WO2024237108A1 - Anti-aplp1 monoclonal antibody and use thereof - Google Patents
Anti-aplp1 monoclonal antibody and use thereof Download PDFInfo
- Publication number
- WO2024237108A1 WO2024237108A1 PCT/JP2024/016798 JP2024016798W WO2024237108A1 WO 2024237108 A1 WO2024237108 A1 WO 2024237108A1 JP 2024016798 W JP2024016798 W JP 2024016798W WO 2024237108 A1 WO2024237108 A1 WO 2024237108A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- aplp1
- extracellular vesicles
- antibody
- monoclonal antibody
- recovering
- 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.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING 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
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/02—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
Definitions
- Patent document 1 WO2021/132352
- Non-patent document 1 Alzheimer's & Dementia 15 (2019) 1071-1080
- the entire disclosures of Patent Document 1 and Non-Patent Document 1 are each expressly incorporated herein by reference.
- amino acid sequences represented by the sequence numbers of the light and heavy chains are shown in Tables 1 and 2 below.
- the anti-APLP1 monoclonal antibody of the present invention can be produced using the methods described in, for example, Kurosawa et al., “Rapid production of antigen-specific monoclonal antibodies from a variety of animals” BMC Biology 2021, 10:80 (Non-Patent Document 2), Kurosawa et al., “Novel method for the high-throughput production site-specific monoclonal antibodies” SCIENTIFIC REPORT 6:25174 DOI:10.1038 srep25174 (Non-Patent Document 3) and WO2011/027808 (Patent Document 2) and WO2012/133572 (Patent Document 3).
- the entire disclosures of Patent Documents 2 and 3, and Non-Patent Documents 2 and 3, respectively, are expressly incorporated herein by reference.
- Monoclonal antibodies with specific light and heavy chain amino acid sequences as well as anti-APLP1 monoclonal antibodies with specific light chain amino acid sequences CD1, CD2, CD3, FR1, FR2, FR3, and FR4, and heavy chain CD1, CD2, CD3, FR1, FR2, FR3, and FR4, can be produced by known methods, including those described in Non-Patent Documents 2 and 3, and Patent Documents 2 and 3, but are not intended to be limited thereto.
- the method for recovering extracellular vesicles includes a step of mixing an anti-APLP1 monoclonal antibody (hereinafter, sometimes referred to as anti-APLP1 antibody) with a sample containing extracellular vesicles, and a step of recovering a complex of the anti-APLP1 antibody and the extracellular vesicles.
- anti-APLP1 antibody an anti-APLP1 monoclonal antibody
- the anti-APLP1 antibody and the extracellular vesicles can form a complex.
- Extracellular vesicles include exosomes, microvesicles, apoptotic bodies, etc.
- biomolecules are present in extracellular vesicles.
- exosomes or microvesicles contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides (RNA such as mRNA, miRNA, non-coding RNA, and DNA).
- apoptotic bodies contain at least one type selected from the group consisting of fragmented nuclei and organelles.
- Extracellular vesicles preferably contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides.
- extracellular vesicles contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides.
- a polypeptide refers to a compound in which multiple amino acids are bound by peptide bonds, and includes proteins with relatively large molecular weights and peptides with relatively small molecular weights.
- the sample contains extracellular vesicles roughly purified from a specimen containing extracellular vesicles.
- the sample may be a dispersion of extracellular vesicles, or a pellet of extracellular vesicles.
- extracellular vesicles can be crudely purified, for example, by size exclusion chromatography, ultracentrifugation, affinity purification, polymer precipitation, and combinations thereof.
- Size exclusion chromatography is not limited as long as it can fractionate extracellular vesicles by size.
- size exclusion chromatography can be performed using an extracellular vesicle extraction kit qEV (Izon Science).
- Ultracentrifugation can be performed, for example, by ultracentrifugation at 100,000 g to 150,000 g for about 2 to 3 hours to obtain extracellular vesicles.
- Methods for detecting biomarkers for psychiatric and neurological disorders as polypeptides include known methods such as Western blotting and Enzyme-Linked Immunosorbent Assay (ELISA).
- Methods for detecting biomarkers for psychiatric and neurological disorders as RNA include known methods such as RT-PCR (including quantitative RT-PCR), microarrays, and RNA-Seq.
- Methods for detecting biomarkers for psychiatric and neurological disorders as DNA include known methods such as PCR (including quantitative PCR), microarrays, and sequencing.
- components derived from brain nerve cells are recovered using the extracellular vesicles recovered in 2 above.
- the method for recovering components derived from brain nerve cells includes a step of recovering at least one biomolecule selected from the group consisting of sugars, lipids, polypeptides, and polynucleotides from the extracellular vesicles recovered in 1 above.
- the sugars may include monosaccharides, disaccharides, oligosaccharides, and polysaccharides.
- the sugars may also be bound to lipids, proteins, or the like.
- the sugars can be recovered from the extracellular vesicles by known methods using hydrazide-oxyamine-bearing polymers, lectins, and the like.
- Lipids may include fatty acids, eicosanoids, triacylglycerols, wax esters, phospholipids, sphingolipids, isoprenoids, lipoproteins, etc. Lipids can be extracted from extracellular vesicles using the Folch method, a lipid extraction kit (Cell Biolabs, Inc.), etc.
- Polypeptides and polynucleotides may include the biomarkers described in 2. above, as well as components other than the biomarkers. The explanation of the biomarkers is incorporated herein by reference.
- Polypeptides and polynucleotides can be extracted according to known methods. Commercially available extraction kits may also be used.
- the anti-APLP1 monoclonal antibody may be labeled with an enzyme or a fluorescent dye.
- the antibody that binds to adipophilin may be immobilized on a microplate, magnetic beads, etc.
- Test Kit In this section, a kit for recovering extracellular vesicles containing the test reagent described in 5 above will be described.
- variable region genes full-length heavy and light chain immunoglobulin genes were created according to the method of Kurosawa et al. (Non-Patent Documents 2 and 3), and these were introduced into 293FT cells to produce recombinant antibodies.
- the reactivity of the obtained antibodies against the APLP1 peptide was analyzed by ELISA. Among these, antibody #11 showed the highest reactivity against the APLP1 peptide. The results are shown in Figure 4.
- the extract from HEK293 cells in which the human APLP1 gene was forcibly expressed was separated by SDS electrophoresis, and Western blotting was performed using #11 as the primary antibody.
- HEK293 cells in which the human APLP1 gene was forcibly expressed a single band was observed at 75 kDa, which corresponds to the molecular weight of APLP1, but no signal was observed in the negative control wild-type HEK293 cells. The results are shown in Figure 6.
- the amino acid sequence of human APLP1 (SEQ ID NO: 1) is shown in Figure 2
- the #11 light chain amino acid sequence (SEQ ID NO: 2) is shown in Table 3
- the #11 heavy chain amino acid sequence (SEQ ID NO: 3) is shown in Table 4.
- Example 1-2 (Identification of the minimal epitope for human APLP1-specific monoclonal antibodies) Fusion proteins in which the APLP1-derived peptide sequence (RSWPPGSRVE (SEQ ID NO: 20), RSWPPGSRV (SEQ ID NO: 21), WPPGSRVE (SEQ ID NO: 22), PPGSRVE (SEQ ID NO: 23), PGSRVE (SEQ ID NO: 24)) was added to the C-terminus of GFP were expressed in HEK293 cells by gene transfer.
- RSWPPGSRV SEQ ID NO: 21
- WPPGSRVE SEQ ID NO: 22
- PPGSRVE SEQ ID NO: 23
- PGSRVE SEQ ID NO: 24
- Examples 1-3 (1) APLP1 cDNA was introduced into HEK293 cells, and after 48 hours of culture, the cells were treated with 0.1% TritonX-100/PBS to obtain a cell extract. Proteins (equivalent to 10 ⁇ g) in the cell extract were separated using SDS polyacrylamide and transferred to a nitrocellulose membrane, after which APLP1 was detected using #11. The results are shown in Figure 8. A band corresponding to the molecular weight of APLP1 of 75 kDa was detected.
- Example 2 Recovery of brain neuron-derived extracellular vesicles from plasma using human APLP1-specific monoclonal antibody #11 (1) Sample Blood was collected from healthy adults using EDTA-2K tubes, and 20 mL of plasma was separated. The plasma was stored at -80°C until testing. When used, the plasma was thawed and centrifuged at 2,500 ⁇ g and 4°C for 10 minutes, and the supernatant was collected.
- Example 3 Recovery of extracellular vesicles from culture supernatant of neurons differentiated from iPS cells (1) Preparation of beads for immunoprecipitation Human APLP1-specific monoclonal antibody #11 prepared in Example 1 was biotinylated using EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) according to the protocol included in the kit, and excess biotin was removed and purified using Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) according to the protocol included in the kit.
- EZ-Link NHS-PEG4-Biotin No-Weigh Format Thermo Fisher Scientific
- the antibody-bound magnetic beads were added to 800 ⁇ L of blocking buffer containing 2% (w/v) ECL TM Prime Blocking Reagent, and incubated overnight at 4° C. to block the surface.
- Blocking was performed at room temperature for 5 minutes using Blocking One (Nacalai Tesque, Inc.).
- APLP1 C-terminus antibody (Calbiochem 171615) diluted 10,000 times in 5% Blocking One dissolved in TBS-T, CD63 antibody (Santa Cruz sc-5275) diluted 2,000 times, and CD81 antibody (Santa Cruz sc-23962) diluted 2,000 times were added and incubated overnight at 4°C with shaking. After incubation, the membrane was washed six times for 5 minutes with 0.1% TBS-T.
- Lane 1 is 1 ⁇ g of iPS neuron lysate
- lane 2 is EV before immunoprecipitation
- lane 3 is input
- lane 4 is the supernatant from immunoprecipitating EV
- lane 5 is antibody-coupled beads from immunoprecipitating EV. 1/40 of the total was electrophoresed for input and supernatant.
- Example 4 Comparison of total tau amount in CSF and NDE, comparison of total tau amount in NDE with phosphorylated tau (pTau181) and A ⁇ 42/A ⁇ 40 in CSF (1) Samples Blood was collected from patients using EDTA-2K tubes, and 10 mL of plasma was separated. The plasma was stored at -80°C until testing. When used, the plasma was thawed and centrifuged at 10,000 ⁇ g at room temperature for 5 minutes, and the supernatant was collected. 100 ⁇ L of D-PBS(-) was added to 400 ⁇ L of plasma.
- Simoa Tau 2.0 assay The supernatant collected in the previous step was dispensed into a dedicated 96-well plate, and the Tau concentration was measured using the Simoa HD-1 Analyzer instrument and the Simoa Tau 2.0 Advantage Kit according to the kit instructions. This resulted in the determination of the total amount of Tau contained in extracellular vesicles isolated from plasma using human APLP1-specific monoclonal antibody #11.
- Cerebrospinal fluid was collected by lumbar puncture on the same day as plasma was collected in (1).
- the cerebrospinal fluid was centrifuged at 430xg for 5 minutes at room temperature, aliquoted, and stored at -80°C.
- the amounts of total tau, phosphorylated tau (pTau181), A ⁇ 42, and A ⁇ 40 were measured by sandwich ELISA.
- Figure 12 shows a scatter plot showing the correlation between the measured total tau amount in plasma brain neuron-derived extracellular vesicles and the total tau in cerebrospinal fluid
- Figure 13 shows a scatter plot showing the correlation between the measured total tau amount in plasma brain neuron-derived extracellular vesicles and phosphorylated tau (pTau181) (left figure) and the A ⁇ 42/A ⁇ 40 ratio (right figure).
- the total amount of tau contained in the extracellular vesicles isolated by immunoprecipitation using human APLP1-specific monoclonal antibody #11 and plasma correlated well with the total tau, phosphorylated (pTau181), and A ⁇ 42/A ⁇ 40 ratio contained in the cerebrospinal fluid.
- APLP1-positive EVs which are extracellular vesicles derived from brain neurons, could be isolated and reflect the amount of biomarkers in the cerebrospinal fluid.
- the total amount of tau contained in the extracellular vesicles isolated from plasma using human APLP1-specific monoclonal antibody #11 could be a biomarker for neuropsychiatric disorders, and that the total amount of tau detected by this method could be used to detect neuropsychiatric disorders using plasma, which is easy to collect, as a sample instead of cerebrospinal fluid, which is difficult to collect.
- the present invention is useful in fields related to biomarkers for neuropsychiatric disorders.
- SEQ ID NO: 1 Human APLP1 amino acid sequence (NCBI accession no. NP_031493.2) SEQ ID NO:2: #11 light chain amino acid sequence SEQ ID NO:3: #11 heavy chain amino acid sequence SEQ ID NO:4: #11 light chain FR1 SEQ ID NO:5: #11 Light chain CDR1 SEQ ID NO:6: #11 Light chain FR2 SEQ ID NO:7: #11 Light chain CDR2 SEQ ID NO: 8: #11 Light chain FR3 SEQ ID NO: 9: #11 Light chain CDR3 SEQ ID NO: 10: #11 light chain FR4 SEQ ID NO: 11: #11 heavy chain FR1 SEQ ID NO: 12: #11 heavy chain CDR1 SEQ ID NO: 13: #11 heavy chain FR2 SEQ ID NO: 14: #11 heavy chain CDR2 SEQ ID NO: 15: #11 heavy chain FR3 SEQ ID NO: 16: #11 heavy chain CDR3 SEQ ID NO: 17: #11 heavy chain FR4 SEQ
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Hematology (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Urology & Nephrology (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Food Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Cell Biology (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
本発明は、抗APLP1モノクローナル抗体及びその利用に関する。
関連出願の相互参照
本出願は、2023年5月12日出願の日本特願2023-79084の優先権を主張し、その全記載は、ここに特に開示として援用される。
The present invention relates to an anti-APLP1 monoclonal antibody and uses thereof.
CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority to Japanese Patent Application No. 2023-79084, filed May 12, 2023, the entire disclosure of which is expressly incorporated herein by reference.
認知症対策は現代の最重要課題であり、バイオマーカーを利用した認知症の診断が開発されている。しかしながらそれらはPETなどの画像診断や脳脊髄液診断であり、費用面や侵襲性の問題が指摘されている。そこで、侵襲性が少なく安価な抹消血液を利用するバイオマーカーの開発が期待されている。 Measures to combat dementia are a major issue in modern times, and methods to diagnose dementia using biomarkers are being developed. However, these involve imaging diagnostics such as PET or cerebrospinal fluid diagnostics, and problems have been raised in terms of cost and invasiveness. Therefore, there is hope for the development of biomarkers that use peripheral blood, which are less invasive and less expensive.
細胞外小胞(エクソソーム)は様々な細胞から血液などの体液中に放出される。従って、脳神経細胞由来の細胞外小胞(NDE: neuron-derived extracellular vesicle)が血液中に存在することになる。NDEは脂質二重層で囲まれており、蛋白分解酵素や核酸分解酵素から内容物は保護されていて、脳細胞の環境を保存している。また、NDEの膜は脳神経細胞のそれを反映しているため、NDE膜上には神経特異的な蛋白が存在する。その一つがAPLP1(amyloid beta precursor like protein 1)である。NDE膜上にAPLP1を提示したNDEの模式図を図1に示す。 Exosomes are released from various cells into body fluids such as blood. Therefore, neuron-derived extracellular vesicles (NDEs) are present in the blood. NDEs are surrounded by a lipid bilayer, which protects the contents from proteolytic and nucleolytic enzymes and preserves the environment of brain cells. In addition, since the membrane of NDEs reflects that of brain neurons, neurospecific proteins exist on the NDE membrane. One of these is APLP1 (amyloid beta precursor like protein 1). A schematic diagram of an NDE presenting APLP1 on the NDE membrane is shown in Figure 1.
