molecule (J. Biol. Chem. 249, pp. 7270-7281, 1974). [0017]
  • We have demonstrated that the binding ability between the antibody obtained from rabbit uimmunized with synthetic peptide of the hinge region in IgA1 and the serum IgA1 of patients with IgA nephropathy was higher than the binding ability between the above antibody and the serum IgA1 of healthy subjects or patients with non-IgA nephropathy. As a result, we have reported that IgA1 of the patients with IgA nephropathy was abnormal in the O-linked sugar chain bound to the hinge region, and in conclusion, the core peptide of the hinge region (core peptide of hinge region means the peptide constructing the hinge region) was in an exposed state (J. Am. Renal. Soc. 8, pp. 538A, 1997). [0018]
  • The core peptide of the hinge region has structural similarity with mucin insofar as containing large amounts of proline, serine and threonine. [0019]
  • The immunologically activating ability of human mucin core peptide has been known. For example, Kotera et al. reported that the mucin which is frequently expressed in some types of cancer cells has less sugar chain, and, as a result, immune reaction against the exposed core peptide occurs in the host (Cancer Res., 54, pp. 2856-2860, 1994). [0020]
  • We have made extensive studies based on the above findings. As a result, we have found that, in patients with IgA nephropathy, 0-linked sugar chain bonded with the hinge region of IgA1 is decreased, and this results in exposing the core peptide of the hinge region, such that an immune reaction against the core peptide of the hinge region in IgA1 is generated and the antibody titer recognizing the core peptide of the hinge region in IgA1 is increased; and that these findings can be applied for testing IgA nephropathy, and completed the present invention. [0021]
  • A rapid test for IgA nephropathy can be performed by determining antibody, which recognizes the core peptide of the hinge region of IgA1, in specimens such as body fluid of the patients. Namely, the present invention relates to a test method for IgA nephropathy based on determining antibody, which recognizes the core peptide of the hinge region in IgA1, in specimens. [0022]
  • In the present invention, the hinge region of IgA1 indicates the region between CH1 domain and CH2 domain in the heavy chains, which construct IgA1 molecule, and may optionally include a structure having added thereto a sequence of Pro-Va1- in the N-terminal region. [0023]
  • This region contains disulfide bond between heavy chains, and is rich in proline, serine and threonine. [0024]
  • In the present invention, the core peptide of the hinge region in IgA1 as an antigen can include the peptide constructing the hinge region in IgA1 or a partial structure thereof. [0025]
  • Amino acid sequences of the peptide of the formula (SEQ ID NO: 1), [0026]
    Pro Val Pro Ser Thr Pro Pro Thr Pro
    Ser Pro Ser Thr Pro Pro Thr Pro Ser
    Pro Ser Cys
  • or peptides having partial structure thereof consisting of at least three amino acids thereof are preferably used. Examples of peptides are peptides comprising amino acid sequences of the formulas: [0027]
    Val Pro Ser, Ser Thr Pro, Thr Pro Pro,
    Pro Pro Thr, Pro Thr Pro, Thr Pro Ser,
    Pro Ser Pro, Ser Pro Ser or Pro Ser Thr.
  • More preferably, peptides consisting of at least five amino acids, for example, formulas, [0028]
    ProValProSerThr (SEQ ID NO: 2),
    ProSerThrProPro (SEQ ID NO: 3),
    ThrProProThrPro (SEQ ID NO: 4),
    ProThrProSerPro (SEQ ID NO: 5),
    ProSerProSerThr (SEQ ID NO: 6) or
    ProSerProSerCys (SEQ ID NO: 7)
  • can be mentioned. [0029]
  • In the present invention, so far as enabling, all peptides or proteins including peptide constructing the hinge region of IgA1 or a part thereof can be used. [0030]
  • Further, the core peptide of the hinge region in IgA1 modified by sugars, lipids, functional groups, protective groups or cross linking agents, and bound to carrier such as proteins, plastics or latex particles can be used. [0031]
  • In the present invention, any core peptide of the hinge region in IgA1, i.e. antigen, produced by any method, for example, a product by synthesis, a product derived from IgA1 originated from patients or subjects with non-IgA nephropathy, or a product by gene recombinant technology, can be used. [0032]
  • Examples of specimens in the present invention are biological fluid such as blood, plasma, serum, saliva or urine, biological tissue extracts and supernatant solutions of the tissue culture. [0033]
  • Specimens can be used without dilution or with aliquot dilution of buffer solution depending on the principle of detection method, detection sensitivity, detection range and other factors in the present invention. [0034]
  • Specimens can contain anticoagulants such as sodium EDTA, sodium citrate or heparin sodium, materials for stabilizing the specimen, or other substances. [0035]
  • With respect to buffer solutions used in the present invention, buffers which are customarily used in immunological determination methods and preferably have a pH value in the range of from 5 to 9 can be employed. [0036]
  • For example, phosphate buffer, borate buffer, carbonate buffer, Tris-HCl buffer, veronal buffer and Good's buffer such as HEPES buffer, PIPES buffer and MES buffer, can be used. [0037]
  • For the purpose of preventing non-specific adsorption of proteins to the carrier used in the determination method of the present invention such as microtiter plate and latex, any type of additives, for example, bovine serum albumin (BSA), protein such as gelatin or skimmed milk, surface active agents, sugars, chelating agents, reducing agents or salt can be added in the buffer solution. [0038]
  • The concentration of the additives can be selected from the concentration used in the immunological determination method of antibody, which is applied in known antigen-antibody reactions. [0039]
  • Examples of antibody to be determined in the present invention are all antibodies that react with the core peptide of the hinge region in IgA1 in specimens, and are preferably IgG class, IgM class or IgA class. [0040]
  • In the present invention, a method for immunologically determining the antibody, which recognizes the core peptide of the hinge region in IgA1 as the antigen, in specimens, is characterized by reacting the above described core peptide of the hinge region in IgA1 with the specimen under the condition to form antigen-antibody complex and qualitating or quantitating the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen based on assaying the thus generated product of the antigen-antibody reaction. [0041]
  • For example, enzyme immunoassay (EIA) (Proteins, Nucleic Acids and Enzymes, Supplement No. 31, Enzyme immunoassay, pp. 13-26, 1987) can be used. [0042]
  • In one embodiment, a carrier having immobilized thereon core peptide of the hinge region in IgA1 and a specimen are reacted, and a labeled antibody (a complex of anti-human IgG antibody, anti-human IgM antibody, anti-human IgA antibody or fragments thereof having binding activity to antigen and labeled enzyme) is reacted therewith, then the coloring agent (substrate) for developing color by applying the reaction with labeled enzyme bound with the carrier through the antigen-antibody complex, is added and subjected to reaction. [0043]
  • The reaction is terminated, if necessary, by adding enzyme reaction terminator, and optical absorption of the thus generated coloration is measured at a suitable wavelength to quantitatively or qualitatively assay the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen. [0044]
  • In a further detailed embodiment, in case that a complex bound with the synthetic core peptide of the hinge region in IgA1 consisting of peptide of at least three amino acids, preferably at least five amino acids, selected from the amino acid sequence of the core peptide of the hinge region in IgA1 such as amino acid sequence of the formula, [0045]
    Pro Val Pro Ser Thr Pro Pro Thr Pro
    Ser Pro Ser Thr Pro Pro Thr Pro Ser
    Pro Ser Cys
  • and the bovine serum albumin (BSA) at a binding ratio of 1:1-15:1 (peptide: BSA), preferably, 1:1-10:1 (peptide: BSA), is used as an antigen, the antigen adjusted within a concentration range generally from 0.01 μg/ml-100 μg/ml is added to the plastic microtiter plate, generally at 25 μl -200 μl/well, allowed to stand at 4° C.-40° C., for 10 min.-48 hours and is washed with suitable buffer to immobilize the antigen. Further, the specimens (such as serum of the patients) diluted with suitable buffer, generally 25 μl-200 μl, are added to the microtiter plate with the immobilized antigen, and allowed to stand generally at 4° C.-40° C., for 5 min.-48 hours then are washed with suitable buffer. [0046]
  • Next, the self-known labeled antibody (a complex of anti-human IgG antibody, anti-human IgM antibody, anti-human IgA antibody or fragments thereof having binding activity to antigen, and the labeling enzyme, for example generally used in the enzyme immunoassay, such as peroxidase, alkaline phosphatase, β-galactosidase or glucose oxidase) diluted with suitable buffer, generally 25 μl-200 μl, is added and allowed to react generally at 4° C.-40° C., for 10 min.-48 hours then are washed with suitable buffer. [0047]
  • Further, coloring agent (substrate) for developing color with a reaction of the labeled enzyme is added generally 25 μl-200 μl, and is reacted generally within a range at 4° C. -40° C., for 1 min.-48 hours. [0048]
  • The reaction is terminated, if necessary, by adding enzyme reaction terminator. Optical absorb any of the supernatant of the enzymatic reaction mixture is colorimetrically measured and the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen is qualitatively or quantitatively measured. [0049]
  • In the above, in case that peroxidase is selected as the labeled enzyme, orthophenylenediamine and the like can be used as a substrate, and the resultant coloring of the enzyme reaction is measured by absorbancy at 490 nm. [0050]
  • Further, in case that alkaline phosphatase is selected as the labeled enzyme, paranitrophenyl phosphate can be used as a substrate, and the resultant color development of the enzyme reaction is measured by absorbancy at 405 nm. [0051]
  • If the antibody titer, which recognizes the core peptide of the hinge region in IgA1 (IgG class antibody titer, IgM class antibody titer or IgA class antibody titer), is high, the above absorbancy values are also high. [0052]
  • For example, the examination for IgA nephropathy can be performed as follows: the absorbancy of the specimens obtained from the healthy subjects or patients with non-IgA nephropathy, or the concentration values in these specimens, which are quantitatively determined by comparing the obtained absorbancy with the calibration curve calculated from the standard substance, are obtained. Measurement above the average of these values +2× standard deviation are deemed to be positive, and those below or not above the value thereof are considered to be negative. Using these criteria, the examination of subjects, whether or not the patient is suffering from IgA nephropathy, can be determined by detecting the absorbancy of the patients or the quantitative value of concentration obtained from the same manner as above. [0053]
  • The determination method is not limited within the enzyme immunoassay, and all methods, which can determine antibody recognizing the core peptide of the hinge region in IgA1, can be used in the present test method. Examples thereof are as follows: [0054]
  • (a) Conventionally used latex agglutination photometric immunoassay; LAPIA (Sakurabayashi, 1. et al., Nihon Rinsho, Vol. 48, supplement, pp. 13 56-1361, 1990). Latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed. Turbidity generated from the result of antigen-antibody reaction is measured as absorbancy. The antibody recognizing the core peptide of the hinge region in IgA1 in the specimen is determined quantitatively or qualitatively based on measurement of changes of the absorbancies. [0055]