特許文献1: WO2021/132352 Patent document 1: WO2021/132352
非特許文献1: Alzheimer’s&Dementia 15(2019)1071-1080
特許文献1及び非特許文献1の全記載は、それぞれ、ここに特に開示として援用される。
Non-patent document 1: Alzheimer's & Dementia 15 (2019) 1071-1080
The entire disclosures of Patent Document 1 and Non-Patent Document 1 are each expressly incorporated herein by reference.
特許文献1には、抗APLP1抗体を用いた細胞外小胞の回収方法、回収した細胞外小胞を用いる精神神経系疾患の検出方法、検査薬及び検査キットなどが開示されている。特許文献1に記載の発明で用いられている抗APLP1抗体は市販の抗体であり、ポリクローナル抗体である。 Patent Document 1 discloses a method for recovering extracellular vesicles using an anti-APLP1 antibody, a method for detecting neuropsychiatric disorders using the recovered extracellular vesicles, a test agent, a test kit, and the like. The anti-APLP1 antibody used in the invention described in Patent Document 1 is a commercially available antibody and is a polyclonal antibody.
非特許文献1には、抗NCAM抗体(mouse anti-human neural cell adhesion molecule (NCAM))を用いたNDEの回収方法が記載されている。抗NCAM抗体は、反応性を示す対象が抗APLP1抗体とは全く異なる抗体である。 Non-Patent Document 1 describes a method for recovering NDE using an anti-NCAM antibody (mouse anti-human neural cell adhesion molecule (NCAM)). The anti-NCAM antibody is an antibody that shows reactivity with a completely different target than the anti-APLP1 antibody.
本発明は、NDEに対する特異性がより高い新たな抗体である抗APLP1モノクローナル抗体を提供し、この抗APLP1モノクローナル抗体を用いた細胞外小胞の回収方法、回収した細胞外小胞を用いる精神神経系疾患バイオマーカーの検出方法、検査薬及び検査キットなどを提供することを目的とする。 The present invention aims to provide an anti-APLP1 monoclonal antibody, which is a new antibody with higher specificity for NDE, and to provide a method for recovering extracellular vesicles using this anti-APLP1 monoclonal antibody, a method for detecting biomarkers for neuropsychiatric disorders using the recovered extracellular vesicles, a test agent, a test kit, etc.
本発明者らは、NDEの内在性膜タンパク質に着目し、かつこのタンパク質の特定部位をエピトープとして認識する複数のモノクローナル抗体を調製し、その中からNDEに対して特異的に結合する抗APLP1モノクローナル抗体を見いだし、この抗APLP1モノクローナル抗体を用いた、NDEの回収方法、回収したNDEを用いる精神神経系疾患バイオマーカーの検出方法、検査薬及び検査キットに関する発明を完成させた。 The inventors focused on the integral membrane protein of NDE and prepared multiple monoclonal antibodies that recognize specific sites of this protein as epitopes. From among these, they discovered an anti-APLP1 monoclonal antibody that specifically binds to NDE, and completed an invention relating to a method for recovering NDE using this anti-APLP1 monoclonal antibody, a method for detecting biomarkers for neuropsychiatric disorders using recovered NDE, and a test agent and test kit.
本発明は以下に示す通りである。
[1]
配列表の配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列をエピトープとする抗APLP1モノクローナル抗体。
[2]
軽鎖のCDR1、CDR2及びCDR3が配列表の配列番号5、7及び9のアミノ酸配列をそれぞれ有し、重鎖のCDR1、CDR2及びCDR3が配列表の配列番号12、14及び16に記載のアミノ酸配列をそれぞれ有する、抗APLP1モノクローナル抗体。
[3]
軽鎖のFR1、FR2、FR3及びFR4が配列表の配列番号4、6、8及び10のアミノ酸配列をそれぞれ有し、重鎖のFR1、FR2、FR3及びFR4が配列表の配列番号11、13、15及び17に記載のアミノ酸配列をそれぞれ有する、[2]に記載の抗APLP1モノクローナル抗体。
[4]
抗APLP1抗体及び細胞外小胞を含む試料を混合し、抗APLP1抗体と細胞外小胞の複合体を形成する工程、及び
前記抗APLP1抗体と細胞外小胞の複合体を回収する工程、
を含み、
抗APLP1抗体が[1]~[3]のいずれかに記載の抗APLP1モノクローナル抗体である、
脳神経細胞に由来する細胞外小胞の回収方法。
[5]
細胞外小胞を含む試料が血漿である、[4]に記載の回収方法。
[6]
[4]または[5]に記載の方法により細胞外小胞を回収し、回収された細胞外小胞から精神神経系疾患のバイオマーカーとなるポリペプチド、及び/又はポリヌクレオチドの測定値を取得する工程を含む、精神神経系疾患バイオマーカーの検出方法。
[7]
取得した測定値を対応する基準値と比較し、前記測定値が基準範囲内であるか基準範囲外であるかを決定する工程をさらに含む、[6]に記載の検出方法。
[8]
バイオマーカーがタウタンパク質またはリン酸化タウタンパク質である、[6]または[7]に記載の検出方法。
[9]
[4]または[5]に記載の方法により細胞外小胞を回収し、回収された細胞外小胞から、糖、脂質、ポリペプチド、及びポリヌクレオチドからなる群より選択される少なくとも一種の生体分子を回収する工程を含む、脳神経細胞由来成分の回収方法。
[10]
[1]~[3]のいずれかに記載の抗APLP1モノクローナル抗体を含む、細胞外小胞を回収するために使用される検査試薬。
[11]
[4]または[5]に記載の細胞外小胞の回収方法を実施するために使用される、[10]に記載の検査試薬。
[12]
[6]~[8]のいずれかに記載の精神神経系疾患バイオマーカーの検出方法を実施するために使用される、[10]に記載の検査試薬。
[13]
[9]に記載の脳神経細胞由来成分の回収方法を実施するために使用される、[10]に記載の検査試薬。
[14]
[10]に記載の検査試薬を含む、検査キット。
The present invention is as follows.
[1]
An anti-APLP1 monoclonal antibody having as its epitope the amino acid sequence from positions 232 to 238 of the amino acid sequence set forth in SEQ ID NO:1 in the sequence listing.
[2]
An anti-APLP1 monoclonal antibody, wherein the light chain CDR1, CDR2 and CDR3 have the amino acid sequences set forth in SEQ ID NOs: 5, 7 and 9, respectively, and the heavy chain CDR1, CDR2 and CDR3 have the amino acid sequences set forth in SEQ ID NOs: 12, 14 and 16, respectively.
[3]
The anti-APLP1 monoclonal antibody according to [2], wherein FR1, FR2, FR3 and FR4 of the light chain have the amino acid sequences of SEQ ID NOs: 4, 6, 8 and 10, respectively, and FR1, FR2, FR3 and FR4 of the heavy chain have the amino acid sequences of SEQ ID NOs: 11, 13, 15 and 17, respectively.
[4]
A step of mixing a sample containing an anti-APLP1 antibody and extracellular vesicles to form a complex of the anti-APLP1 antibody and the extracellular vesicles, and a step of recovering the complex of the anti-APLP1 antibody and the extracellular vesicles.
Including,
The anti-APLP1 antibody is an anti-APLP1 monoclonal antibody according to any one of [1] to [3].
A method for recovering extracellular vesicles derived from brain neurons.
[5]
The method according to [4], wherein the sample containing extracellular vesicles is plasma.
[6]
A method for detecting a biomarker for a psychiatric and neurological disorder, comprising the steps of recovering extracellular vesicles by the method according to [4] or [5], and obtaining measurements of polypeptides and/or polynucleotides that serve as biomarkers for the psychiatric and neurological disorder from the recovered extracellular vesicles.
[7]
The detection method according to [6], further comprising a step of comparing the acquired measurement value with a corresponding reference value and determining whether the measurement value is within or outside the reference range.
[8]
The detection method according to [6] or [7], wherein the biomarker is tau protein or phosphorylated tau protein.
[9]
A method for recovering components derived from brain and nerve cells, comprising the steps of recovering extracellular vesicles by the method described in [4] or [5], and recovering at least one biological molecule selected from the group consisting of sugars, lipids, polypeptides, and polynucleotides from the recovered extracellular vesicles.
[10]
A test reagent used for recovering extracellular vesicles, comprising the anti-APLP1 monoclonal antibody according to any one of [1] to [3].
[11]
The test reagent described in [10], which is used to carry out the extracellular vesicle recovery method described in [4] or [5].
[12]
The test reagent according to [10], which is used for carrying out the method for detecting a biomarker for a psychiatric and neurological disorder according to any one of [6] to [8].
[13]
The test reagent described in [10] is used to carry out the method for recovering components derived from brain nerve cells described in [9].
[14]
A test kit comprising the test reagent according to [10].
本発明によれば、NDEに対して特異的に結合する抗APLP1モノクローナル抗体を提供できる。さらに、本発明によれば、NDEを回収する方法、及び回収したNDEを用いる精神神経系疾患バイオマーカーの検出方法を提供することができる。 The present invention can provide an anti-APLP1 monoclonal antibody that specifically binds to NDE. Furthermore, the present invention can provide a method for recovering NDE and a method for detecting biomarkers for neuropsychiatric disorders using the recovered NDE.
1.抗APLP1モノクローナル抗体
本発明の抗APLP1モノクローナル抗体の第一の態様は、配列表の配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列をエピトープとする抗APLP1モノクローナル抗体である。配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列をエピトープとする抗APLP1モノクローナル抗体は、配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列を有するペプチドを抗原として認識する抗体である。配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列を有するペプチドを抗原として認識することは、抗体が、232番から238番のアミノ酸配列を有するペプチドと反応性を示すことで判定できる。抗体とペプチドとの反応性は、常法により試験することができ、常法としては、例えば、フローサイトメータ(FACS)、酵素結合免疫吸着検定法(ELISA)、western-blot、蛍光微量測定技術(FMAT)表面プラズモン共鳴(BIAcore)、免疫染色、免疫沈降などが挙げられる。
1. Anti-APLP1 Monoclonal Antibody The first aspect of the anti-APLP1 monoclonal antibody of the present invention is an anti-APLP1 monoclonal antibody having an epitope of the amino acid sequence of 232 to 238 of the amino acid sequence described in SEQ ID NO: 1 in the sequence listing. The anti-APLP1 monoclonal antibody having an epitope of the amino acid sequence of 232 to 238 of the amino acid sequence described in SEQ ID NO: 1 is an antibody that recognizes a peptide having an amino acid sequence of 232 to 238 of the amino acid sequence described in SEQ ID NO: 1 as an antigen. The recognition of a peptide having an amino acid sequence of 232 to 238 of the amino acid sequence described in SEQ ID NO: 1 as an antigen can be determined by the antibody showing reactivity with a peptide having an amino acid sequence of 232 to 238. The reactivity of the antibody with the peptide can be tested by standard methods, such as flow cytometry (FACS), enzyme-linked immunosorbent assay (ELISA), western blot, fluorescence microanalysis (FMAT), surface plasmon resonance (BIAcore), immunostaining, immunoprecipitation, and the like.
APLP1(amyloid beta precursor like protein 1)は、脳神経細胞に由来する細胞外小胞(NDE)の内在性膜タンパク質の一種であり、アミロイド前駆体タンパク質遺伝子ファミリーのメンバーの1つである。APLP1タンパク質は、脳に発現し、アミロイドベータA4前駆体タンパク質の切断と同様に、セクレターゼにより切断される膜結合糖タンパク質である。この切断により、転写活性化因子として作用する可能性のある細胞内細胞質断片が遊離する。 APLP1 (amyloid beta precursor like protein 1) is an integral membrane protein of brain neuronal-derived extracellular vesicles (NDEs) and a member of the amyloid precursor protein gene family. APLP1 protein is a membrane-bound glycoprotein expressed in the brain that is cleaved by secretases similar to the cleavage of the amyloid beta A4 precursor protein. This cleavage releases intracellular cytoplasmic fragments that may act as transcriptional activators.
脳神経細胞としては、例えば、神経細胞及びグリア細胞を含み得る。グリア細胞には、アストロサイト、オリゴデンドロサイト及びマイクログリア細胞を含み得る。細胞外小胞は、細胞から放出されるリン脂質を主成分とする膜で覆われた数十から数千nm程度の大きさを有する粒子である。 Brain nerve cells may include, for example, neurons and glial cells. Glial cells may include astrocytes, oligodendrocytes, and microglial cells. Extracellular vesicles are particles with a size of tens to thousands of nanometers that are covered with a membrane mainly composed of phospholipids and are released from cells.
ヒトAPLP1は、例えばGene ID:333で登録されている。NCBI Reference Sequenceは、例えばNM_001024807.3である。本発明においてヒトAPLP1ペプチドは、配列表の配列番号1に記載のアミノ酸配列を有するものである。 Human APLP1 is registered, for example, under Gene ID: 333. The NCBI Reference Sequence is, for example, NM_001024807.3. In the present invention, the human APLP1 peptide has the amino acid sequence set forth in SEQ ID NO: 1 in the sequence listing.
本発明の抗APLP1モノクローナル抗体の第一の態様は、配列表の配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列をエピトープとする抗APLP1モノクローナル抗体である。配列表の配列番号1に記載のアミノ酸配列の232番から238番のアミノ酸配列は、実施例で示すように、エピトープとしての最小のアミノ酸配列である。 The first embodiment of the anti-APLP1 monoclonal antibody of the present invention is an anti-APLP1 monoclonal antibody having, as an epitope, the amino acid sequence from positions 232 to 238 of the amino acid sequence set forth in SEQ ID NO:1 in the sequence listing. As shown in the examples, the amino acid sequence from positions 232 to 238 of the amino acid sequence set forth in SEQ ID NO:1 in the sequence listing is the minimum amino acid sequence that serves as an epitope.
本発明の抗APLP1モノクローナル抗体の第二の態様は、軽鎖のCDR1、CDR2及びCDR3が配列表の配列番号5、7及び9のアミノ酸配列をそれぞれ有し、重鎖のCDR1、CDR2及びCDR3が配列表の配列番号12、14及び16に記載のアミノ酸配列をそれぞれ有する、抗APLP1モノクローナル抗体である。本発明の抗APLP1モノクローナル抗体の第二の態様は、好ましくは、軽鎖のFR1、FR2、FR3及びFR4が配列表の配列番号4、6、8及び10のアミノ酸配列をそれぞれ有し、重鎖のFR1、FR2、FR3及びFR4が配列表の配列番号11、13、15及び17に記載のアミノ酸配列をそれぞれ有する。 A second embodiment of the anti-APLP1 monoclonal antibody of the present invention is an anti-APLP1 monoclonal antibody in which CDR1, CDR2, and CDR3 of the light chain have the amino acid sequences of SEQ ID NOs: 5, 7, and 9, respectively, and CDR1, CDR2, and CDR3 of the heavy chain have the amino acid sequences of SEQ ID NOs: 12, 14, and 16, respectively. In the second embodiment of the anti-APLP1 monoclonal antibody of the present invention, preferably, FR1, FR2, FR3, and FR4 of the light chain have the amino acid sequences of SEQ ID NOs: 4, 6, 8, and 10, respectively, and FR1, FR2, FR3, and FR4 of the heavy chain have the amino acid sequences of SEQ ID NOs: 11, 13, 15, and 17, respectively.