  • (b) Nephelometric immunoassay; NIA (Yamagishi, Y. et al., Rinsho-Kensa (Clinical Examinations), Vol. 23, Supplement, pp.1286-1289, 1979). Latex particles immobilized with the core peptide of the hinge region in IgA1 are mixed with a specimen. The thus produced antigen-antibody complex is irradiated with light (laser) and changes of the scattering strength are measured, and the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen is quantitatively or qualitatively measured. [0056]
  • (c) Latex slide agglutination (Shibata, S. et al., Equipment and Reagents, Vol. 11, pp. 338 -342, 1988). Latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed on the plate, and agglutination of latex particles generated as a result of antigen-anitibody reaction is confirmed macroscopically to qualitate the antibody, which recognizes the core peptide of the hinge region in IgA1, in the specimen. [0057]
  • (d) Counting immunoassay; CIA (Hashimoto, K. et al., Test and Technology, Vol. 22, No. 5, pp. 67-68, 1994). Latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed. Sizes and numbers of aggregates generated as a result of antigen-antibody reaction are measured. The antibody recognizing the core peptide of the hinge region in IgA1 in the specimen can be qualitatively or quantitatively measured. [0058]
  • (e) Turbidimetric immunoassay; TIA (Toyama, T., Clinical pathology, Vol. 35, pp. 868-873, 1987). The core peptide of the hinge region in IgA1 and a specimen are mixed. Turbidity generated as a result of antigen-antibody reaction is measured and changes of absorbancy are measured to qualitate or quantitate the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen. [0059]
  • (f) Luminescent immunoassay; LIA (“Proteins, Nucleic Acids and Enzymes”, Suppl. 31, “Enzyme Immunoassay”, pp. 252-263, 1987). [0060]
  • (g) Fluorescence polarization immunoassay; FPIA (Kurata, K., “New case studies on immunoassay and application for development of diagnostic reagents and therapeutic agents”, Keiei-Kyoiku Publ., Jun. 20, 1985, pp. 91-102). [0061]
  • (h) Immunoelectrophoresis (Urushizaki, I. et al., “New case studies on immunoassay and application for development of diagnostic reagents and therapeutic agents”, Keiei-Kyoiku Publ., Jun. 20, 1985, pp. 63-72). [0062]
  • (i) Spin immunoassay; SIA (Sago, H., “New case studies on immunoassay and application for development of diagnostic reagents and therapeutic agents”, Keiei-Kyoiku Publ., Jun. 20, 1985, pp. 52-62). [0063]
  • (j) Fluorescence immunoassay; FIA (Hashimoto, T. et al., Test and Technology, Vol. 22, No.5, pp.61-66, 1994), and [0064]
  • (k) Radioimmunoassay; RIA (Kurata,K., “New case studies on immunoassay and application for development of diagonostic reagents and therapeutic agents”, Keiei-Kyoiku Publ., Jun. 20, 1985, pp. 33-41). [0065]
  • Qualitative and quantitative assay methods for a product of antigen-antibody reaction in the present invention are not limited to those performed by manual operation but can be performed by applying automatic analysis. [0066]
  • In the enzyme immunoassay, in case of determining antibody recognizing the core peptide of the hinge region in IgA1 in specimens using microtiter plate immobilized with the core peptide of the hinge region in IgA1, for example, BECKMAN Biomek 1000 Automated Laboratory Workstation (Beckman Inc.) or CELL ASSAY-2000 ROBOTIC ASSAY SYSTEM (Moritex Inc.), both of which are automated system equipment with supplying specimens, supplying reagents, washing operation, measurement of absorbancy and data processing, can be used. [0067]
  • In detail, a specimen, enzyme labeled antibody and substrate solution are added, in this order, together with washing operation among each addition, in the microtiter plate immobilized with the core peptide of the hinge region in IgA1, and the reaction is terminated, if necessary, by adding a terminator for enzyme reaction, then absorbancy of the color development as a result of the enzyme reaction between the labeled enzyme and substrate used is measured at appropriate wave length selected from the customary measurement conditions to determine the antibody recognizing the core peptide of the hinge region in IgA1 in the specimen. [0068]
  • In the latex agglutination photometric immunoassay, in order to determine antibody, which recognizes the core peptide of the hinge region in IgA1 in specimen by using latex particles having immobilized thereon the core peptide of the hinge region in IgA1, for example, a Hitachi 705, 7050, 7150, 736 or 7070 automatic analyzer, which are automatic equipment systems for supplying specimens, supplying reagents, measurement of absorbance and data processing, can be used. [0069]
  • In detail, latex particles immobilized with the core peptide of the hinge region in IgA1 and a specimen are mixed in a reaction cell, and the blank is corrected by measuring with suitable wave length selected from the known measurement conditions, preferably at 400 nm-950 nm, then changes of absorbancy after a constant time are measured to determine antibody recognizing the core peptide of the hinge region in IgA1 in the specimen. [0070]
  • In the method of the present invention, measurement operations may be facilitated if the necessary reagents for practicing the present invention are prepared in a kit. Further, it is convenient to practice the present invention using automated analytical equipment. [0071]
  • Example of reagent kits used in the present invention is a kit comprising a combination of a carrier (such as microtiter plate or latex particles) immobilized with the core peptide of the hinge region in IgA1 and reagents necessary for practice in the present invention such as buffer, standard substance, labeled antibody, substrate, solubilizing agent for substrate or reaction terminator.[0072]
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 is a schematic drawing showing the outline of a molecular structure of IgA1. [0073]
  • FIG. 2 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy and a group of non-IgA nephropathy in case of adding anti-human IgG. [0074]
  • FIG. 3 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy and a group of non-IgA nephropathy in case of adding anti-human IgM. [0075]