軽鎖及び重鎖の各配列番号で表示されるアミノ酸配列を以下の表1及び2に示す。 The amino acid sequences represented by the sequence numbers of the light and heavy chains are shown in Tables 1 and 2 below.
本発明の抗APLP1モノクローナル抗体は、免疫グロブリンのクラス及びサブクラスとしては特に制限されないが、例えば、IgGである。さらに、抗APLP1モノクローナル抗体は、断片(例えば、Fab、F(ab’)、F(ab)2等)や、キメラ抗体、scFv等であってもよい。 The anti-APLP1 monoclonal antibody of the present invention is not particularly limited in terms of immunoglobulin class and subclass, and may be, for example, IgG. Furthermore, the anti-APLP1 monoclonal antibody may be a fragment (e.g., Fab, F(ab'), F(ab) 2 , etc.), a chimeric antibody, scFv, etc.
本発明の抗APLP1モノクローナル抗体の製造方法
本発明の抗APLP1モノクローナル抗体は、例えば、Kurosawaら、“Rapid production of antigen-specific monoclonal antibodies from a variety of animals” BMC Biology 2021、10:80(非特許文献2)、Kurosawaら、“Novel method for the high-throughput production site-specific monoclonal antibodies”SCIENTIFIC REPORT 6:25174 DOI:10.1038 srep25174(非特許文献3)及びWO2011/027808(特許文献2)及びWO2012/133572(特許文献3)に記載の方法を用いて製造することができる。特許文献2及び3、並びに非特許文献2及び3の全記載は、それぞれ、ここに特に開示として援用される。
Method for Producing Anti-APLP1 Monoclonal Antibody of the Present Invention The anti-APLP1 monoclonal antibody of the present invention can be produced using the methods described in, for example, Kurosawa et al., “Rapid production of antigen-specific monoclonal antibodies from a variety of animals” BMC Biology 2021, 10:80 (Non-Patent Document 2), Kurosawa et al., “Novel method for the high-throughput production site-specific monoclonal antibodies” SCIENTIFIC REPORT 6:25174 DOI:10.1038 srep25174 (Non-Patent Document 3) and WO2011/027808 (Patent Document 2) and WO2012/133572 (Patent Document 3). The entire disclosures of Patent Documents 2 and 3, and Non-Patent Documents 2 and 3, respectively, are expressly incorporated herein by reference.
軽鎖及び重鎖のアミノ酸配列が特定されているモノクローナル抗体、並びに軽鎖のCD1、CD2及びCD3、FR1、FR2、FR3及びFR4並びに重鎖のCD1、CD2及びCD3、FR1、FR2、FR3及びFR4のアミノ酸配列が特定されている抗APLP1モノクローナル抗体は公知の方法により製造方法することでき、公知の方法として、非特許文献2及び3、並びに特許文献2及び3に記載の方法を挙げることができるが、これに限定される意図ではない。 Monoclonal antibodies with specific light and heavy chain amino acid sequences, as well as anti-APLP1 monoclonal antibodies with specific light chain amino acid sequences CD1, CD2, CD3, FR1, FR2, FR3, and FR4, and heavy chain CD1, CD2, CD3, FR1, FR2, FR3, and FR4, can be produced by known methods, including those described in Non-Patent Documents 2 and 3, and Patent Documents 2 and 3, but are not intended to be limited thereto.
脳神経細胞に由来する細胞外小胞の内在性膜タンパク質をエピトープとするモノクローナル抗体は、エピトープとしたいアミノ酸断片(目的抗原)で動物を免疫し、免疫した動物からリンパ液またはリンパ組織などを採取し、リンパ液またはリンパ組織などから細胞を採取し、採取した細胞からを目的抗原と反応性を示す、特異的形質細胞を回収する。回収した目的抗原特異的形質細胞からcDNAを合成し、これを鋳型としてγ鎖及びκ鎖可変領域遺伝子を増幅し、増幅した可変領域を発現ベクターに組み込み、可変領域遺伝子を得る。得られたγ鎖及びκ鎖可変領域遺伝子を用いて重鎖及軽鎖免疫グロブリン遺伝子を作成し、例えば、293FT細胞に導入して組み換え抗体を作成する。必要により、作成した複数の組み換え抗体から、目的抗原に対して反応性の高い抗体を選択することもできる。具体的な方法は実施例1に記載する。 A monoclonal antibody that uses an integral membrane protein of extracellular vesicles derived from brain nerve cells as an epitope is produced by immunizing an animal with an amino acid fragment (target antigen) that is to be used as an epitope, collecting lymph or lymphatic tissue from the immunized animal, collecting cells from the lymph or lymphatic tissue, and collecting specific plasma cells that are reactive with the target antigen from the collected cells. cDNA is synthesized from the collected target antigen-specific plasma cells, and the cDNA is used as a template to amplify the γ-chain and κ-chain variable region genes. The amplified variable regions are incorporated into an expression vector to obtain the variable region genes. The obtained γ-chain and κ-chain variable region genes are used to create heavy and light chain immunoglobulin genes, which are then introduced into, for example, 293FT cells to create a recombinant antibody. If necessary, an antibody with high reactivity to the target antigen can be selected from the multiple recombinant antibodies created. A specific method is described in Example 1.
2.細胞外小胞の回収方法
本実施形態は、脳神経細胞に由来する細胞外小胞の回収方法に関する。細胞外小胞の回収方法は、抗APLP1モノクローナル抗体(以下、抗APLP1抗体と記載することもある)と細胞外小胞を含む試料とを混合する工程と、抗APLP1抗体と細胞外小胞の複合体を回収する工程と、を含む。抗APLP1抗体と細胞外小胞を含む試料とを混合することにより、抗APLP1抗体と細胞外小胞とが複合体を形成し得る。
2. Method for recovering extracellular vesicles This embodiment relates to a method for recovering extracellular vesicles derived from brain nerve cells. The method for recovering extracellular vesicles includes a step of mixing an anti-APLP1 monoclonal antibody (hereinafter, sometimes referred to as anti-APLP1 antibody) with a sample containing extracellular vesicles, and a step of recovering a complex of the anti-APLP1 antibody and the extracellular vesicles. By mixing the anti-APLP1 antibody with a sample containing extracellular vesicles, the anti-APLP1 antibody and the extracellular vesicles can form a complex.
細胞外小胞には、エクソソーム、マイクロベシクル、アポトーシス小体等が含まれる。多くの場合、細胞外小胞には、生体分子が存在している。例えば、エクソソーム又はマイクロベシクルは、ポリペプチド及びポリペプチド(mRNA、miRNA、ノン・コーディングRNA等のRNA、及びDNA)よりなる群から選択される少なくとも一種の生体分子を含む。例えば、アポトーシス小体は、断片化された核及び細胞小器官よりなる群から選択される少なくとも一種を含む。細胞外小胞は、好ましくは、ポリペプチド及びポリペプチドよりなる群から選択される少なくとも一種の生体分子を含む。より好ましくは、細胞外小胞は、ポリペプチド及びポリペプチドよりなる群から選択される少なくとも一種の生体分子を含む。ここで、ポリペプチドは、複数のアミノ酸がペプチド結合で結合した化合物をいい、分子量の比較的大きいタンパク質及び分子量の比較的小さいペプチドを含む。 Extracellular vesicles include exosomes, microvesicles, apoptotic bodies, etc. In many cases, biomolecules are present in extracellular vesicles. For example, exosomes or microvesicles contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides (RNA such as mRNA, miRNA, non-coding RNA, and DNA). For example, apoptotic bodies contain at least one type selected from the group consisting of fragmented nuclei and organelles. Extracellular vesicles preferably contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides. More preferably, extracellular vesicles contain at least one biomolecule selected from the group consisting of polypeptides and polypeptides. Here, a polypeptide refers to a compound in which multiple amino acids are bound by peptide bonds, and includes proteins with relatively large molecular weights and peptides with relatively small molecular weights.
試料は、細胞外小胞を含む検体から粗精製した細胞外小胞を含む。試料は、細胞外小胞分散液であってもよいが、細胞外小胞のペレットであってもよい。 The sample contains extracellular vesicles roughly purified from a specimen containing extracellular vesicles. The sample may be a dispersion of extracellular vesicles, or a pellet of extracellular vesicles.
検体から、細胞外小胞を粗精製する方法は公知であり、例えばサイズ排除クロマトグラフィー法、超遠心法、アフィニティー精製法、ポリマー沈殿法及びこれらの組み合わせにより、細胞外小胞を粗精製することができる。これらの粗精製方法として、公知の方法を使用できる。サイズ排除クロマトグラフィー法は、細胞外小胞をサイズで分画できる限り制限されない。例えば、サイズ排除クロマトグラフィーは、細胞外小胞抽出キット qEV(Izon Science社)等を用いて行うことができる。超遠心法は、例えば100,000gから150,000gで2時間から3時間程度超遠心することで細胞外小胞を取得できる。好ましくは、検体は、必要に応じて、超遠心を行う際に比重が1.000から1.010程度となるようにPBS、HEPESバッファー、細胞培養用培地等で希釈することが好ましい。アフィニティー精製法は、例えば、ホスファチジルセリンアフィニティー精製法、CD63アフィニティー精製法、陰イオンアフィニティー精製法等を含み得る。ポリマー沈殿法は、ポリエチレングリコール、ポリプロピレングリコール、ポリテトラメチレングリコール等のポリエーテルを使って細胞外小胞を沈殿させる方法である。細胞外小胞の粗精製は、70nmから1000nm程度の細胞外小胞を取得できる方法で行うことが好ましい。 Methods for crudely purifying extracellular vesicles from a specimen are known, and extracellular vesicles can be crudely purified, for example, by size exclusion chromatography, ultracentrifugation, affinity purification, polymer precipitation, and combinations thereof. Known methods can be used as these crude purification methods. Size exclusion chromatography is not limited as long as it can fractionate extracellular vesicles by size. For example, size exclusion chromatography can be performed using an extracellular vesicle extraction kit qEV (Izon Science). Ultracentrifugation can be performed, for example, by ultracentrifugation at 100,000 g to 150,000 g for about 2 to 3 hours to obtain extracellular vesicles. Preferably, the specimen is diluted with PBS, HEPES buffer, cell culture medium, or the like, as necessary, so that the specific gravity is about 1.000 to 1.010 when ultracentrifuging. Affinity purification methods may include, for example, phosphatidylserine affinity purification methods, CD63 affinity purification methods, anion affinity purification methods, etc. Polymer precipitation methods are methods for precipitating extracellular vesicles using polyethers such as polyethylene glycol, polypropylene glycol, and polytetramethylene glycol. It is preferable to roughly purify extracellular vesicles using a method that can obtain extracellular vesicles of about 70 nm to 1000 nm.
検体は、動物、好ましくは、ヒト、マウス、ラット、ウサギ、イヌ、ネコ、ウシ、ブタ、ウマ等の哺乳動物の生体から採取されたものであり、細胞外小胞を含む限り制限されない。例えば、検体は、全血、血清、血漿、リンパ液、尿、腹水、胸水、脳脊髄液、細胞間質液、涙液、鼻汁、唾液等を含む。 The sample is collected from an animal, preferably a mammalian living body such as a human, mouse, rat, rabbit, dog, cat, cow, pig, or horse, and is not limited as long as it contains extracellular vesicles. For example, the sample includes whole blood, serum, plasma, lymph, urine, ascites, pleural fluid, cerebrospinal fluid, interstitial fluid, tears, nasal secretion, saliva, etc.
抗APLP1抗体は、本発明の抗APLP1モノクローナル抗体である。但し、抗体は、担体と結合した複合体であってもよい。担体として、免疫沈降に使用される公知の担体を挙げることができる。例えば、担体は、磁性ビーズ、アガロースビーズ、セルロースビーズ、マイクロプレート、チューブ等である。 The anti-APLP1 antibody is the anti-APLP1 monoclonal antibody of the present invention. However, the antibody may be a complex bound to a carrier. Examples of the carrier include known carriers used in immunoprecipitation. For example, the carrier is magnetic beads, agarose beads, cellulose beads, a microplate, a tube, etc.
抗APLP1抗体と、細胞外小胞を含む試料との混合は、抗APLP1抗体が、細胞外小胞に存在するAPLP1と結合できる条件で行われる限り、制限されない。例えば、pH6からpH9程度のTris-HClバッファー、リン酸バッファー、HEPESバッファー、マレイン酸バッファー、CHAPSバッファー等を挙げることができる。これらのバッファーには、生理食塩水と同程度の塩化ナトリウムを添加することが好ましい。また、ブロッキング試薬として、ウシ血清アルブミン、スキムミルク等を添加してもよい。反応温度は、4℃から37℃程度である。また、抗体と細胞外小胞との反応は、攪拌しながら行うことが好ましい。反応時間は、反応温度にも依存するが、反応温度が4℃程度の場合には4時間から48時間程度、反応温度が37℃程度の場合には0.5時間から4時間程度である。 The mixing of the anti-APLP1 antibody with the sample containing extracellular vesicles is not limited as long as the anti-APLP1 antibody is mixed with the APLP1 present in the extracellular vesicles under conditions that allow the antibody to bind to APLP1 present in the extracellular vesicles. Examples of the buffer include Tris-HCl buffer, phosphate buffer, HEPES buffer, maleic acid buffer, CHAPS buffer, etc., with a pH of about 6 to about 9. It is preferable to add the same amount of sodium chloride as physiological saline to these buffers. In addition, bovine serum albumin, skim milk, etc. may be added as a blocking reagent. The reaction temperature is about 4°C to 37°C. In addition, it is preferable to carry out the reaction between the antibody and the extracellular vesicles while stirring. The reaction time depends on the reaction temperature, but is about 4 to 48 hours when the reaction temperature is about 4°C, and about 0.5 to 4 hours when the reaction temperature is about 37°C.
上記反応により、抗APLP1抗体が細胞外小胞に結合し、抗APLP1抗体と細胞外小胞の複合体(「抗APLP1抗体-細胞外小胞複合体」ともいう)が形成される。 The above reaction causes the anti-APLP1 antibody to bind to the extracellular vesicles, forming a complex of the anti-APLP1 antibody and the extracellular vesicles (also called the "anti-APLP1 antibody-extracellular vesicle complex").