  • FIG. 4 is a graph plotting absorbancies at 490 nm showing the reactivity of the serum of patient with IgA nephropathy, previously reacted with various synthetic peptide or blank solution, and the core peptide of the hinge region in IgA1.[0076]
  • MODE FOR CARRYING OUT THE INVENTION
  • Examples of the present invention are illustrated in detail with reference to the drawings, but the present invention is not restricted by these examples. [0077]
  • EXAMPLE 1
  • (I) Preparation of Antigen [0078]
  • BSA (bovine serum albumin, purchased from SIGMA Inc.) was dissolved to 13.4 mg/ml with PBS (0.01 M phosphate buffer, pH 7.2, containing 0.15 M NaCl), and 1.5 ml thereof was added to 15 ml plastic centrifugal tube (purchased from Sumitomo Bakelite Co.). A solution (100 μl) of EMCS (N-(ε-maleimidocaproyloxy) succinimide, purchased from Dojin Chemical Lab. Inc.) in DMSO (dimethyl sulfoxide purchased from Wako Pure Chemicals Co.) (100 mg/ml) was added to the above BSA solution, and incubated at room temperature for 60 minutes. [0079]
  • The incubation mixture was charged on an Econo-Pac IODG column (purchased from Bio-Rad Inc.) equilibrated with PBS, and eluated with PBS. Each 1 ml of the eluates was fractionated into 15 ml plastic centrifugal tube (purchased from Sumitomo Bakelite Co.), and absorbancy was measured at 280 nm to collect the protein fractions. [0080]
  • A solution (250 μl) of synthetic core peptide of the hinge region in IgA1 [0081]
    Pro Val Pro Ser Thr Pro Pro Thr Pro
    Ser Pro Ser Thr Pro Pro Thr Pro Ser
    Pro Ser Cys
  • (molecular weight 2031.3) (synthesized in Acord Inc.) in PBS (20 mg/ml) was added to the above collected protein fraction (3 ml) and incubated at 4° C. for 36 hours. [0082]
  • The incubation mixture was charged on the Econo-Pac 10DG column equilibrated with PBS and eluated with PBS. Each 1 ml of the eluates was fractionated in the 15 ml plastic centrifugal tube (purchased from Sumitomo Bakelite Co.), and was measured absorbancy at 280 nm and the protein fractions were collected to obtain a protein solution (19.3 mg/ml, 3 ml). The obtained solution was used as an antigen in the following experiments. [0083]
  • (2) Measurement of Antibody Titer [0084]
  • The above antigen solution was dissolved to 10 μg/ml in 0.015 M carbonate buffer (pH 9.6) and each 100 μl was added in each well of the 96 well microtiter plate (A Flow General Co., Linbro/Titertek EIA Microtitration plate, Cat. No. 76-381-04), then incubated at 4° C. overnight (standing culture). [0085]
  • PBS (0.01 M phosphate buffer, pH 7.5, containing 0. 1 5 M NaCl) solution (200 1) containing 1% bovine serum albumin (fraction V, purchased from SIGMA Inc.) was added in each well, and incubated at 4° C. overnight. Each well was washed with 200 μl of PBS. Sera of 32 cases in a group of patients with IgA nephropathy and sera of 32 cases in a group of patients with non-IgA nephropathy (a group of patients with kidney diseases without IgA nephropathy) were diluted to 1/100 with PBS, and the diluted solution (each 100 μl) was added in each well. After incubation at room temperature for 3 hours, each well was washed three times with PBS (200 μl) containing 0.1% bovine serum albumin and 0.05% Tween-20. [0086]
  • A solution (100 μl) of peroxidase labeled anti-human IgG purchased from Organon Tecknika Inc.) or a solution (100 μl) of peroxidase labeled anti-human IgM (purchased from Organon Tecknika Inc.), each of which was diluted to 1/200 with PBS, was added in each well. [0087]
  • After incubation at room temperature for 1 hour, each well was washed three times with PBS (200 μl) containing 0.1% bovine serum albumin and 0.05% Tween-20. [0088]
  • Coloring agent (100 μl)(orthophenylenediamine 20 mg, sodium dihydrogen phosphate 12H[0089] 2O 1.795 g, citric acid 0.525 g, and hydrogen peroxide 15 μl were dissolved in water 50 ml) was added in each well, and incubated at room temperature for 1 hour.
  • Absorbancy at 490 nm was measured using a microplate reader (Bio-Rad Inc., Microplate reader Model 450). [0090]
  • (3) Result of IgG Class Antibody Titer [0091]
  • FIG. 2 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy (in FIG. 2, IgAN group) and a group of non-IgA nephlopathy (in FIG. 2, non-IgAN group) in case of adding peroxidase labeled anti-human IgG. [0092]
  • In this case, absorbancy showing above the average values (0.316) of 32 cases of a group of patients with non-IgA nephropathy+2× standard deviation=0.606 is set as positive. [0093]
  • As a result of the experiments, in the group of patients with non-IgA nephropathy, all of 32 cases were negative, on the contrary, in the group of patients with IgA nephropathy, 16 out of 32 cases were positive, and a statistically significant difference between the two groups was noted in the positive rate. [0094]
  • (4) Result of IgM Class Antibody Titer [0095]
  • FIG. 3 is a graph plotting absorbancies of wave length at 490 nm in specimens of 32 patients with a group of IgA nephropathy (in FIG. 3, IgAN group) and a group of non-IgA nephropathy (in FIG. 3, non-IgAN group) in case of adding peroxidase labeled anti-human IgM. [0096]
  • In this case, absorbancy showing above the average values (0.134) of 32 cases of a group of patients with non-IgA nephropathy+2×standard deviation=0.272 is set as positive. [0097]
  • As a result of the experiments, in the group of patients with non-IgA nephropahty, all of 32 cases were negative, on the contrary, in the group of patients with IgA nephropathy, 13 out of 32 cases were positive, and a statistically significant difference between the two groups was noted in the positive rate. [0098]
  • EXAMPLE 2
  • (1) Preparation of Antigen [0099]
  • Antigen was prepared according to the method described in the above example 1 (1). (2) Assay of inhibitory activities of various synthetic peptides The above antigen solution was dissolved to 10 μg/ml in 0.015 M carbonate buffer (pH 9.6) and each 100 μl was added in each well of the 96 well microtiter plate (A Flow General Co., Linbro/Titeltek EIA Microtitration plate, Cat. No. 76-381-04), then incubated at 4° C. overnight (standing culture). [0100]
  • PBS (0.01M phosphate buffer, pH 7.5 containing 0.15M NaCl) solution (200 μl) containing 1% bovine serum albumin (fraction V, purchased from SIGMA Inc.) was added in each well, and incubated at 4° C. overnight. Each well was washed with 200 μl of PBS. The antigen was immobilized. [0101]
  • Serum of a patient with IgA nephropathy which showed positive in example 1 was diluted to 1/50 with PBS. Synthetic peptide [the core peptide constructing a part of the hinge region in IgA1 as follows: [0102]
  • Pro Val Pro Ser Thr (SEQ ID NO: 2) (hereinafter designated as PVPST) [0103]
  • Pro Ser Thr Pro Pro (SEQ ID NO: 3) (hereinafter designated as PSTPP) [0104]
  • Thr Pro Pro Thr Pro (SEQ ID NO: 4) (hereinafter designated as TPPTP) [0105]
  • Pro Thr Pro Ser Pro (SEQ ID NO: 5) (hereinafter designated as PTPSP) [0106]
  • Pro Ser Pro Ser Thr (SEQ ID NO: 6) (hereinafter designated as PSPST) [0107]
  • Pro Ser Pro Ser Cys (SEQ ID NO: 7) (hereinafter designated as PSPSC) [0108]
  • and the core peptide constructing a part of the bovine serum albumin as follows: [0109]
  • Ser Pro Asp Leu Pro (SEQ ID NO: 8) (hereinafter designated as SPDLP); [0110]
  • Leu Lys Pro Asp Pro (SEQ ID NO: 9) (hereinafter designated as LKPDP); [0111]
  • Leu Pro Pro Leu Thr (SEQ ID NO: 10) (hereinafter designated as LPPLT); [0112]
  • Thr Pro Val Glu Ser (SEQ ID NO: 11) (hereinafter designated as TPVES); [0113]
  • Thr Pro Asp Glu Ser (SEQ ID NO: 12) (hereinafter designated as TPDET)] [0114]
  • was dissolved to 1 μg/ml with PBS. The above diluted serum of patient with IgA nephropathy and the solution of synthetic peptide were mixed in equal volume (1:1). Also the above diluted serum of patient with IgA nephropathy and PBS without containing synthetic peptide (hereinafter designated as blank solution) were mixed in equal volume (1:1). These mixtures were incubated at room temperature for 3 hours. [0115]
  • After termination of the incubation, each solution (100 μl) was added in each well, which was provided with the antigen immobilized therein, of the 96 well microtiter plate and incubated at room temperature for 3 hours. [0116]
  • Each well was washed three times with 200 μl of PBS containing 0.1% bovine serum albumin, 0.05% Tween-20 (hereinafter designated as PS/BSA/Tween). [0117]
  • A solution (100 μl) of peroxidase labeled anti-human IgG (purchased from Organon Tecknika Inc.), which was diluted to 1/200 with PBS, was added in each well. [0118]
  • After incubation at room temperature for 1 hour, each well was washed three times with PS/B SA/Tween (200 μl). [0119]
  • Coloring agent (100 μl)(orthophenylenediamine 20 mg, sodium dihydrogen phosphate. 12H[0120] 2O 1.795 g, citric acid 0.525 g, and hydrogen peroxide 15 μl were dissolved in water 50 ml) was added in each well, and incubated at room temperature for 1 hour.
  • After the incubation was terminated, absorbency at 490 nm was measured using microplate reader (Bio-Red Inc., Microplate reader Model 450). [0121]
  • (3) Results of Measurement of Inhibitory Activities of Various Synthetic Peptides [0122]
  • FIG. 4 is a graph plotting absorbancies at 490 nm showing the reactivity of the serum of patient with IgA nepliropathy, previously reacted with various synthetic peptide or blank solution, and the core peptide of the hinge region in IgA1. [0123]
  • The results of the experiments indicated that no inhibitory action on the reaction was observed in case of adding the core peptide (in FIG. 4, SPDLP, LKPDP, LPPLT, TPVES and TPDET) constructing a partial structure of bovine serum albumin, and less than 10% of difference in the absorption between the added group and the blank solution (in FIG. 4, Blank) was noted. [0124]
  • On the other hand, when the core peptide (in FIG. 4, PVPST, PSTPP, TPPTP, PTPSP, PSPST and PSPSC) constructing a partial structure of the hinge region in IgA1 was added, the inhibitory action on the reaction was observed, and more than 10% of difference in the absorption between the added group and the blank solution (in FIG. 4, Blank) was noted. [0125]
  • It was demonstrated that as a result of the above experiments, the test method of the present invention could specifically measure the antibody recognizing the core peptide of the hinge region in IgA1, which was represented by the amino acid sequence of the formula [0126]
    Pro Val Pro Ser Thr Pro Pro Thr Pro
    Ser Pro Ser Thr Pro Pro Thr Pro Ser
    Pro Ser Cys
  • in specimens, and when the peptide, which constructed a partial structure of the core peptide of the hinge region in IgA1 and consisting of at least 5 amino acids, was used, the test can be performed. [0127]
  • Sequence listings of the core peptide of the hinge region in IgA1 and the core peptide constructing a partial structure of bovine serum albumin are shown hereinbelow for reference. [0128]
  • EFFECT OF THE INVENTION
  • The test method of the present invention is advantageous as a rapid and simple test method for IgA nephropathy, since the test method has low risk of emotional distress for the patients, reduced risk of peripheral hemorrhage in the kidney and less financial burden to the patients, and operability with simple operation within short time. [0129]
  • 1 13 1 21 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 1 Pro Val Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Thr Pro Pro Thr 1 5 10 15 Pro Ser Pro Ser Cys 20 2 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 2 Pro Val Pro Ser Thr 1 5 3 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 3 Pro Ser Thr Pro Pro 1 5 4 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 4 Thr Pro Pro Thr Pro 1 5 5 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 5 Pro Thr Pro Ser Pro 1 5 6 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 6 Pro Ser Pro Ser Thr 1 5 7 5 PRT Artificial Sequence Description of Artificial Sequence Formula amino acid sequence 7 Pro Ser Pro Ser Cys 1 5 8 5 PRT Bovine sp. 8 Ser Pro Asp Leu Pro 1 5 9 5 PRT Bovine sp. 9 Leu Lys Pro Asp Pro 1 5 10 5 PRT Bovine sp. 10 Leu Pro Pro Leu Thr 1 5 11 5 PRT Bovine sp. 11 Thr Pro Val Glu Ser 1 5 12 5 PRT Bovine sp. 12 Thr Pro Asp Glu Thr 1 5 13 19 PRT Unknown Organism Description of Unknown Organism IgA1 13 Pro Ser Thr Pro Pro Thr Pro Ser Pro Ser Thr Pro Pro Thr Pro Ser 1 5 10 15 Pro Ser Cys