抗APLP1抗体-細胞外小胞複合体の回収は、公知の方法により行うことができる。抗APLP1抗体があらかじめ担体に結合している場合には、担体の性状にあわせて、回収方法を選択できる。例えば、担体が磁性ビーズである場合には、抗APLP1抗体-細胞外小胞複合体を磁石に吸着し、回収することができる。担体が、アガロースビーズ、セルロースビーズ等の非磁性ビーズである場合には、遠心分離により、抗APLP1抗体-細胞外小胞複合体を回収することができる。 The anti-APLP1 antibody-extracellular vesicle complex can be collected by known methods. When the anti-APLP1 antibody is bound to a carrier in advance, the collection method can be selected according to the properties of the carrier. For example, when the carrier is magnetic beads, the anti-APLP1 antibody-extracellular vesicle complex can be adsorbed to a magnet and collected. When the carrier is non-magnetic beads such as agarose beads or cellulose beads, the anti-APLP1 antibody-extracellular vesicle complex can be collected by centrifugation.
抗APLP1抗体があらかじめ担体に結合していない場合には、用いた抗体にアフィニティーを持つ物質、例えば、抗APLP1抗体に結合する二次抗体、プロテインA又はプロテインGと結合した担体を使用して、抗APLP1抗体-細胞外小胞複合体を回収することができる。担体、及び担体と結合した抗APLP1抗体-細胞外小胞複合体の回収方法は、抗APLP1抗体があらかじめ担体に結合している場合と同様である。 When the anti-APLP1 antibody is not bound to a carrier in advance, the anti-APLP1 antibody-extracellular vesicle complex can be recovered using a substance that has affinity for the antibody used, such as a secondary antibody that binds to the anti-APLP1 antibody, or a carrier bound to protein A or protein G. The method for recovering the carrier and the anti-APLP1 antibody-extracellular vesicle complex bound to the carrier is the same as when the anti-APLP1 antibody is bound to a carrier in advance.
抗APLP1抗体-細胞外小胞複合体の回収過程において、適宜抗APLP1抗体と未反応の細胞外小胞、及び細胞外小胞と未反応の抗APLP1抗体を除去する、B(bound)/F(free)分離を行う抗APLP1抗体-細胞外小胞複合体の洗浄工程を含んでいてもよい。 The process for recovering the anti-APLP1 antibody-extracellular vesicle complex may include a washing step of the anti-APLP1 antibody-extracellular vesicle complex by performing B (bound)/F (free) separation to remove extracellular vesicles that have not reacted with the anti-APLP1 antibody, and anti-APLP1 antibody that has not reacted with the extracellular vesicles.
APLP1は、脳神経細胞に特異的に発現するタンパク質であるため、上記回収方法により脳神経細胞に由来する細胞外小胞を回収することができる。本発明の抗APLP1モノクローナル抗体は、ヒトAPLP1ペプチドに対する特異的反応性が高いため、APLP1ペプチドを膜タンパク質として有する細胞外小胞を特異的に回収することができる。 APLP1 is a protein that is specifically expressed in brain neurons, so the above recovery method can recover extracellular vesicles derived from brain neurons. The anti-APLP1 monoclonal antibody of the present invention has high specific reactivity to human APLP1 peptide, so it can specifically recover extracellular vesicles that have APLP1 peptide as a membrane protein.
細胞外小胞の回収方法の一態様として、
(1)血漿からサイズ排除型クロマトグラフィーで全細胞外小胞を分離する、
(2)全細胞外小胞分画に対し、抗APLP1モノクローナル抗体を用いて免疫沈降を行う、
(3)得られた分画を酸処理して細胞外小胞を分離する、
ことを含む、血漿から細胞外小胞を分離する方法を挙げることができる。尚、酸処理の代りにペプチド処理をして細胞外小胞を分離することもできる。
As one embodiment of the method for recovering extracellular vesicles,
(1) Separating total extracellular vesicles from plasma by size exclusion chromatography;
(2) Immunoprecipitation of the total extracellular vesicle fraction using anti-APLP1 monoclonal antibody;
(3) treating the obtained fraction with acid to separate extracellular vesicles;
Examples of the method for separating extracellular vesicles from plasma include the following. In addition, extracellular vesicles can be separated by peptide treatment instead of acid treatment.
3.精神神経系疾患バイオマーカーの検出方法
本実施形態では、上記2.で回収された細胞外小胞を用いて、精神神経系疾患バイオマーカーの検出を行う。
3. Method for detecting biomarkers for psychiatric and neurological disorders In this embodiment, biomarkers for psychiatric and neurological disorders are detected using the extracellular vesicles collected in 2 above.
精神神経系疾患バイオマーカーの検出方法は、上記2.で回収された細胞外小胞から、精神神経系疾患のバイオマーカーのポリペプチド、及び/又はポリヌクレオチドの測定値を取得することを含む。精神神経系疾患バイオマーカーの検出方法は、さらに、前記測定値を対応する基準値と比較し、前記測定値が基準範囲内であるか基準範囲外であるかを決定する工程を含むこともできる。また、測定値が基準範囲外である場合には、検体を採取した被検体が精神神経系疾患に罹患していると決定することができる。あるいは、測定値が基準範囲内である場合には、検体を採取した患者が精神神経系疾患に罹患していないと決定することができる。 The method for detecting a biomarker for a psychiatric disorder includes obtaining a measurement value of a polypeptide and/or polynucleotide of the biomarker for a psychiatric disorder from the extracellular vesicles collected in 2 above. The method for detecting a biomarker for a psychiatric disorder may further include a step of comparing the measurement value with a corresponding reference value and determining whether the measurement value is within or outside the reference range. If the measurement value is outside the reference range, it can be determined that the subject from whom the sample was collected is suffering from a psychiatric disorder. Alternatively, if the measurement value is within the reference range, it can be determined that the patient from whom the sample was collected is not suffering from a psychiatric disorder.
さらに、測定値が、基準値からどの程度解離しているかを判定することにより、精神神経系疾患の重症度を決定してもよい。 Furthermore, the severity of the neuropsychiatric disorder may be determined by determining the extent to which the measured values deviate from the baseline values.
精神神経系疾患には、精神疾患及び神経系疾患を含み得る。神経系疾患には、例えば、神経変性疾患、脳脊髄外傷後の神経機能障害、脳腫瘍、感染に伴う脳脊髄疾患、及び多発性硬化症等が含まれ得る。神経変性疾患には、例えば、認知症、パーキンソン病、筋萎縮性側索硬化症、進行性核上麻痺、多系統萎縮症、及びトリプレットリピート病等が含まれ得る。認知症には、例えば、アルツハイマー病、老人性認知症、レビー小体病、前頭側頭型認知症、血管性認知症、アルコール性認知症及び大脳皮質基底核変性症を含み得る。感染に伴う脳脊髄疾患には、例えば、髄膜炎、脳膿瘍、クロッツフェルト-ヤコブ病、及びエイズ認知症を含み得る。脳腫瘍は、例えば、アストロサイトーマを含み得る。 Psychiatric and neurological disorders may include psychiatric disorders and neurological disorders. Neurological disorders may include, for example, neurodegenerative disorders, neurological dysfunction after brain and spinal cord trauma, brain tumors, brain and spinal cord disorders associated with infections, and multiple sclerosis. Neurodegenerative disorders may include, for example, dementia, Parkinson's disease, amyotrophic lateral sclerosis, progressive supranuclear palsy, multiple system atrophy, and triplet repeat disease. Dementia may include, for example, Alzheimer's disease, senile dementia, Lewy body disease, frontotemporal dementia, vascular dementia, alcoholic dementia, and corticobasal degeneration. Brain and spinal cord disorders associated with infections may include, for example, meningitis, brain abscess, Crotzfeldt-Jakob disease, and AIDS dementia. Brain tumors may include, for example, astrocytoma.
精神疾患には、例えば、統合失調症、うつ病、及び双極性障害等を含み得る。 Mental illnesses may include, for example, schizophrenia, depression, and bipolar disorder.
例えば、細胞外小胞に含まれるタウタンパク質、特にリン酸化タウタンパク質は、アルツハイマー病のバイオマーカーとなる。タウタンパク質、又はリン酸化タウタンパク質は、被検体から採取した検体における測定値が、基準値よりも高い場合には、アルツハイマー病であると決定することができる。 For example, tau protein, particularly phosphorylated tau protein, contained in extracellular vesicles can be a biomarker for Alzheimer's disease. If the measured value of tau protein or phosphorylated tau protein in a sample taken from a subject is higher than the reference value, it can be determined that the subject has Alzheimer's disease.
各疾患において、報告されているバイオマーカーとして、パーキンソン病及びレビー小体型認知症ではα-シヌクレイン;筋萎縮性側索硬化症及び前頭側頭型認知症ではTAR DNA-binding protein (TDP-43);クロイツフェルトヤコブ病では異常型プリオン蛋白;脳脊髄外傷後の神経機能障害、多発性硬化症、筋萎縮性側索硬化症、進行性核上麻痺、多系統萎縮症、トリプレットリピート病、アルツハイマー病、レビー小体病、前頭側頭型認知症、血管性認知症、大脳皮質基底核変性症ではNeurofilament Light Chain等を挙げることができる。また、うつ病では、インスリン様成長因子(IGF-1)及び脳由来神経栄養因子(BDNF);統合失調症では、microRNAのhsa-miR-34a、hsa-miR-432;双極性障害では、IGF-1やBDNF等を挙げることができる。 Biomarkers reported for each disease include α-synuclein for Parkinson's disease and dementia with Lewy bodies; TAR DNA-binding protein (TDP-43) for amyotrophic lateral sclerosis and frontotemporal dementia; abnormal prion protein for Creutzfeldt-Jakob disease; and Neurofilament Light Chain for neurological dysfunction after brain and spinal cord trauma, multiple sclerosis, amyotrophic lateral sclerosis, progressive supranuclear palsy, multiple system atrophy, triplet repeat disease, Alzheimer's disease, disease with Lewy bodies, frontotemporal dementia, vascular dementia, and corticobasal degeneration. Other examples include insulin-like growth factor (IGF-1) and brain-derived neurotrophic factor (BDNF) for depression; microRNAs hsa-miR-34a and hsa-miR-432 for schizophrenia; and IGF-1 and BDNF for bipolar disorder.
細胞外小胞に含まれる精神神経系疾患のバイオマーカーは、ポリペプチドとして、又はポリヌクレオチドとして検出することができる。ポリヌクレオチドは、RNAであってもよいが、DNAであってもよい、また、RNAには、mRNAの他、microRNA、ncRNA等を含み得る。精神神経系疾患のバイオマーカーのポリペプチド、及び/又はポリヌクレオチドには、完全長のものの他、断片を含み得る。 The biomarkers for neuropsychiatric disorders contained in extracellular vesicles can be detected as polypeptides or polynucleotides. The polynucleotides may be RNA or DNA, and the RNA may include not only mRNA but also microRNA, ncRNA, etc. The polypeptides and/or polynucleotides of the biomarkers for neuropsychiatric disorders may include full-length ones as well as fragments.
精神神経系疾患のバイオマーカーをポリペプチドとして検出する方法は、ウエスタンブロッティング、Enzyme-Linked Immuno Sorbent Assay(ELISA)等の公知の方法を挙げることができる。また、精神神経系疾患のバイオマーカーをRNAとして検出する方法は、RT-PCR(定量的RT-PCRを含む)、マイクロアレイ、RNA-Seq等の公知の方法を挙げることができる。精神神経系疾患のバイオマーカーをDNAとして検出する方法は、PCR(定量的PCRを含む)、マイクロアレイ、シーケンシング等の公知の方法を挙げることができる。 Methods for detecting biomarkers for psychiatric and neurological disorders as polypeptides include known methods such as Western blotting and Enzyme-Linked Immunosorbent Assay (ELISA). Methods for detecting biomarkers for psychiatric and neurological disorders as RNA include known methods such as RT-PCR (including quantitative RT-PCR), microarrays, and RNA-Seq. Methods for detecting biomarkers for psychiatric and neurological disorders as DNA include known methods such as PCR (including quantitative PCR), microarrays, and sequencing.
精神神経系疾患のバイオマーカーをポリペプチドとしてウエスタンブロッティング、ELISA等で検出する場合には、前処理として、細胞外小胞を所定の溶解バッファーで溶解する。溶解バッファーで溶解されたサンプルを検査サンプルとする。 When biomarkers for psychiatric and neurological disorders are detected as polypeptides using Western blotting, ELISA, etc., the extracellular vesicles are dissolved in a specified dissolution buffer as a pretreatment. The sample dissolved in the dissolution buffer is used as the test sample.
ウエスタンブロッティング、ELISA等によって精神神経系疾患のバイオマーカーを検出するための一次抗体は、精神神経系疾患のバイオマーカーを検出できる限り制限されない。 The primary antibody for detecting a biomarker for a psychiatric and neurological disorder by Western blotting, ELISA, etc. is not limited as long as it can detect the biomarker for a psychiatric and neurological disorder.
精神神経系疾患のバイオマーカーをRNAとしてRT-PCR、マイクロアレイ、RNA-Seq等で検出する場合には、前処理として、細胞外小胞からRNAを抽出する。また、必要に応じて、抽出したRNAを鋳型として逆転写を行い、相補的DNA(cDNA)を合成してもよい。RNA、又はcDNAをバイオマーカーの検出に使用することができる。 When biomarkers for psychiatric and neurological disorders are detected as RNA using RT-PCR, microarrays, RNA-Seq, etc., RNA is extracted from extracellular vesicles as a pretreatment. If necessary, the extracted RNA may be used as a template for reverse transcription to synthesize complementary DNA (cDNA). RNA or cDNA can be used to detect the biomarkers.
RT-PCRに使用するプライマー(定量的RT-PCRの場合にはプローブを含んでいてもよい)は市販されているものを使用することができる。また、マイクロアレイも市販されているものを使用することができる。 The primers used in RT-PCR (which may include a probe in the case of quantitative RT-PCR) can be commercially available. Commercially available microarrays can also be used.
RNA-Seqは、次世代シーケンサー(例えば、イルミナ社製)等を使用して、精神神経系疾患のバイオマーカーmRNAのリード数を得ることができる。 RNA-Seq can obtain the number of reads of biomarker mRNA for neuropsychiatric disorders using a next-generation sequencer (e.g., manufactured by Illumina).
精神神経系疾患のバイオマーカーをDNAとしてPCR、マイクロアレイ、シーケンシング等で検出する場合には、前処理として、細胞外小胞からDNAを抽出する。また、必要に応じて、抽出したDNAを鋳型として増幅反応を行ってもよい。細胞外小胞から抽出したDNA、又は増幅したDNAをバイオマーカーの検出に使用することができる。 When biomarkers for psychiatric and neurological disorders are detected as DNA using PCR, microarrays, sequencing, etc., DNA is extracted from extracellular vesicles as a pretreatment. If necessary, an amplification reaction may be performed using the extracted DNA as a template. The DNA extracted from extracellular vesicles or the amplified DNA can be used to detect the biomarkers.
PCRに使用するプライマー(定量的PCRの場合にはプローブを含んでいてもよい)は市販されているものを使用することができる。また、マイクロアレイも市販されているものを使用することができる。 Commercially available primers can be used for PCR (which may include a probe in the case of quantitative PCR). Commercially available microarrays can also be used.
シーケンシングは、次世代シーケンサー(例えば、イルミナ社製)等を使用して、精神神経系疾患のバイオマーカーに関連するDNAのリード数を得ることができる。 Sequencing can be performed using a next-generation sequencer (e.g., manufactured by Illumina) to obtain the number of DNA reads associated with biomarkers for neuropsychiatric disorders.
ウエスタンブロッティング、ELISA、RT-PCR、PCR、RNA-Seq、シーケンシング、マイクロアレイ等によって、精神神経系疾患のバイオマーカーを検出する場合、細胞外小胞において精神神経系疾患のバイオマーカーが検出された場合に、「精神神経系疾患のバイオマーカーが検出された」又は、「精神神経系疾患のバイオマーカーの発現が陽性である」と決定してもよい。あるいは、被検体の細胞外小胞に由来する精神神経系疾患のバイオマーカーと健常個体の細胞外小胞に由来する精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量を比較して、被検体に由来する検査サンプルにおける精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量が健常個体に由来する細胞外小胞の精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量よりも高値を示す場合に、「精神神経系疾患のバイオマーカーが検出された」、又は「精神神経系疾患のバイオマーカーの発現が陽性である」と決定してもよい。また、被検体に由来する細胞外小胞における精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量が健常個体に由来する細胞外小胞の精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量と同程度である場合に、「精神神経系疾患のバイオマーカーが検出されていない」、又は「精神神経系疾患のバイオマーカーの発現が陰性である」と決定してもよい。ここで、「高値を示す」とは、1.2倍以上、好ましくは1.5倍以上、より好ましくは2倍以上、さらに好ましくは5倍以上高い値を示す場合を例示できる。「同程度」とは、0.8倍から1.2倍未満程度を例示できる。また、精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量を比較する前に、各検体から精製した細胞外小胞量を、細胞外小胞のマーカーである、CD9、CD63、CD81等のポリペプチド量で正規化してもよい。また、細胞外小胞数の正規化は、Nanoparticle Tracking Analysis法等で測定した粒子数等で行ってもよい。ポリペプチド量は質量、又は濃度で表されてもよいが、基質の発光強度等で表されてもよい。ポリヌクレオチド量は、ポリヌクレオチドのコピー数又はリード数であってもよいが、蛍光強度等で表されてもよい。 When a biomarker for a neuropsychiatric disorder is detected by Western blotting, ELISA, RT-PCR, PCR, RNA-Seq, sequencing, microarray, or the like, if the biomarker for a neuropsychiatric disorder is detected in extracellular vesicles, it may be determined that "the biomarker for a neuropsychiatric disorder has been detected" or that "the expression of the biomarker for a neuropsychiatric disorder is positive." Alternatively, by comparing the amount of the polypeptide or the amount of the polynucleotide of the biomarker for a neuropsychiatric disorder derived from the extracellular vesicles of a subject with the amount of the polypeptide or the polynucleotide of the biomarker for a neuropsychiatric disorder derived from the extracellular vesicles of a healthy individual, it may be determined that "the biomarker for a neuropsychiatric disorder has been detected" or that "the expression of the biomarker for a neuropsychiatric disorder is positive." If the amount of the polypeptide or the amount of the polynucleotide of the biomarker for a neuropsychiatric disorder in the test sample derived from the subject is higher than the amount of the polypeptide or the polynucleotide of the biomarker for a neuropsychiatric disorder in the extracellular vesicles derived from the healthy individual, it may be determined that "the biomarker for a neuropsychiatric disorder has been detected" or that "the expression of the biomarker for a neuropsychiatric disorder is positive." In addition, when the amount of the polypeptide of the biomarker for a neuropsychiatric disease or the amount of the polynucleotide of the biomarker for a neuropsychiatric disease in the extracellular vesicles derived from the subject is comparable to the amount of the polypeptide of the biomarker for a neuropsychiatric disease or the amount of the polynucleotide of the biomarker for a neuropsychiatric disease in the extracellular vesicles derived from a healthy individual, it may be determined that "the biomarker for a neuropsychiatric disease is not detected" or that "the expression of the biomarker for a neuropsychiatric disease is negative." Here, "showing a high value" can be exemplified by a value that is 1.2 times or more, preferably 1.5 times or more, more preferably 2 times or more, and even more preferably 5 times or more higher. "Similar" can be exemplified by about 0.8 times to less than 1.2 times. In addition, before comparing the amount of the polypeptide of the biomarker for a neuropsychiatric disease or the amount of the polynucleotide of the biomarker for a neuropsychiatric disease, the amount of extracellular vesicles purified from each specimen may be normalized by the amount of the polypeptide of CD9, CD63, CD81, etc., which are markers of extracellular vesicles. The number of extracellular vesicles may be normalized based on the number of particles measured by the Nanoparticle Tracking Analysis method or the like. The amount of polypeptide may be expressed as mass or concentration, or may be expressed as the luminescence intensity of the substrate. The amount of polynucleotide may be the copy number or read number of the polynucleotide, or may be expressed as fluorescence intensity or the like.
別の態様として、精神神経系疾患のバイオマーカーポリペプチド量又はRNA量の基準値をあらかじめ決定しておき、被検体に由来する細胞外小胞における精神神経系疾患のバイオマーカーポリペプチド量又はRNA量が基準範囲外である場合に、「精神神経系疾患のバイオマーカーが検出された」、又は「精神神経系疾患のバイオマーカーの発現が陽性である」と決定してもよい。また、被検体に由来する細胞外小胞に由来する精神神経系疾患のバイオマーカーポリペプチド量又はRNA量が基準範囲内である場合に、「精神神経系疾患のバイオマーカーが検出されない」又は、「精神神経系疾患のバイオマーカーの発現が陰性である」と決定してもよい。基準値は、精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量が検出されているか、又は発現が陽性であるかを判別できる値である限り制限されず、公知の方法により決定することができる。精神神経系疾患のバイオマーカーのポリペプチド量、又は精神神経系疾患のバイオマーカーのポリヌクレオチド量が検出されているか、又は発現が陽性であるかを判別できる値は、ROC(receiver operating characteristic curve)曲線、判別分析法、モード法、Kittler法、3σ法、p‐tile法等により決定することもできる。また、基準値として、感度、特異度、陰性的中率、陽性的中率、第一四分位数等を例示できる。 In another embodiment, a reference value for the amount of a biomarker polypeptide or RNA for a neuropsychiatric disorder may be determined in advance, and when the amount of a biomarker polypeptide or RNA for a neuropsychiatric disorder in extracellular vesicles derived from a subject is outside the reference range, it may be determined that a "biomarker for a neuropsychiatric disorder has been detected" or that the expression of a biomarker for a neuropsychiatric disorder is positive. In addition, when the amount of a biomarker polypeptide or RNA for a neuropsychiatric disorder derived from extracellular vesicles derived from a subject is within the reference range, it may be determined that a "biomarker for a neuropsychiatric disorder has not been detected" or that the expression of a biomarker for a neuropsychiatric disorder is negative. The reference value is not limited as long as it is a value that can determine whether the amount of a polypeptide for a biomarker for a neuropsychiatric disorder or the amount of a polynucleotide for a biomarker for a neuropsychiatric disorder has been detected or that the expression is positive, and can be determined by a known method. The value that can determine whether the amount of a polypeptide of a biomarker for a neuropsychiatric disorder or the amount of a polynucleotide of a biomarker for a neuropsychiatric disorder is detected or whether expression is positive can also be determined by a receiver operating characteristic curve (ROC) curve, discriminant analysis method, mode method, Kittler method, 3σ method, p-tile method, etc. Examples of reference values include sensitivity, specificity, negative predictive value, positive predictive value, first quartile, etc.
精神神経系疾患バイオマーカーの検出方法の一態様として、前記した血漿から細胞外小胞を分離する方法において得られた細胞外小胞内の全タウを超高感度デジタルELISAで測定することを含む、細胞外小胞内の全タウの測定方法を挙げることができる。 One embodiment of the method for detecting biomarkers for psychiatric and neurological disorders is a method for measuring the total amount of tau in extracellular vesicles, which includes measuring the total amount of tau in extracellular vesicles obtained by the method for isolating extracellular vesicles from plasma using an ultrasensitive digital ELISA.
4.脳神経細胞由来成分の回収方法
本実施形態では、上記2.で回収された細胞外小胞を用いて、脳神経細胞由来成分の回収を行う。
4. Method for recovering components derived from brain nerve cells In this embodiment, components derived from brain nerve cells are recovered using the extracellular vesicles recovered in 2 above.
脳神経細胞由来成分の回収方法は、上記1.で回収された細胞外小胞から、糖、脂質、ポリペプチド、及びポリヌクレオチドからなる群より選択される少なくとも一種の生体分子を回収する工程を含む。 The method for recovering components derived from brain nerve cells includes a step of recovering at least one biomolecule selected from the group consisting of sugars, lipids, polypeptides, and polynucleotides from the extracellular vesicles recovered in 1 above.
糖には、単糖、二糖、オリゴ糖、ポリサッカライドを含み得る。また、糖は、脂質、又はタンパク質等と結合していてもよい。細胞外小胞からの糖の回収は、ヒドラジド・オキシアミン担持ポリマー、レクチン等を使用する公知の方法により行うことができる。 The sugars may include monosaccharides, disaccharides, oligosaccharides, and polysaccharides. The sugars may also be bound to lipids, proteins, or the like. The sugars can be recovered from the extracellular vesicles by known methods using hydrazide-oxyamine-bearing polymers, lectins, and the like.
脂質には、脂肪酸、エイコサノイド、トリアシルグリセロール、ろうエステル、リン脂質、スフィンゴ脂質、イソプレノイド、リポタンパク質等を含み得る。細胞外小胞からの脂質の抽出は、Folch法、脂質抽出キット(Lipid Extraction Kit:Cell Biolabs, Inc.)等を使用して行うことができる。 Lipids may include fatty acids, eicosanoids, triacylglycerols, wax esters, phospholipids, sphingolipids, isoprenoids, lipoproteins, etc. Lipids can be extracted from extracellular vesicles using the Folch method, a lipid extraction kit (Cell Biolabs, Inc.), etc.
ポリペプチド、及びポリヌクレオチドには、上記2.で述べたバイオマーカーと、上記バイオマーカー以外の成分も含みうる。バイオマーカーの説明は、ここに援用する。 Polypeptides and polynucleotides may include the biomarkers described in 2. above, as well as components other than the biomarkers. The explanation of the biomarkers is incorporated herein by reference.
ポリペプチド、及びポリヌクレオチドの回収は、公知の方法にしたがって行うことができる。また、市販の抽出キットを使用してもよい。 Polypeptides and polynucleotides can be extracted according to known methods. Commercially available extraction kits may also be used.
5.検査試薬
本項において、細胞外小胞を回収するための検査試薬について説明する。検査試薬は、本発明の抗APLP1モノクローナル抗体を含む。抗APLP1モノクローナル抗体の説明は、上記1.の説明をここに援用する。
5. Test Reagent In this section, a test reagent for recovering extracellular vesicles will be described. The test reagent contains the anti-APLP1 monoclonal antibody of the present invention. The description of the anti-APLP1 monoclonal antibody is incorporated herein by reference in the above 1.
検査試薬に含まれる抗APLP1モノクローナル抗体は、1種であっても2種以上であってもよい。 The test reagent may contain one or more types of anti-APLP1 monoclonal antibodies.
検査試薬に含まれる抗APLP1モノクローナル抗体は、乾燥状態であってもよく、リン酸緩衝生理食塩水等のバッファーに溶解されていてもよい。さらに、検査試薬は、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。 The anti-APLP1 monoclonal antibody contained in the test reagent may be in a dry state or may be dissolved in a buffer such as phosphate-buffered saline. Furthermore, the test reagent may contain at least one of the following: a stabilizer such as β-mercaptoethanol or DTT; a protective agent such as albumin; a surfactant such as polyoxyethylene (20) sorbitan monolaurate or polyoxyethylene (10) octylphenyl ether; or a preservative such as sodium azide.
抗APLP1モノクローナル抗体は、酵素や蛍光色素で標識されていてもよい。アディポフィリンと結合する抗体はマイクロプレート、磁性ビーズ等に固定されていてもよい。 The anti-APLP1 monoclonal antibody may be labeled with an enzyme or a fluorescent dye. The antibody that binds to adipophilin may be immobilized on a microplate, magnetic beads, etc.
6.検査キット
本項において、細胞外小胞を回収するための、上記5.で述べた検査試薬を含むキットについて説明する。
6. Test Kit In this section, a kit for recovering extracellular vesicles containing the test reagent described in 5 above will be described.
検査キットは、検査試薬と試薬の使用方法を記載した又は試薬の使用方法を記載するウェブページのURLが記載された添付文書を含む検査キットとして提供されてもよい。また、抗APLP1抗体が、担体や固相と結合していない場合には、担体や固相と抗APLP1抗体を結合するための、二次抗体結合ビーズ、プロテインAビーズ、プロテインGビーズ、二次抗体固相化マイクロプレート、プロテインA固相化マイクロプレート、プロテインG固相化マイクロプレート、二次抗体固相化チューブ等を含んでいてもよい。 The test kit may be provided as a test kit including a test reagent and a package insert that describes how to use the reagent or that lists the URL of a web page that describes how to use the reagent. In addition, when the anti-APLP1 antibody is not bound to a carrier or solid phase, the test kit may include secondary antibody-bound beads, protein A beads, protein G beads, secondary antibody-immobilized microplates, protein A-immobilized microplates, protein G-immobilized microplates, secondary antibody-immobilized tubes, etc., for binding the anti-APLP1 antibody to a carrier or solid phase.
さらに、検査キットは、細胞外小胞を粗精製するための、サイズ排除クロマトグラフィー用カラム、アフィニティー精製用カラム、ポリエチレングリコール等のポリエーテル等を含んでいてもよい。 Furthermore, the test kit may contain a size exclusion chromatography column, an affinity purification column, a polyether such as polyethylene glycol, etc., for crude purification of extracellular vesicles.
さらにまた、細胞外小胞に存在する精神神経系疾患のバイオマーカーを検出するための抗体、又はプローブ、プライマー等を含んでいてもよい。 Furthermore, the sample may contain antibodies, probes, primers, etc. for detecting biomarkers for neuropsychiatric disorders present in the extracellular vesicles.
以下、本発明を実施例に基づいて更に詳細に説明する。但し、実施例は本発明の例示であって、本発明は実施例に限定される意図ではない。 The present invention will be described in more detail below based on examples. However, the examples are merely illustrative of the present invention, and the present invention is not intended to be limited to the examples.
実施例1-1
(ヒトAPLP1特異的モノクローナル抗体の作製)
(A)ヒトAPLP1特異的モノクローナル抗体を作製するため、ヒトAPLP1ペプチド(GTAVGDPSTRSWPPGSRVEG)(配列番号1)を免疫したモルモットから腸骨リンパ節を採取し、これより細胞懸濁液を調製した。細胞(1x107個)を2%パラフォルムアルデヒドPBS溶液にて4℃で10分間の固定を行った(固定化)。
Example 1-1
(Generation of human APLP1-specific monoclonal antibodies)
(A) To prepare a human APLP1-specific monoclonal antibody, iliac lymph nodes were collected from guinea pigs immunized with human APLP1 peptide (GTAVGDPSTRSWPPGSRVEG) (SEQ ID NO: 1) and a cell suspension was prepared from the lymph nodes. The cells ( 1x107 cells) were fixed in 2% paraformaldehyde in PBS at 4°C for 10 minutes (fixation).
遠心にて細胞を回収し、これに250μlの染色液(抗モルモットIgG-Dylight650,APLP1ペプチド-ストレプトアビジンDylight488, ストレプトアビジンDylight550、および200unitのRNaseOUTを含むPBST)を加え、氷上にて15分間放置することで、染色を行った(細胞膜溶解及び標識目的抗原反応)。 Cells were collected by centrifugation, and 250 μl of staining solution (PBST containing anti-guinea pig IgG-Dylight650, APLP1 peptide-streptavidin Dylight488, streptavidin Dylight550, and 200 units of RNaseOUT) was added to the cells, and the cells were left on ice for 15 minutes for staining (cell membrane lysis and reaction of the antigen to be labeled).
本溶液に0.2μg/mlのDAPIを含むPBS溶液を3ml加え細胞核の染色を行った。 3 ml of PBS solution containing 0.2 μg/ml DAPI was added to this solution to stain the cell nuclei.
これらの細胞を、フローサイトメータを用いて解析し、APLP1ペプチド特異的形質細胞の分離を行った。R1ゲート(FSC vs SSC)およびR2ゲート(DAPI)を設定することで、単一細胞集団を同定した。更に単一細胞集団から、自己蛍光を有する細胞、非特異的染色を受けた細胞をFL2チャンネルによって除去した(R3ゲート)。これらの細胞のなかで、抗マウスIgG 強陽性、APLP1ペプチド強陽性、ストレプトアビジンDylight550陰性細胞を、APLP1ペプチド特異的形質細胞として同定した(R4ゲート)。結果を図3に示す。 These cells were analyzed using a flow cytometer to separate APLP1 peptide-specific plasma cells. Single cell populations were identified by setting the R1 gate (FSC vs SSC) and R2 gate (DAPI). Autofluorescent cells and non-specifically stained cells were further removed from the single cell population using the FL2 channel (R3 gate). Among these cells, cells that were strongly anti-mouse IgG positive, strongly APLP1 peptide positive, and streptavidin Dylight550 negative were identified as APLP1 peptide-specific plasma cells (R4 gate). The results are shown in Figure 3.
(C)R4ゲート内の細胞を、シングルセルソーティングを行った(細胞単離)。その後、セルソーターで分離された細胞からKurosawaらの方法(非特許文献2及び3)に従ってcDNAを合成し、これを鋳型として5'-RACE PCR法によりラットγ鎖及びκ鎖可変領域遺伝子を増幅した。増幅された可変領域を線状化発現ベクターに組み込み、モルモットγ鎖及びκ鎖免疫グロブリン発現ユニットを作製した。増幅されたDNAのアガロース電気泳動の結果を示した。 (C) Single cell sorting was performed on the cells in the R4 gate (cell isolation). After that, cDNA was synthesized from the cells separated by the cell sorter according to the method of Kurosawa et al. (Non-Patent Documents 2 and 3), and the rat γ-chain and κ-chain variable region genes were amplified using this as a template by 5'-RACE PCR. The amplified variable regions were incorporated into a linearized expression vector to create guinea pig γ-chain and κ-chain immunoglobulin expression units. The results of agarose electrophoresis of the amplified DNA are shown.
得られた可変領域遺伝子より、Kurosawaらの方法(非特許文献2及び3)に従って、全長の重鎖、軽鎖免疫グロブリン遺伝子を作成し、これを293FT細胞に導入することで組み換え抗体を作成した。得られた抗体のAPLP1ペプチドに対する反応性をELISA法にて解析した。このうち#11がAPLP1ペプチドに対して最も高い反応性を示した。結果を図4に示す。 From the obtained variable region genes, full-length heavy and light chain immunoglobulin genes were created according to the method of Kurosawa et al. (Non-Patent Documents 2 and 3), and these were introduced into 293FT cells to produce recombinant antibodies. The reactivity of the obtained antibodies against the APLP1 peptide was analyzed by ELISA. Among these, antibody #11 showed the highest reactivity against the APLP1 peptide. The results are shown in Figure 4.
得られたAPLP1ペプチド陽性抗体クローンを用いて内在性のAPLP1を発現するヒト神経芽細胞腫であるSHSY-5Y細胞、及びSHSY-5Y細胞のAPLP1遺伝子ノックアウト細胞を用いて免疫染色を行った。#11はSHSY-5Y細胞の細胞質にドット状の局在を示したが、ノックアウト細胞では、シグナルが認められなかった結果を図5に示す。 Using the obtained APLP1 peptide-positive antibody clone, immunostaining was performed on SHSY-5Y cells, a human neuroblastoma that expresses endogenous APLP1, and on APLP1 gene knockout SHSY-5Y cells. #11 showed dot-like localization in the cytoplasm of SHSY-5Y cells, but no signal was observed in the knockout cells. The results are shown in Figure 5.
ヒトAPLP1遺伝子を強制発現させたHEK293細胞抽出液をSDS電気泳動法により分離し、一次抗体として#11を用いたウエスタンブロッティングを行った。ヒトAPLP1遺伝子を強制発現させたHEK293細胞では、APLP1の分子量に相当する75kDaの位置に単一のバンドが認めれれたが、陰性コントロールの野生型HEK293細胞ではシグナルが観察されなかった。結果を図6に示す。 The extract from HEK293 cells in which the human APLP1 gene was forcibly expressed was separated by SDS electrophoresis, and Western blotting was performed using #11 as the primary antibody. In HEK293 cells in which the human APLP1 gene was forcibly expressed, a single band was observed at 75 kDa, which corresponds to the molecular weight of APLP1, but no signal was observed in the negative control wild-type HEK293 cells. The results are shown in Figure 6.
ヒトAPLP1のアミノ酸配列(配列番号1)を図2に示し、#11軽鎖アミノ酸配列(配列番号2)を表3、#11重鎖アミノ酸配列(配列番号3)を表4に示す。 The amino acid sequence of human APLP1 (SEQ ID NO: 1) is shown in Figure 2, the #11 light chain amino acid sequence (SEQ ID NO: 2) is shown in Table 3, and the #11 heavy chain amino acid sequence (SEQ ID NO: 3) is shown in Table 4.
実施例1-2
(ヒトAPLP1特異的モノクローナル抗体の最小エピトープの特定)
APLP1由来のペプチド配列(RSWPPGSRVE(配列番号20)、RSWPPGSRV(配列番号21)、WPPGSRVE(配列番号22)、PPGSRVE(配列番号23)、PGSRVE(配列番号24))をGFPのC末に付加した融合タンパク質を遺伝子導入によりHEK293細胞に発現させた。培養24時間後に細胞を4%ホルマリン/PBSにて固定し、0.1% TritonX-100/PBSにて膜可溶化を行なった後、#11 (0.1μg/mLPBS) にて1時間反応させた。抗モルモット抗体(Dylight 650)にて細胞染色を行うことで、シグナルを検出した。結果を図7に示す。
Example 1-2
(Identification of the minimal epitope for human APLP1-specific monoclonal antibodies)
Fusion proteins in which the APLP1-derived peptide sequence (RSWPPGSRVE (SEQ ID NO: 20), RSWPPGSRV (SEQ ID NO: 21), WPPGSRVE (SEQ ID NO: 22), PPGSRVE (SEQ ID NO: 23), PGSRVE (SEQ ID NO: 24)) was added to the C-terminus of GFP were expressed in HEK293 cells by gene transfer. After 24 hours of culture, the cells were fixed with 4% formalin/PBS, membranes were solubilized with 0.1% TritonX-100/PBS, and then reacted with #11 (0.1 μg/mL PBS) for 1 hour. Signals were detected by staining the cells with an anti-guinea pig antibody (Dylight 650). The results are shown in Figure 7.
結果
GFPにペプチドRSWPPGSRVE を付加した場合ではGFP陽性細胞が#11陽性となっているが、GFPにペプチドRSWPPGSRV を付加した場合では、GFP陽性細胞は#11陽性となっていない。このことから、#11が認識するC末エピトープにはEが必要であることが判明した。
Results: When the peptide RSWPPGSRVE was added to GFP, GFP-positive cells were positive for #11, but when the peptide RSWPPGSRV was added to GFP, GFP-positive cells were not positive for #11. This indicates that E is required for the C-terminal epitope recognized by #11.
次に、GFPにペプチドPPGSRVEを付加した場合はGFP陽性細胞が#11陽性となっているが、GFPにペプチドPGSRVEを付加した場合ではGFP陽性細胞が#11陽性となっていない。以上の結果から#11が認識する最小エピトープはPPGSRVEであることが明らかになった。 Next, when the peptide PPGSRVE was added to GFP, GFP-positive cells were positive for #11, but when the peptide PGSRVE was added to GFP, GFP-positive cells were not positive for #11. These results demonstrated that the minimal epitope recognized by #11 is PPGSRVE.
実施例1-3
(1)APLP1 cDNAをHEK293細胞に導入し、培養48時間後の細胞を0.1% TritonX-100/PBSにて処理することで細胞抽出液を得た。細胞抽出液中のタンパク質(10μg相当)をSDSポリアクリルアミドにて分離し、ニトロセルロース膜に転写後、#11を用いてAPLP1を検出した。結果を図8に示す。75kDaのAPLP1の分子量に相当するバンドが検出された。
Examples 1-3
(1) APLP1 cDNA was introduced into HEK293 cells, and after 48 hours of culture, the cells were treated with 0.1% TritonX-100/PBS to obtain a cell extract. Proteins (equivalent to 10 μg) in the cell extract were separated using SDS polyacrylamide and transferred to a nitrocellulose membrane, after which APLP1 was detected using #11. The results are shown in Figure 8. A band corresponding to the molecular weight of APLP1 of 75 kDa was detected.
(2)SHSY-5Y細胞を4%ホルマリン/PBSにて固定し、0.1% TritonX-100/PBSにて膜可溶化を行なった後、#11 (0.1μg/mL PBS) にて12時間反応させた。抗モルモット抗体(Dylight 650)にて細胞染色を行うことで、シグナルを検出した。結果を図9に示す。 (2) SHSY-5Y cells were fixed in 4% formalin/PBS, membranes were solubilized in 0.1% TritonX-100/PBS, and then reacted with #11 (0.1 μg/mL PBS) for 12 hours. Signals were detected by cell staining with anti-guinea pig antibody (Dylight 650). The results are shown in Figure 9.
実施例2:ヒトAPLP1特異的モノクローナル抗体#11を用いた血漿からの脳神経細胞由来細胞外小胞の回収
(1)検体
健常成人からEDTA-2Kチューブを使って採血し、20 mLの血漿を分離した。血漿は検査まで、-80℃下で保存した。使用時に、血漿を融解し、2,500×g, 4℃で10分間遠心し、上清を回収した。
Example 2: Recovery of brain neuron-derived extracellular vesicles from plasma using human APLP1-specific monoclonal antibody #11 (1) Sample Blood was collected from healthy adults using EDTA-2K tubes, and 20 mL of plasma was separated. The plasma was stored at -80°C until testing. When used, the plasma was thawed and centrifuged at 2,500×g and 4°C for 10 minutes, and the supernatant was collected.
(2)血漿細胞外小胞の粗精製
qEV10(Izon Science社)を用いて、キット添付プロトコルにしたがって、血漿10mLからサイズ排除クロマトグラフィー(SEC)分画 20 mLを回収した。上記を計2回実施し、SEC分画 40mLを回収した。Amicon Ultra-15 100kDa MWCOを用いてSEC分画を3,000μLまで濃縮した。
(2) Crude purification of plasma extracellular vesicles Using qEV10 (Izon Science), 20 mL of size exclusion chromatography (SEC) fraction was collected from 10 mL of plasma according to the kit protocol. The above procedure was repeated twice, and 40 mL of SEC fraction was collected. The SEC fraction was concentrated to 3,000 μL using an Amicon Ultra-15 100 kDa MWCO.
(3)免疫沈降用ビーズの作製
EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) を用いて、実施例1で調製したヒトAPLP1特異的モノクローナル抗体#11をキット付属のプロトコルに従いビオチン化し、Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) をキット付属のプロトコルに従って使用し、余分なビオチンを除去・精製した。
(3) Preparation of beads for immunoprecipitation Human APLP1-specific monoclonal antibody #11 prepared in Example 1 was biotinylated using EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) according to the protocol included with the kit, and excess biotin was removed and purified using Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) according to the protocol included with the kit.
Dynabeads(登録商標) MyOne Streptavidin C1 (Thermo Fisher Scientific) 20μLと10μgのビオチン化したヒトAPLP1特異的モノクローナル抗体#11を 0.01% Tween-20入りのDulbecco’s PBS内で混合し、ヒトAPLP1特異的モノクローナル抗体#11が結合した磁性ビーズを作製した。 20 μL of Dynabeads® MyOne Streptavidin C1 (Thermo Fisher Scientific) and 10 μg of biotinylated human APLP1-specific monoclonal antibody #11 were mixed in Dulbecco's PBS containing 0.01% Tween-20 to produce magnetic beads bound to human APLP1-specific monoclonal antibody #11.
更に、ECLTM Prime Blocking Reagentを2%(w/v)含むブロッキングバッファー 800μL中に抗体結合磁気ビーズを加えて4℃下で1晩インキュベートし、表面のブロッキングを行った。 Furthermore, the antibody-bound magnetic beads were added to 800 μL of blocking buffer containing 2% (w/v) ECL ™ Prime Blocking Reagent, and incubated overnight at 4° C. to block the surface.
(4)脳神経細胞由来細胞外小胞の免疫沈降
上記(3)で作製した免疫沈降用ビーズに濃縮したSEC分画 1,444μLを添加し、4℃下で6時間転倒混和しながらインキュベーションした。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。続いて、磁性ビーズを洗浄するため、D-PBS(-) 800μLをチューブに加えて転倒混和した。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。再度スピンダウンして磁気スタンドに装着し1分間待ち、上清を完全に取り除いた。
(4) Immunoprecipitation of brain neuron-derived extracellular vesicles 1,444 μL of concentrated SEC fraction was added to the immunoprecipitation beads prepared in (3) above, and incubated at 4°C for 6 hours with inversion mixing. The tube was spun down, attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. Next, to wash the magnetic beads, 800 μL of D-PBS(-) was added to the tube and mixed by inversion. The tube was spun down, attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. The tube was spun down again, attached to a magnetic stand, waited for 1 minute, and the supernatant was completely removed.
磁性ビーズが残ったチューブに0.1M glycine-HCl (pH 2.8) 17.4μLを加え、ボルテックス後、4℃で10分間インキュベートした。チューブをスピンダウンした後、磁気スタンドに装着し1分間待ち、上清を回収し、1M Tris-HCl (pH 8.0) 2.6μL加えてpHを調整し、解析に供した。 17.4μL of 0.1M glycine-HCl (pH 2.8) was added to the tube containing the remaining magnetic beads, which was then vortexed and incubated at 4°C for 10 minutes. The tube was then spun down, placed on a magnetic stand and left for 1 minute. The supernatant was then collected and 2.6μL of 1M Tris-HCl (pH 8.0) was added to adjust the pH before preparation for analysis.
(5)Western Blot
前ステップで回収した上清を変性バッファーと混合した後、加熱し、アプライ用のサンプルを調製した。サンプルを5-15% Tris-glycine SDS gelにロードし、200Vで55分間泳動した。泳動が終わったSDSゲルを、セミドライ条件で、Towbin Buffer (5%メタノール入り)を用いて15Vで30分間泳動し、ゲルからPVDFメンブレンにトランスファーした。
(5) Western Blot
The supernatant collected in the previous step was mixed with a denaturing buffer and heated to prepare a sample for application. The sample was loaded onto a 5-15% Tris-glycine SDS gel and electrophoresed at 200 V for 55 minutes. After electrophoresis, the SDS gel was electrophoresed at 15 V for 30 minutes in Towbin Buffer (containing 5% methanol) under semi-dry conditions, and the gel was transferred to a PVDF membrane.
Blocking One (ナカライテスク株式会社)を用いて、室温で5分間ブロッキングした。TBS-Tに溶解した5% Blocking Oneで一次抗体を希釈し、4℃で一晩反応させた。使用した抗体は、抗APLP1抗体 (R&D Systems, AF3129)、抗CD81抗体 (Cosmo Bio, SHI-EXO-M03)、抗CD9抗体 (Cosmo Bio, SHI-EXO-M01)であり、抗APLP1抗体および抗CD81抗体は2,000倍希釈、抗CD9抗体は1,000倍希釈して用いた。一次抗体を反応させた後、TBS-Tを使って、1回5分の洗浄を6回行った。 Blocking was performed at room temperature for 5 minutes using Blocking One (Nacalai Tesque, Inc.). Primary antibodies were diluted with 5% Blocking One dissolved in TBS-T and incubated overnight at 4°C. The antibodies used were anti-APLP1 antibody (R&D Systems, AF3129), anti-CD81 antibody (Cosmo Bio, SHI-EXO-M03), and anti-CD9 antibody (Cosmo Bio, SHI-EXO-M01). Anti-APLP1 antibody and anti-CD81 antibody were diluted 2,000-fold, and anti-CD9 antibody was diluted 1,000-fold. After incubation with the primary antibodies, the cells were washed six times with TBS-T for 5 minutes each.
TBS-Tに溶解した5% Blocking Oneで二次抗体を希釈し、室温で1時間反応させた。使用した抗体は、HRP標識抗マウス抗体 (Promega, W4021)、HRP標識抗ラビット抗体 (Promega, W4011)であり、全て10,000希釈して用いた。TBS-Tを使って、1回5分の洗浄を6回行った。 The secondary antibody was diluted with 5% Blocking One dissolved in TBS-T and incubated at room temperature for 1 hour. The antibodies used were HRP-labeled anti-mouse antibody (Promega, W4021) and HRP-labeled anti-rabbit antibody (Promega, W4011), all diluted 10,000. Six washes were performed using TBS-T for 5 minutes each.
メンブレンにImmunoStar(登録商標) LD A+B液 1,400μLを加え、室温で5分間反応させた。MyECL Imager (Thermo Scientific)でHRPを発光させ、シグナルを撮像した。 1,400μL of ImmunoStar (registered trademark) LD A+B solution was added to the membrane and allowed to react at room temperature for 5 minutes. HRP was induced to emit light using a MyECL Imager (Thermo Scientific) and the signal was imaged.
(6)結果
Western Blotの結果を図10-1に示す。
(6) Results The results of Western Blot are shown in FIG. 10-1.
血漿を用いた免疫沈降により、標的であるAPLP1に加えて、EVタンパク質であるCD81とCD9が検出された。このことから、特異的モノクローナル抗体#11を用いることで脳神経細胞由来細胞外小胞であるAPLP1陽性EVが分離できていることが明らかとなった。 In addition to the target APLP1, EV proteins CD81 and CD9 were detected by immunoprecipitation using plasma. This demonstrated that APLP1-positive EVs, which are extracellular vesicles derived from brain neurons, can be isolated by using specific monoclonal antibody #11.
さらに、血漿を用いた免疫沈降において、本発明の特異的モノクローナル抗体#11またはポリクローナル抗体の市販品(Human APLP-1 Antibody (R&D Systems, #AF3129))を用いて、APLP1並びにEVタンパク質であるCD9及びCD63の検出を比較した。Western Blotの結果を図10-2に示す。その結果、特異的モノクローナル抗体#11を用いることでAPLP1、CD9及びCD63を検出できた。それに対して、ポリクローナル抗体の市販品を用いた場合にはAPLP1は検出できず、CD9とCD63の検出も困難であった。 Furthermore, in immunoprecipitation using plasma, the detection of APLP1 and the EV proteins CD9 and CD63 was compared using the specific monoclonal antibody #11 of the present invention or a commercially available polyclonal antibody (Human APLP-1 Antibody (R&D Systems, #AF3129)). The results of Western Blot are shown in Figure 10-2. As a result, APLP1, CD9, and CD63 could be detected using specific monoclonal antibody #11. In contrast, when a commercially available polyclonal antibody was used, APLP1 could not be detected, and it was difficult to detect CD9 and CD63.
実施例3:iPS細胞から分化させた神経細胞の培養上清からの細胞外小胞の回収
(1)免疫沈降用ビーズの作製
EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) を用いて、実施例1で調製したヒトAPLP1特異的モノクローナル抗体#11をキット付属のプロトコルに従いビオチン化し、Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) をキット付属のプロトコルに従って使用し、余分なビオチンを除去・精製した。
Example 3 Recovery of extracellular vesicles from culture supernatant of neurons differentiated from iPS cells (1) Preparation of beads for immunoprecipitation Human APLP1-specific monoclonal antibody #11 prepared in Example 1 was biotinylated using EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) according to the protocol included in the kit, and excess biotin was removed and purified using Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) according to the protocol included in the kit.
Dynabeads(登録商標) MyOne Streptavidin C1 (Thermo Fisher Scientific) 20μLと10μgのビオチン化したヒトAPLP1特異的モノクローナル抗体#11を 0.01% Tween-20入りのDulbecco’s PBS内で混合し、ヒトAPLP1特異的モノクローナル抗体#11が結合した磁性ビーズを作製した。 20 μL of Dynabeads® MyOne Streptavidin C1 (Thermo Fisher Scientific) and 10 μg of biotinylated human APLP1-specific monoclonal antibody #11 were mixed in Dulbecco's PBS containing 0.01% Tween-20 to produce magnetic beads bound to human APLP1-specific monoclonal antibody #11.
更に、ECLTM Prime Blocking Reagentを2%(w/v)含むブロッキングバッファー 800μL中に抗体結合磁気ビーズを加えて4℃下で1晩インキュベートし、表面のブロッキングを行った。 Furthermore, the antibody-bound magnetic beads were added to 800 μL of blocking buffer containing 2% (w/v) ECL ™ Prime Blocking Reagent, and incubated overnight at 4° C. to block the surface.
(2)培養上清中の細胞外小胞(EV)の調製
iPS細胞から分化させた神経細胞の培養上清を2,500g, 10分間遠心し、上清80mLをAmicon Ultra-15 100kDa MWCOを用いて20mLまで濃縮した。qEV10(Izon Science社)を用いて、キット添付プロトコルにしたがって、濃縮培養上清10mLからサイズ排除クロマトグラフィー(SEC)分画 20 mLを回収した。上記を計2回実施し、SEC分画 40mLを回収した。Amicon Ultra-15 100kDa MWCOを用いてSEC分画を1,300μLまで濃縮した。
(2) Preparation of extracellular vesicles (EVs) in culture supernatant Culture supernatant of neural cells differentiated from iPS cells was centrifuged at 2,500g for 10 minutes, and 80 mL of the supernatant was concentrated to 20 mL using an Amicon Ultra-15 100kDa MWCO. Using qEV10 (Izon Science), 20 mL of size exclusion chromatography (SEC) fraction was collected from 10 mL of concentrated culture supernatant according to the kit protocol. The above was carried out twice in total, and 40 mL of SEC fraction was collected. The SEC fraction was concentrated to 1,300 μL using an Amicon Ultra-15 100kDa MWCO.
(3)免疫沈降及びWestern Blot
免疫沈降用ビーズに濃縮したSEC分画 300μLを添加し、4℃下で転倒混和しながら8時間インキュベーションした。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。続いて、磁性ビーズを洗浄するため、D-PBS(-) 800μLをチューブに加えて転倒混和した。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。再度スピンダウンして磁気スタンドに装着し1分間待ち、上清を完全に取り除いた。
(3) Immunoprecipitation and Western Blot
300 μL of concentrated SEC fraction was added to the immunoprecipitation beads and incubated for 8 hours at 4°C with inversion mixing. The tube was spun down and then attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. Next, to wash the magnetic beads, 800 μL of D-PBS(-) was added to the tube and mixed by inversion. The tube was spun down and then attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. The tube was spun down again, attached to a magnetic stand, waited for 1 minute, and the supernatant was completely removed.
キット説明書に従って調製した1x RIPA buffer (SDSなし) (ナカライテスク株式会社, 08714-04) 64μLをビーズの入ったチューブに加えてvortex後、4℃で10分間インキュベートした。遠心後磁気スタンドに付け、1分静置して上清を回収し、変性バッファーと混合した後、加熱し、アプライ用のサンプルを調製した。サンプルを5-15% Tris-glycine SDS gelにロードし、200Vで55分間泳動した。泳動が終わったSDSゲルを、セミドライ条件で、Towbin Buffer (5%メタノール入り)を用いて15Vで30分間泳動し、ゲルからPVDFメンブレンにトランスファーした。 64μL of 1x RIPA buffer (without SDS) (Nacalai Tesque, Inc., 08714-04) prepared according to the kit instructions was added to the tube containing the beads, vortexed, and incubated at 4℃ for 10 minutes. After centrifugation, the tube was attached to a magnetic stand and left to stand for 1 minute to collect the supernatant, which was then mixed with denaturing buffer and heated to prepare the sample for application. The sample was loaded onto a 5-15% Tris-glycine SDS gel and electrophoresed at 200V for 55 minutes. After electrophoresis, the SDS gel was electrophoresed under semi-dry conditions in Towbin Buffer (containing 5% methanol) at 15V for 30 minutes, and the gel was transferred to a PVDF membrane.
Blocking One (ナカライテスク株式会社)を用いて、室温で5分間ブロッキングした。TBS-Tに溶解した5% Blocking Oneで10,000希釈したAPLP1 C末抗体(Calbiochem 171615)、2,000倍希釈したCD63抗体(Santa Cruz sc-5275)、2,000倍希釈したCD81抗体(Santa Cruz sc-23962)のそれぞれを加えて4℃で一晩振盪しながらインキュベーションした。インキュベーション後、0.1% TBS-Tで5分、6回メンブレンを洗浄した。 Blocking was performed at room temperature for 5 minutes using Blocking One (Nacalai Tesque, Inc.). APLP1 C-terminus antibody (Calbiochem 171615) diluted 10,000 times in 5% Blocking One dissolved in TBS-T, CD63 antibody (Santa Cruz sc-5275) diluted 2,000 times, and CD81 antibody (Santa Cruz sc-23962) diluted 2,000 times were added and incubated overnight at 4°C with shaking. After incubation, the membrane was washed six times for 5 minutes with 0.1% TBS-T.
TBS-Tに溶解した5% Blocking Oneで二次抗体を希釈し、室温で1時間反応させた。使用した抗体は、HRP標識抗マウス抗体 (Promega, W4021)、HRP標識抗ラビット抗体 (Promega, W4011)であり、全て10,000希釈して用いた。TBS-Tを使って、1回5分の洗浄を6回行った。 The secondary antibody was diluted with 5% Blocking One dissolved in TBS-T and incubated at room temperature for 1 hour. The antibodies used were HRP-labeled anti-mouse antibody (Promega, W4021) and HRP-labeled anti-rabbit antibody (Promega, W4011), all diluted 10,000. Six washes were performed using TBS-T for 5 minutes each.
ImmunoStar(登録商標) LD(Wako)のA液とB液を800μLずつ混ぜ、メンブレンを浸して5分静置した。MY ECL Imager (Thermo Fisher Scientific)でシグナルを検出した(10-300秒)。 800 μL each of ImmunoStar® LD (Wako) solution A and B were mixed, the membrane was immersed in the solution and left to stand for 5 minutes. Signals were detected (10-300 seconds) using a MY ECL Imager (Thermo Fisher Scientific).
(4)結果
Western Blotの結果を図11に示す。
(4) Results The results of Western Blot are shown in FIG.
レーン1はiPS neuronのlysate 1μg、レーン2は、免疫沈降前のEV、レーン3はinput、レーン4はEVを免疫沈降した上清、レーン5はEVを免疫沈降した抗体カップリングビーズである。inputと上清は全体の1/40を泳動した。 Lane 1 is 1μg of iPS neuron lysate, lane 2 is EV before immunoprecipitation, lane 3 is input, lane 4 is the supernatant from immunoprecipitating EV, and lane 5 is antibody-coupled beads from immunoprecipitating EV. 1/40 of the total was electrophoresed for input and supernatant.
APLP1、EVマーカータンパク(CD63, Flotillin-1)、及び神経由来タンパク(L1CAM, SNAP25)が抗体ビーズで検出された。このことから、ヒトAPLP1特異的モノクローナル抗体#11を用いることにより、細胞外小胞が回収できることが示された。 APLP1, EV marker proteins (CD63, Flotillin-1), and neural-derived proteins (L1CAM, SNAP25) were detected with antibody beads. This demonstrated that extracellular vesicles can be recovered by using human APLP1-specific monoclonal antibody #11.
実施例4
CSF中及びNDE中の全タウ量の対比、NDE中の全タウ量とCSF中のリン酸化タウ(pTau181)及びAβ42/Aβ40の対比
(1)検体
患者からEDTA-2Kチューブを使って採血し、10mLの血漿を分離した。血漿は検査まで、-80℃下で保存した。使用時に、血漿を融解し、10,000×g, 室温で5分間遠心し、上清を回収した。血漿400μLに対してD-PBS(-) 100μLを加えた。
Example 4
Comparison of total tau amount in CSF and NDE, comparison of total tau amount in NDE with phosphorylated tau (pTau181) and Aβ42/Aβ40 in CSF (1) Samples Blood was collected from patients using EDTA-2K tubes, and 10 mL of plasma was separated. The plasma was stored at -80°C until testing. When used, the plasma was thawed and centrifuged at 10,000×g at room temperature for 5 minutes, and the supernatant was collected. 100 μL of D-PBS(-) was added to 400 μL of plasma.
(2)血漿細胞外小胞の粗精製
qEV original(Izon Science社)を用いて、キット添付プロトコルにしたがって、D-PBS(-)添加血漿500μLからサイズ排除クロマトグラフィー(SEC)分画 1,500μLを回収した。
(2) Crude purification of plasma extracellular vesicles Using qEV original (Izon Science), 1,500 μL of size exclusion chromatography (SEC) fraction was collected from 500 μL of D-PBS(-)-added plasma according to the protocol attached to the kit.
(3)免疫沈降用ビーズの作製
EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) を用いて、実施例1で調製したヒトAPLP1特異的モノクローナル抗体#11をキット付属のプロトコルに従いビオチン化し、Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) をキット付属のプロトコルに従って使用し、余分なビオチンを除去・精製した。
(3) Preparation of beads for immunoprecipitation Human APLP1-specific monoclonal antibody #11 prepared in Example 1 was biotinylated using EZ-Link NHS-PEG4-Biotin No-Weigh Format (Thermo Fisher Scientific) according to the protocol included with the kit, and excess biotin was removed and purified using Zeba Spin Desalting Columns 40K MWCO 0.5mL (Thermo Fisher Scientific) according to the protocol included with the kit.
Dynabeads(登録商標) MyOne Streptavidin C1 (Thermo Fisher Scientific) 10μLと4μgのビオチン化したヒトAPLP1特異的モノクローナル抗体#11を 0.01% Tween-20入りのD-PBS(-)内で混合し、特異的モノクローナル抗体#11が結合した磁性ビーズを作製した。 10 μL of Dynabeads (registered trademark) MyOne Streptavidin C1 (Thermo Fisher Scientific) and 4 μg of biotinylated human APLP1-specific monoclonal antibody #11 were mixed in D-PBS(-) containing 0.01% Tween-20 to prepare magnetic beads bound to specific monoclonal antibody #11.
更に、ECLTM Prime Blocking Reagentを2%(w/v)含むブロッキングバッファー 800μL中に抗体結合磁気ビーズを加えて4℃下で1晩インキュベートし、表面のブロッキングを行った。 Furthermore, the antibody-bound magnetic beads were added to 800 μL of blocking buffer containing 2% (w/v) ECL ™ Prime Blocking Reagent, and incubated overnight at 4° C. to block the surface.
(4)脳神経細胞由来細胞外小胞の免疫沈降
上記(3)で作製した免疫沈降用ビーズに濃縮したSEC分画 1,444μLを添加し、4℃下で6時間転倒混和しながらインキュベーションした。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。続いて、磁性ビーズを洗浄するため、D-PBS(-) 800μLをチューブに加えて転倒混和した。チューブをスピンダウンしたのち磁気スタンドに装着し1分間待ち、上清を除去した。再度スピンダウンして磁気スタンドに装着し1分間待ち、上清を完全に取り除いた。
(4) Immunoprecipitation of brain neuron-derived extracellular vesicles 1,444 μL of concentrated SEC fraction was added to the immunoprecipitation beads prepared in (3) above, and incubated at 4°C for 6 hours with inversion mixing. The tube was spun down, attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. Next, to wash the magnetic beads, 800 μL of D-PBS(-) was added to the tube and mixed by inversion. The tube was spun down, attached to a magnetic stand, waited for 1 minute, and the supernatant was removed. The tube was spun down again, attached to a magnetic stand, waited for 1 minute, and the supernatant was completely removed.
磁性ビーズが残ったチューブに0.1M glycine-HCl (pH 2.8) 105μLを加え、ボルテックス後、4℃で10分間インキュベートした。チューブをスピンダウンした後、磁気スタンドに装着し1分間待ち、上清を回収し、1M Tris-HCl (pH 8.0) 15.8μL加えてpHを調整し、解析に供した。 105μL of 0.1M glycine-HCl (pH 2.8) was added to the tube containing the remaining magnetic beads, which was then vortexed and incubated at 4°C for 10 minutes. The tube was then spun down, placed on a magnetic stand and left for 1 minute. The supernatant was then collected and 15.8μL of 1M Tris-HCl (pH 8.0) was added to adjust the pH before preparation for analysis.
(5)Simoa Tau 2.0 assay
前ステップで回収した上清を専用96ウェルプレートに分注し、キット説明書に基づいてSimoa HD-1 Analyzer機器およびSimoa Tau 2.0 Advantage Kitを用いてTau濃度を測定した。これにより、ヒトAPLP1特異的モノクローナル抗体#11を用い血漿から分離された細胞外小胞に含まれる総タウ量を求めた。
(5) Simoa Tau 2.0 assay
The supernatant collected in the previous step was dispensed into a dedicated 96-well plate, and the Tau concentration was measured using the Simoa HD-1 Analyzer instrument and the Simoa Tau 2.0 Advantage Kit according to the kit instructions. This resulted in the determination of the total amount of Tau contained in extracellular vesicles isolated from plasma using human APLP1-specific monoclonal antibody #11.
(1)で血漿を採取した日と同日に腰椎穿刺によって脳脊髄液を採取した。脳脊髄液は室温にて430xgで5分間遠心し、分注して-80℃下で保存した。総タウ量およびリン酸化タウ(pTau181)量、Aβ42、Aβ40量はサンドイッチELISA法によって測定した。 Cerebrospinal fluid was collected by lumbar puncture on the same day as plasma was collected in (1). The cerebrospinal fluid was centrifuged at 430xg for 5 minutes at room temperature, aliquoted, and stored at -80°C. The amounts of total tau, phosphorylated tau (pTau181), Aβ42, and Aβ40 were measured by sandwich ELISA.
(6)結果
測定された血漿脳神経細胞由来細胞外小胞中の総タウ量と、脳脊髄液中の総タウとの相関を示す散布図を図12に示し、測定された血漿脳神経細胞由来細胞外小胞中の総タウ量と、リン酸化タウ(pTau181)(左図)、Aβ42/Aβ40比(右図)との相関を示す散布図を図13に示す。
(6) Results Figure 12 shows a scatter plot showing the correlation between the measured total tau amount in plasma brain neuron-derived extracellular vesicles and the total tau in cerebrospinal fluid, and Figure 13 shows a scatter plot showing the correlation between the measured total tau amount in plasma brain neuron-derived extracellular vesicles and phosphorylated tau (pTau181) (left figure) and the Aβ42/Aβ40 ratio (right figure).
ヒトAPLP1特異的モノクローナル抗体#11及び血漿を用いた、免疫沈降により、分離された細胞外小胞に含まれる総タウ量は、脳脊髄液中に含まれる総タウ、リン酸化(pTau181)、Aβ42/Aβ40比と良く相関していた。このことから、脳神経細胞由来細胞外小胞であるAPLP1陽性EVが分離できており、脳脊髄液中のバイオマーカー量を反映していることが明らかとなった。さらに、ヒトAPLP1特異的モノクローナル抗体#11を用いて血漿から分離された細胞外小胞に含まれる総タウ量は、精神神経系疾患バイオマーカーとなり得ること、この方法で検出した総タウ量を用いることで、採取が容易ではない脳脊髄液に代わって、採取が容易な血漿を検体として用いた精神神経系疾患の検出が可能であることが分かった。 The total amount of tau contained in the extracellular vesicles isolated by immunoprecipitation using human APLP1-specific monoclonal antibody #11 and plasma correlated well with the total tau, phosphorylated (pTau181), and Aβ42/Aβ40 ratio contained in the cerebrospinal fluid. This demonstrated that APLP1-positive EVs, which are extracellular vesicles derived from brain neurons, could be isolated and reflect the amount of biomarkers in the cerebrospinal fluid. Furthermore, it was found that the total amount of tau contained in the extracellular vesicles isolated from plasma using human APLP1-specific monoclonal antibody #11 could be a biomarker for neuropsychiatric disorders, and that the total amount of tau detected by this method could be used to detect neuropsychiatric disorders using plasma, which is easy to collect, as a sample instead of cerebrospinal fluid, which is difficult to collect.
本発明は、精神神経系疾患のバイオマーカーに関連する分野において有用である。 The present invention is useful in fields related to biomarkers for neuropsychiatric disorders.
配列番号1:ヒトAPLP1アミノ酸配列(NCBI accession No. NP_031493.2)
配列番号2:#11軽鎖アミノ酸配列
配列番号3:#11重鎖アミノ酸配列
配列番号4:#11 軽鎖FR1
配列番号5:#11 軽鎖CDR1
配列番号6:#11 軽鎖FR2
配列番号7:#11 軽鎖CDR2
配列番号8:#11 軽鎖FR3
配列番号9:#11 軽鎖CDR3
配列番号10:#11 軽鎖FR4
配列番号11:#11重鎖FR1
配列番号12:#11重鎖CDR1
配列番号13:#11重鎖FR2
配列番号14:#11重鎖CDR2
配列番号15:#11重鎖FR3
配列番号16:#11重鎖CDR3
配列番号17:#11重鎖FR4
配列番号18:#11軽鎖シグナル配列
配列番号19:#11重鎖シグナル配列
配列番号20:最小エピトープの特定用ペプチド配列
配列番号21:最小エピトープの特定用ペプチド配列
配列番号22:最小エピトープの特定用ペプチド配列
配列番号23:最小エピトープの特定用ペプチド配列
配列番号24:最小エピトープの特定用ペプチド配列
SEQ ID NO: 1: Human APLP1 amino acid sequence (NCBI accession no. NP_031493.2)
SEQ ID NO:2: #11 light chain amino acid sequence SEQ ID NO:3: #11 heavy chain amino acid sequence SEQ ID NO:4: #11 light chain FR1
SEQ ID NO:5: #11 Light chain CDR1
SEQ ID NO:6: #11 Light chain FR2
SEQ ID NO:7: #11 Light chain CDR2
SEQ ID NO: 8: #11 Light chain FR3
SEQ ID NO: 9: #11 Light chain CDR3
SEQ ID NO: 10: #11 light chain FR4
SEQ ID NO: 11: #11 heavy chain FR1
SEQ ID NO: 12: #11 heavy chain CDR1
SEQ ID NO: 13: #11 heavy chain FR2
SEQ ID NO: 14: #11 heavy chain CDR2
SEQ ID NO: 15: #11 heavy chain FR3
SEQ ID NO: 16: #11 heavy chain CDR3
SEQ ID NO: 17: #11 heavy chain FR4
SEQ ID NO:18: #11 light chain signal sequence SEQ ID NO:19: #11 heavy chain signal sequence SEQ ID NO:20: Peptide sequence for identifying minimal epitope SEQ ID NO:21: Peptide sequence for identifying minimal epitope SEQ ID NO:22: Peptide sequence for identifying minimal epitope SEQ ID NO:23: Peptide sequence for identifying minimal epitope SEQ ID NO:24: Peptide sequence for identifying minimal epitope
Claims (14)
前記抗APLP1抗体と細胞外小胞の複合体を回収する工程、
を含み、
抗APLP1抗体が請求項1または2に記載の抗APLP1モノクローナル抗体である、
脳神経細胞に由来する細胞外小胞の回収方法。 A step of mixing a sample containing an anti-APLP1 antibody and extracellular vesicles to form a complex of the anti-APLP1 antibody and the extracellular vesicles, and a step of recovering the complex of the anti-APLP1 antibody and the extracellular vesicles.
Including,
The anti-APLP1 antibody is an anti-APLP1 monoclonal antibody according to claim 1 or 2.
A method for recovering extracellular vesicles derived from brain neurons.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2023-079084 | 2023-05-12 | ||
| JP2023079084 | 2023-05-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024237108A1 true WO2024237108A1 (en) | 2024-11-21 |
Family
ID=93519084
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2024/016798 Pending WO2024237108A1 (en) | 2023-05-12 | 2024-05-01 | Anti-aplp1 monoclonal antibody and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024237108A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JPH06128294A (en) * | 1992-04-20 | 1994-05-10 | General Hospital Corp | Amyloid precursor-like protein and use thereof |
| WO2021132352A1 (en) * | 2019-12-27 | 2021-07-01 | 国立大学法人大阪大学 | Method for collecting extracellular vesicles derived from nervous system cells |
| US20220099686A1 (en) * | 2018-11-30 | 2022-03-31 | Cha University Industry-Academic Cooperation Foundation | Brain-derived vesicle-specific marker and brain disease diagnostic method using same |
-
2024
- 2024-05-01 WO PCT/JP2024/016798 patent/WO2024237108A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JPH06128294A (en) * | 1992-04-20 | 1994-05-10 | General Hospital Corp | Amyloid precursor-like protein and use thereof |
| US20220099686A1 (en) * | 2018-11-30 | 2022-03-31 | Cha University Industry-Academic Cooperation Foundation | Brain-derived vesicle-specific marker and brain disease diagnostic method using same |
| WO2021132352A1 (en) * | 2019-12-27 | 2021-07-01 | 国立大学法人大阪大学 | Method for collecting extracellular vesicles derived from nervous system cells |
Non-Patent Citations (1)
| Title |
|---|
| COUSINS SARAH L., DAI WEI, STEPHENSON F. ANNE: " APLP1 and APLP2, members of the APP family of proteins, behave similarly to APP in that they associate with NMDA receptors and enhance NMDA receptor surface expression", JOURNAL OF NEUROCHEMISTRY, WILEY-BLACKWELL PUBLISHING LTD., GB, vol. 133, no. 6, 1 June 2015 (2015-06-01), GB , pages 879 - 885, XP093240174, ISSN: 0022-3042, DOI: 10.1111/jnc.13063 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP2021511510A (en) | Detection of exosomes and exosome biomarkers for diagnosis and prognosis of diseases and disorders | |
| JP7511938B2 (en) | Apparatus for producing exosome liquid biopsy samples and method for producing same | |
| JP7645514B2 (en) | Method for recovering extracellular vesicles derived from nervous system cells | |
| WO2008016186A1 (en) | Antibody specific to intact human autotaxin, method of screening the same and method and reagent for examining malignant lymphoma by assaying autotaxin | |
| JP2025098086A (en) | Tau protein detection method using blood sample as specimen | |
| CN110133272A (en) | Method for enrichment or detection of astrocyte-derived exosomes from biological fluids | |
| US20220341948A1 (en) | Methods and compositions for monitoring phagocytic activity | |
| KR101658620B1 (en) | Method for Detecting Aggregate Form of Aggregate-Forming Polypeptides | |
| CN114127561A (en) | Affinity separation systems and methods using switch-like binding reactions | |
| US20140011192A1 (en) | Particle complex and method of isolating target cell | |
| JP2024507755A (en) | Method for detecting neurofilament light chains in plasma and cerebrospinal fluid | |
| KR102498862B1 (en) | Method for analyzing exosomes in liquid biopsy sample | |
| WO2024237108A1 (en) | Anti-aplp1 monoclonal antibody and use thereof | |
| WO2025080894A1 (en) | Detection of disease state macromolecules binding to normal macromolecules as a biomarker for disease identification | |
| CN112430569B (en) | Application of protein SFTPC as lung cancer diagnosis marker and kit | |
| WO2023224919A1 (en) | Isolation and diagnostic methods using cell type-specific and/or organ-specific extracellular vesicle (ev) markers | |
| CN113785200B (en) | Marker composition based on exosomes overexpressing TUBA1C protein | |
| CN115066617A (en) | Methods for predicting the effectiveness of treatment with immune checkpoint inhibitors for lung cancer patients | |
| WO2020241812A1 (en) | Anti-oxytocin monoclonal antibody and kit containing said antibody | |
| WO2025075789A1 (en) | Immunoassay for detecting tau phosphorylated at serine 413 | |
| JP7007665B2 (en) | Method for determining the tissue regeneration state of a living body | |
| WO2025004683A1 (en) | Cancer examination method and cancer treatment agent | |
| WO2025182761A1 (en) | Gfap detection method, method for assisting diagnosis of alzheimer 's disease, and kit used therefor | |
| TWI382178B (en) | Methods for detecting biomolecules in a sample | |
| Risha | The proteomic analysis of exosomes from breast cell lines reveals potential biomarkers of breast cancer |
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: 24807062 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2025520511 Country of ref document: JP Kind code of ref document: A |