Claims (3)

What is claimed:
1. A test method for detecting IgA nephropathy in a patient suspected of having IgA nephropathy comprising obtaining a sample comprising immunoglobulins from the patient; contacting the sample with a hinge-region core peptide of IgA1 comprising amino acid sequence SEQ ID NO:1 or a fragment thereof comprising at least three sequential amino acids, and determining presence or amount of antibody in the sample which specifically binds to the core peptide, wherein an increased amount of said antibody determined in the sample relative to an amount determined in control samples is indicative of IgA nephropathy in the patient.
2. The method of claim 1, wherein the core peptide is immobilized on a solid phase.
3. The method of claim 2, wherein the solid phase is selected from latex particles or a multi-well microtiter plate.
US10/117,798 1998-03-31 2002-04-09 Test method for IgA nephropathy Abandoned US20030017619A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/117,798 US20030017619A1 (en) 1998-03-31 2002-04-09 Test method for IgA nephropathy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP10175998 1998-03-31
JP10-101759 1998-03-31
US09/646,154 US6429024B1 (en) 1998-03-31 1999-03-25 Test method for IgA nephropathy
US10/117,798 US20030017619A1 (en) 1998-03-31 2002-04-09 Test method for IgA nephropathy

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US09/646,154 Division US6429024B1 (en) 1998-03-31 1999-03-25 Test method for IgA nephropathy
PCT/JP1999/001525 Division WO1999050663A1 (en) 1998-03-31 1999-03-25 METHOD FOR EXAMINING IgA NEPHROPATHY

Publications (1)

Publication Number Publication Date
US20030017619A1 true US20030017619A1 (en) 2003-01-23

Family

ID=14309172

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/646,154 Expired - Lifetime US6429024B1 (en) 1998-03-31 1999-03-25 Test method for IgA nephropathy
US10/117,798 Abandoned US20030017619A1 (en) 1998-03-31 2002-04-09 Test method for IgA nephropathy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/646,154 Expired - Lifetime US6429024B1 (en) 1998-03-31 1999-03-25 Test method for IgA nephropathy

Country Status (6)

Country Link
US (2) US6429024B1 (en)
EP (1) EP1069432B1 (en)
JP (1) JP4197393B2 (en)
AT (1) ATE396399T1 (en)
DE (1) DE69938780D1 (en)
WO (1) WO1999050663A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100015044A1 (en) * 2005-08-03 2010-01-21 Robert Qiu Methods and compositions for diagnosis of iga-and igm-mediated kidney diseases
US20110201519A1 (en) * 2008-08-18 2011-08-18 Sarwal Minnie M Methods and Compositions for Determining a Graft Tolerant Phenotype in a Subject
WO2012139051A2 (en) * 2011-04-06 2012-10-11 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for iga nephropathy
US8932808B1 (en) 2004-01-21 2015-01-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
US9290813B2 (en) 2009-12-02 2016-03-22 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for determining an allograft tolerant phenotype
US9535075B2 (en) 2010-03-25 2017-01-03 The Board Of Trustees Of The Leland Stanford Junior University Protein and gene biomarkers for rejection of organ transplants
US9938579B2 (en) 2009-01-15 2018-04-10 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
USRE46843E1 (en) 2005-03-14 2018-05-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
USRE47057E1 (en) 2005-03-14 2018-09-25 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2360291T3 (en) * 2003-03-07 2011-06-02 New England Medical Center Hospitals, Inc. TREATMENT OF DISEASES BY DEPOSITION OF IgA1.
CA2789196A1 (en) * 2009-12-28 2011-07-07 Kyowa Hakko Kirin Co., Ltd. Anti-iga1 antibody
US9598733B2 (en) 2011-02-17 2017-03-21 The Trustees Of Columbia University In The City Of New York Methods for identifying subjects with a genetic risk for developing IgA nephropathy
US10202647B2 (en) 2013-04-12 2019-02-12 The Trustees Of Columbia University In The City Of New York Mutations in DSTYK cause dominant urinary tract malformations

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE461616B (en) * 1987-05-08 1990-03-05 Biocarb Ab PROCEDURE AND TEST RATE FOR DIAGNOSIS OF IGA NEPHROPATI WITH DETERMINATION OF FIBRONECTIN-IGA COMPLEX
WO1997044663A1 (en) * 1996-05-22 1997-11-27 Asahi Kasei Kogyo Kabushiki Kaisha EXAMINATION METHOD AND EXAMINATION KIT FOR IgA NEPHROPATHY
JPH09311132A (en) 1996-05-22 1997-12-02 Asahi Chem Ind Co Ltd Diagnostic method of immunoglobulin-a nephropathy

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8932808B1 (en) 2004-01-21 2015-01-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
USRE47057E1 (en) 2005-03-14 2018-09-25 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
USRE46843E1 (en) 2005-03-14 2018-05-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
US20100015044A1 (en) * 2005-08-03 2010-01-21 Robert Qiu Methods and compositions for diagnosis of iga-and igm-mediated kidney diseases
US9803241B2 (en) 2008-08-18 2017-10-31 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
US20110201519A1 (en) * 2008-08-18 2011-08-18 Sarwal Minnie M Methods and Compositions for Determining a Graft Tolerant Phenotype in a Subject
US10538813B2 (en) 2009-01-15 2020-01-21 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
US9938579B2 (en) 2009-01-15 2018-04-10 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
US9290813B2 (en) 2009-12-02 2016-03-22 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for determining an allograft tolerant phenotype
US9535075B2 (en) 2010-03-25 2017-01-03 The Board Of Trustees Of The Leland Stanford Junior University Protein and gene biomarkers for rejection of organ transplants
US8962261B2 (en) 2011-04-06 2015-02-24 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for IGA nephropathy
WO2012139051A3 (en) * 2011-04-06 2013-03-07 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for iga nephropathy
WO2012139051A2 (en) * 2011-04-06 2012-10-11 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for iga nephropathy

Also Published As

Publication number Publication date
EP1069432A1 (en) 2001-01-17
EP1069432B1 (en) 2008-05-21
WO1999050663A1 (en) 1999-10-07
EP1069432A4 (en) 2004-08-18
DE69938780D1 (en) 2008-07-03
JP4197393B2 (en) 2008-12-17
US6429024B1 (en) 2002-08-06
ATE396399T1 (en) 2008-06-15

Similar Documents

Publication Publication Date Title
EP0615129B1 (en) Methods for selectively detecting perinuclear anti-neutrophil cytoplasmic antibody of ulcerative colitis or primary sclerosing cholangitis
US4886761A (en) Polysilicon binding assay support and methods
US10830771B2 (en) PIVKA-II assay method and method for manufacturing reagent or kit for PIVKA-II immunoassay
WO2013132347A2 (en) Improved elisa immunoassay for calprotectin
WO2013132338A2 (en) Competitive immunoassay for calprotectin
US6429024B1 (en) Test method for IgA nephropathy
JPWO1999050663A6 (en) Testing methods for IgA nephropathy
CN111781372A (en) Alpha 1-AT immunoturbidimetry detection kit based on mixed antibody and preparation and use methods thereof
US20040248216A1 (en) Method of examining cancer by assaying autoantibody against mdm2 and reagent therefor
KR20010025027A (en) Immunoassay reagents and immunoassay method
CA2077906A1 (en) Method for measurement of tissue factor in high sensitivity and measurement kit therefor
JPH0580053A (en) Immunochemical detection method for human corpus uteri cancer cell
JPH03170058A (en) Reagent complex for immunoassay
EP0398292B1 (en) Diagnostic composition for rheumatoid arthritis
JP2021148804A (en) Antibody for detecting helicobacter pylori
US5436132A (en) Quantitative determination of tenascin as glioma marker
EP0125368A1 (en) A method of immunoassay detection by reaction-rate potentiometry using fluoride ion-selective electrode
EP0248038A1 (en) Idiotypic-antigenic conjunction binding assay
WO2023068249A1 (en) Measuring reagent for cross-linked n-telopeptide of type i collagen, preparation method thereof, and immunoassay method using same
JP2651438B2 (en) Enzyme-labeled antibody-sensitized latex and enzyme immunoassay using the same
CN113817025B (en) SLE epitope polypeptides in the identification of SLE and other autoimmune diseases
WO2023068248A1 (en) Immunoassay method for cross-linked n-telopeptide of type i collagen, immunoassay kit, and antibody or antibody fragment thereof
JPH11166931A (en) Method for detecting quantitative or qualitative anomaly in organism component
JPS62184353A (en) Immunochemical measurement method for human epithelia cell growth factor and kit for said method
WO2017159473A1 (en) Method for assessing likelihood of chronic kidney disease patient developing hyperphosphatemia

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION