WO2024076210A1 - Method for screening for candidate material for inhibiting or disaggregating amyloid protein aggregation - Google Patents

Method for screening for candidate material for inhibiting or disaggregating amyloid protein aggregation Download PDF

Info

Publication number
WO2024076210A1
WO2024076210A1 PCT/KR2023/015443 KR2023015443W WO2024076210A1 WO 2024076210 A1 WO2024076210 A1 WO 2024076210A1 KR 2023015443 W KR2023015443 W KR 2023015443W WO 2024076210 A1 WO2024076210 A1 WO 2024076210A1
Authority
WO
WIPO (PCT)
Prior art keywords
amyloid
protein
aggregation
amyloid protein
plate
Prior art date
Application number
PCT/KR2023/015443
Other languages
French (fr)
Korean (ko)
Inventor
김영수
조일환
김혜주
김혜연
Original Assignee
아밀로이드솔루션 주식회사
연세대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 아밀로이드솔루션 주식회사, 연세대학교 산학협력단 filed Critical 아밀로이드솔루션 주식회사
Publication of WO2024076210A1 publication Critical patent/WO2024076210A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • It relates to a method for screening an amyloid protein aggregation inhibitor or disaggregator, a kit, and a method for screening the aggregation inhibition or disaggregator site of an amyloid protein aggregation inhibitor or disaggregator.
  • Amyloidosis is a disease that occurs when abnormal proteins called amyloid accumulate in tissues. Amyloid is a protein mass with a diameter of 7-13 nm and a beta-sheet structure that appears in a fibrous form when viewed under a microscope, and is characterized by staining with Thioflavin T (ThioT) and Congo red. there is. Amyloid is not found in the normal body, and to date, it has been reported that 36 proteins can form it (Picken, Acta Haematol. (2020), 143:322-334). Representative amyloidosis includes neurological diseases such as Alzheimer's disease, Parkinson's disease, Huntington disease, and prion disease. In addition, there are various types of amyloidosis depending on the causative protein and affected organ. There are a number of amyloidoses with different presentations.
  • amyloid-beta An important pathological feature of Alzheimer's disease, one of these amyloidoses, is the formation of peptide aggregates called senile plaques, which cause synaptic dysfunction and neuronal death.
  • amyloid-beta One of the components of these senile plaques is amyloid-beta, which is 40-42 amino acids long.
  • Amyloid-beta monomers are prone to self-assemble into oligomers, protofibrils, and beta-sheet-rich fibers and are associated with the pathogenesis of neurotoxicity.
  • amyloid-beta plaques Although the correlation between amyloid-beta plaques and neurotoxicity has not yet been clearly elucidated, the self-assembly of amyloid-beta into intermediate oligomers or aggregates is believed to be involved in the pathogenesis of cranial nerve diseases such as Alzheimer's disease.
  • amyloid disease treatments developed to date use antibodies against A ⁇ fragments or specific epitopes within A ⁇ that cause an immune response in patients, or apply chemical drugs.
  • chemical drugs there is currently no fundamental treatment for amyloid disease.
  • the present inventors completed the present invention to provide a method of screening for a therapeutic agent for degenerative brain diseases that inhibits the formation of amyloid protein, especially amyloid beta plaques.
  • One aspect is to provide a method of screening for substances that inhibit, inhibit, or dissolve the aggregation of amyloid proteins.
  • Another aspect is to provide a kit for screening substances that inhibit, inhibit, or dissolve the aggregation of amyloid proteins.
  • Another aspect is to provide a method of screening for an aggregation inhibition or dissolution site of action of a substance that inhibits, inhibits, or dissolves the aggregation of amyloid protein.
  • the present invention provides a method for screening for an aggregation inhibitor or an aggregation-dissolving agent of amyloid protein, for example, an amyloid protein aggregation inhibitor or Methods and kits for screening candidate solubilizers are provided.
  • One aspect includes (1) providing a plate to which a first amyloid protein is attached;
  • step (2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or (2-2) contacting a second amyloid protein to which an indicator substance is bound to the plate and then treating the candidate substance as a protein aggregation dissolving agent; And, (3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1) and selecting an aggregation inhibitor candidate changed compared to the untreated control group; or (3-2) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2) and selecting an aggregation resolving agent candidate changed compared to the untreated control group.
  • a method for screening solubilizers is provided.
  • step (2-1) or (2-2) may further include inducing aggregation of the first and second amyloid proteins.
  • the method of screening for the protein aggregation inhibitor or aggregation dissolving agent is an amyloid protein aggregation inhibitor or a candidate material that changes the aggregation rate of amyloid protein compared to the substance that changed the aggregation rate of the amyloid protein or the untreated control. It may further include a step of selecting as a coagulant dissolving agent.
  • amyloid protein refers to a protein that tangles and aggregates in the form of a beta plate structure, and refers to a protein or a fragment thereof capable of forming amyloid deposits.
  • amyloid protein may refer to individual proteins constituting amyloid protein aggregates.
  • the amyloid protein may be amyloid beta, tau protein, alpha-synuclein, prion, amyloidogenic protein, amyloid transthyretin, Lewy body, fliglutamine, Torsin A, and AIMP2 protein.
  • amyloid beta is a peptide molecule containing 36 to 43 amino acids, is a major component of amyloid plaques, and is known to be involved in the development of Alzheimer's disease.
  • the amyloid beta peptide molecules aggregate to form neurotoxic oligomers, causing degenerative brain diseases.
  • the amyloid beta peptide molecule may be obtained by chopping amyloid precursor protein (APP); UniProtKB P05067) with beta secretase and gamma secretase.
  • the amyloid protein may self-assemble.
  • amyloid protein aggregate refers to amyloid protein aggregates that are associated with disease states. These disease states include, but are not limited to, cell death, or partial or complete loss of neuronal signaling between two or more cells. Amyloid protein aggregates may be located inside the cell or outside the cell. Specifically, amyloid protein aggregates can cause neurotoxicity and cause degenerative brain diseases described later.
  • the amyloid protein aggregates may include polymers of amyloid proteins, fibril precursors, fibrils, plaques, protein monomers and polymers bound to homologous proteins.
  • the fibrils may include oligomers of amyloid protein.
  • the self-assembling protein may be soluble amyloid beta aggregates and/or water-insoluble amyloid beta aggregates, amyloid beta oligomers, amyloid beta protofibrils, amyloid beta fibrils, and amyloid beta plaques. You can.
  • the first or second amyloid protein may be a full-length protein and/or a protein fragment.
  • the full-length protein may be an amyloid beta 1-36, amyloid beta 1-38, amyloid beta 1-40, or amyloid beta 1-42 peptide.
  • protein fragment refers to a truncated form of a protein. Specifically, the protein fragment refers to a cleaved portion of the full-length protein.
  • the protein fragment may be a continuous protein within an amyloid protein. Specifically, it may be a continuous 6 to 15 mer within the amyloid protein.
  • the protein fragment may be 6 to 15 mer.
  • the protein fragment may be 6 to 14 mer. In one embodiment, the protein fragment may be 6 to 13 mer. In one embodiment, the protein fragment may be 6 to 12 mer. In one embodiment, the protein fragment may be 6 to 11 mer. In one embodiment, the protein fragment may be 6 to 10 mer. In one embodiment, the protein fragment may be 6 to 9 mer.
  • the protein fragment may be amyloid beta 6mer (hereinafter also referred to as amyloid beta hexamer).
  • the protein fragment may be DAEFRH.
  • the protein fragment may be AEFRHD.
  • the protein fragment may be EFRHDS.
  • the protein fragment may be FRHDSG.
  • the protein fragment may be RHDSGY.
  • the protein fragment may be HDSGYE.
  • the protein fragment may be DSGYEV.
  • the protein fragment may be SGYEVH.
  • the protein fragment may be GYEVHH.
  • the protein fragment may be YEVHHQ.
  • the protein fragment may be EVHHQK.
  • the protein fragment may be VHHQKL.
  • the protein fragment may be HHQKLV.
  • the protein fragment may be HQKLVF.
  • the protein fragment may be QKLVFF.
  • the protein fragment may be KLVFFA.
  • the protein fragment may be LVFFAE.
  • the protein fragment may be VFFAED.
  • the protein fragment may be FFAEDV.
  • the protein fragment may be FAEDVG.
  • the protein fragment may be AEDVGS.
  • the protein fragment may be EDVGSN.
  • the protein fragment may be DVGSNK.
  • the protein fragment may be DVGSNG.
  • the protein fragment may be VGSNGA.
  • the protein fragment may be GSNGAI.
  • the protein fragment may be SNGAII.
  • the protein fragment may be NGAIIG.
  • the protein fragment may be GAIIGL.
  • the protein fragment may be AIIGLM.
  • the protein fragment may be IIGLMV.
  • the protein fragment may be IGLMVG.
  • the protein fragment may be GLMVGG.
  • the protein fragment may be LMVGGV.
  • the protein fragment may be MVGGVV.
  • the protein fragment may be VGGVVI.
  • the protein fragment may be GGVVIA.
  • the first amyloid protein in the step of providing a plate to which the first amyloid protein is attached, may be attached to the plate by cysteine bound to the C-terminus.
  • the full-length protein amyloid beta 1-42 or amyloid beta hexamer may be attached to the plate through a cysteine bound to the C-terminus.
  • the cysteine may be bound to the C-terminus of the first amyloid protein through systemic anhydride activation, but is not limited thereto. Binding of cysteine to the C- or N-terminus of amyloid protein can be performed by methods that are obvious to those skilled in the art.
  • the peptide having cysteine bound to the C-terminus of the amyloid protein may be synthesized through a solid-phase peptide synthesis method. Specifically, it may be by synthesizing 9-fluorenylmethyloxycarbonyl solid phase peptide using microwaves, but is not limited thereto.
  • full-length amyloid beta 1-42 or amyloid beta hexamer with cysteine bound to the C-terminus is formed by binding cysteine to the C-terminus through systemic anhydride activation, and then activating it in the microwave. It can be produced by 9-fluorenylmethyloxycarbonyl solid-phase peptide synthesis using .
  • the full-length amyloid beta 1-42 or amyloid beta hexamer with cysteine bound to the C-terminus may be attached to the C-terminus by reacting cysteine with a functional group of the plate, specifically maleimide. and its derivatives, aziridine and its derivatives, acryloyl and its derivatives, or aryl halide and its derivatives.
  • the first amyloid protein may be attached to the plate in monomeric form and may be attached to the plate to maintain the monomeric form before forming aggregates by contact with the second amyloid protein.
  • the method includes the steps of (1) providing a plate to which full-length amyloid beta 1-42 or amyloid beta hexamer is attached through a cysteine bound to the C terminus; (2-1) contacting the plate with full-length amyloid beta 1-42 to which a protein aggregation inhibitor candidate and an indicator are bound, or (2-2) contacting full-length amyloid beta 1-42 to which an indicator is bound.
  • processing the protein aggregation dissolving candidate material and, (3-1) selecting an aggregation inhibitor candidate that inhibits aggregation between amyloid beta livers compared to the untreated control group;
  • a method of screening a protein aggregation inhibitor or an aggregation-dissolving agent is provided, which includes a step of selecting a candidate for an aggregation-dissolving agent that dissolves aggregation between aggregated amyloid beta compared to an untreated control group.
  • the method may further include measuring the aggregation rate between amyloid beta.
  • the step of contacting the second amyloid protein may further include inducing aggregation with the first amyloid protein.
  • the second amyloid protein may be bound to an indicator substance.
  • the indicator material is for measuring the degree of binding between the first amyloid protein and the second amyloid protein; And it may be used to measure the aggregation inhibition or aggregation dissolution ability of the first amyloid protein and the second amyloid protein by the aggregation inhibitor, aggregation dissolving agent, aggregation inhibitor candidate, or aggregation dissolving agent candidate, specifically, the degree of aggregation inhibition or aggregation dissolution. there is.
  • the indicator may be a fluorescent substance or a fluorescent protein fragment.
  • the fluorescent material is a fluorescent dye having rhodamine, coumarin, EvoBlue, oxazine, carbopyronine, naphthalene, biphenyl, anthracene, phenanthrene, pyrene, or carbazole as a basic skeleton, or It may be a derivative of the fluorescent dye.
  • the fluorescent substance is Fluorescein, CR110:Carboxyrhodamine 110:Rhodamine Green (trade name), TAMRA:Carboxytetramethylrhodamine:TMR, Carboxyrhodamine 6G:CR6G, BODIPY FL (trade name): 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-propionic acid, BODIPY 493/503 (trade name): 4,4-difluoro- 1,3,5,7-Tetramethyl-4-bora-3a,4a-diaza-s-indacene-8-propionic acid, BODIPY R6G (trade name): 4,4-difluoro-5-(4- Phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza-s-indacene-3-propionic acid, BODIPY
  • the fluorescent protein fragments include Venus, Cerulean, Citrine, and mKate, and may be fluorescent proteins of different colors, or may be parts thereof.
  • amyloid protein aggregation inhibitor refers to inhibiting the accumulation of amyloid proteins
  • amyloid protein aggregation dissolving agent may refer to a substance that decomposes the aggregation of already formed amyloid proteins.
  • the aggregation inhibitor or aggregation dissolving agent may refer to a substance that inhibits self-assembly of amyloid proteins or breaks down aggregation of already formed self-assembled proteins. Specifically, it may refer to a substance that inhibits aggregation or disaggregates water-soluble protein aggregates and/or water-insoluble protein aggregates.
  • the aggregation inhibitor or aggregation dissolving agent may inhibit or dissolve the aggregation of amyloid beta, tau protein, or alpha-synuclein.
  • the aggregation inhibitor inhibits amyloid fibrillation and amyloid fiber aggregation
  • the aggregation dissolving agent includes the dissociation of aggregates. Specifically, it dissolves or accumulates soluble amyloid beta aggregates and/or water-insoluble amyloid beta aggregates, amyloid beta oligomers, amyloid beta protofibrils, amyloid beta fibrils, and amyloid beta plaques. It is an inhibitory substance.
  • amyloid protein aggregation inhibitor or aggregation-dissolving agent may be a treatment for degenerative brain disease.
  • the aggregation inhibitor or aggregation inhibitor candidate may be treated simultaneously or sequentially with the second amyloid protein.
  • the aggregation inhibitor may be treated simultaneously with the second amyloid protein.
  • the amyloid beta aggregation inhibitor curcumin or scyllo-inositol may be treated simultaneously with fluor-A ⁇ 1-42 .
  • the aggregation-dissolving agent or aggregation-dissolving agent candidate may be treated after aggregating the first amyloid protein and the second amyloid protein.
  • amyloid beta aggregation dissolving agent necrostatin-1 and sunitnib may be treated sequentially with fluor-A ⁇ 1-42 , for example, after 24 hours.
  • the protein aggregation inhibitor or aggregation-dissolving agent is used in the prevention and treatment of degenerative brain diseases such as (1) accumulation of amyloid beta, (2) aging of brain cells, (3) loss of synapses, and (4) accumulation of peripheral immune cells. It may alleviate the pathological symptoms of degenerative brain disease, specifically Alzheimer's disease.
  • the term “degenerative brain disease” refers to all diseases related to degenerative changes in the brain, especially all diseases (brain diseases) that can be caused by factors such as aggregation of amyloid-beta in the brain and/or brain nerve cells. It is used to describe comprehensively.
  • the degenerative brain disease includes dementia, Alzheimer's disease, preclinical Alzheimer's disease, Parkinson's disease, Huntington's disease, and mild cognitive impairment.
  • the degenerative brain disease may be Alzheimer's disease.
  • Alzheimer's disease is used interchangeably with senile dementia and refers to a disease involving mental deterioration associated with a specific degenerative brain disease characterized by senile plaques, neuroinflammatory tangles, and progressive nerve loss. You can.
  • Parkinson's disease may refer to a chronic and progressive degenerative disease of the central nervous system that often impairs motor function and language ability.
  • Huntington's disease may refer to a neurodegenerative disease that is caused by a 3-base repeat expansion in the gene encoding the huntingtin protein and is accompanied by chorea, psychosis, dementia, etc.
  • the term “Lou Gehrig's disease” refers to a disease in which only motor neurons are selectively killed, and may refer to a disease in which both upper motor neurons in the cerebral cortex and lower motor neurons in the brainstem and spinal cord are gradually destroyed.
  • Pick's disease may refer to a disease that causes progressive destruction of nerve cells in the brain.
  • tauopathy of the present invention may refer to a neurodegenerative disease in which cranial nerves are damaged due to abnormal accumulation of tau protein (a family closely related to intracellular microtubul-related proteins) in brain tissue.
  • the method includes providing a plate to which a first amyloid protein is attached with cysteine bound to the C-terminus;
  • a method for screening a protein aggregation inhibitor or an aggregation dissolving agent comprising the step of selecting a candidate for an aggregation dissolving agent that dissolves the aggregation between the first amyloid protein and the second amyloid protein compared to the untreated control group is provided.
  • the method may further include measuring the aggregation rate between the first amyloid protein and the second amyloid protein.
  • the step of contacting the second amyloid protein may further include inducing aggregation with the first amyloid protein.
  • the full-length protein amyloid beta 1-42 or the protein fragment amyloid beta hexamer may be attached to the plate with cysteine bound to the C-terminus.
  • the concentration of the first amyloid protein treated on the plate may be 0.00001 ⁇ M to 50 ⁇ M. In one embodiment, the concentration of the first amyloid protein may be 0.01 ⁇ M to 20 ⁇ M. In one embodiment, the concentration of the first amyloid protein may be 0.1 ⁇ M to 15 ⁇ M. In one embodiment, the concentration of the first amyloid protein may be 0.1 ⁇ M to 10 ⁇ M. In one embodiment, the concentration of the first amyloid protein may be 0.5 ⁇ M to 5 ⁇ M. Specifically, the concentration of the first amyloid protein may be 1 ⁇ M.
  • the concentration of the first amyloid protein processed on the plate is 0.1 ⁇ M or less, the fixation effect of the first amyloid protein fixed on the plate is low, making it difficult to use it in the screening method of the present invention. If the concentration exceeds 10 ⁇ M, the fixation effect of the first amyloid protein immobilized on the plate is saturated, and the concentration of the first amyloid protein present on the plate without being fixed due to non-specific binding increases, causing an error in measuring the aggregation rate. As it can be produced, the concentration of the first amyloid protein of the present invention should be 0.1 ⁇ M to 10 ⁇ M, preferably 0.5 ⁇ M to 5 ⁇ M.
  • the first amyloid protein in the step of providing a plate to which the first amyloid protein is attached, may be attached to the plate by treating the plate and incubating at 20°C to 40°C. In one embodiment, the amyloid protein may be attached to the plate by incubation at 25 to 35°C. Specifically, the amyloid protein may be attached to the plate by incubation at 28 to 32°C. When the first amyloid protein is attached by incubating at less than 25°C or more than 35°C, the attachment efficiency to the plate is significantly low and cannot be applied to the screening method for aggregation inhibitor or aggregation dissolving agent. Therefore, the first amyloid protein is treated on the plate. It should be adhered to the plate by incubation at 25°C to 35°C, preferably 28 to 32°C.
  • the first amyloid protein in the step of providing a plate to which the first amyloid protein is attached, may be attached to the plate by treating the plate and incubating for 12 to 36 hours. In one embodiment, the amyloid protein may be attached to the plate by incubating for 20 to 26 hours. Specifically, the first amyloid protein may be attached to the plate by incubating for 23 to 25 hours.
  • the fixation effect of the first amyloid protein immobilized on the plate is low, making it difficult to use it in the screening method of the present invention, and when the amyloid protein is incubated for more than 26 hours, the fixation effect is saturated, so the experiment In terms of speed and business feasibility, the first amyloid protein should be attached to the plate by incubating for 20 to 26 hours, preferably 23 to 25 hours.
  • the first amyloid protein in providing a plate to which the first amyloid protein is attached, may be attached to the plate at pH 4 to 9. In one embodiment, the first amyloid protein may be attached to the plate at pH 5 to 8. Specifically, the first amyloid protein may be attached to the plate at pH 6.5 to 7.2. When the first amyloid protein is attached to the plate at pH less than 5 or more than pH 8, the fixation effect of the first amyloid protein is low and the screening efficiency is reduced when used in the screening method of the present invention. Therefore, the first amyloid protein is used at pH 5 or higher. It should adhere to the plate at pH 8, preferably pH 6.5 to 7.2.
  • the second amyloid protein in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, is treated at a concentration in the range of 1 ⁇ M to 15 ⁇ M to the plate to which the first amyloid protein is attached. It can be. In one embodiment, it can be treated at a concentration ranging from 5 ⁇ M to 15 ⁇ M. Specifically, it can be treated at a concentration within the range of 8 ⁇ M to 12 ⁇ M.
  • the second amyloid protein When the second amyloid protein is treated at less than 5 ⁇ M, the aggregation rate between the first and second amyloid proteins is low, making it difficult to use in the screening method of the present invention, and when the second amyloid protein is treated at more than 15 ⁇ M, non-specific binding occurs.
  • the concentration of the second amyloid protein that does not aggregate with the first amyloid protein and is present on the plate increases, which may cause errors in measuring the aggregation rate.
  • the second amyloid protein is added to the plate to which the first amyloid protein is attached at 5 ⁇ M to 15 ⁇ M. , preferably at 8 ⁇ M to 12 ⁇ M.
  • the second amyloid protein in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, is treated to the plate to which the first amyloid protein is attached and incubated at 20°C to 40°C. By incubation, it can be aggregated with the first amyloid protein.
  • the amyloid protein can be aggregated with the first amyloid protein by incubating at 28 to 38°C. Specifically, the amyloid protein may be incubated at 30 to 36° C. to aggregate with the first amyloid protein.
  • the amyloid protein should be treated on a plate to which the first amyloid protein is attached and incubated at 28 to 38°C, preferably 30 to 36°C to aggregate with the first amyloid protein.
  • the second amyloid protein in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, can be brought into contact with the plate to which the first amyloid protein is attached at pH 4 to 9. there is.
  • the second amyloid protein may be brought into contact with a plate to which the first amyloid protein is attached at pH 5 to 8.
  • the second amyloid protein can be brought into contact with the plate to which the first amyloid protein is attached at pH 7 to 8.
  • the second amyloid protein When the second amyloid protein is brought into contact with the plate to which the first amyloid protein is attached at a pH of less than 5 or more than pH 8, the aggregation effect with the first amyloid protein is low, making it difficult to use the second amyloid protein in the screening method of the present invention.
  • the amyloid protein should be brought into contact with the plate to which the first amyloid protein is attached at pH 5 to pH 8, preferably pH 7 to pH 8.
  • Another aspect includes a plate to which a first amyloid protein is attached; and a kit for screening a protein aggregation inhibitor or aggregation dissolving agent containing a second amyloid protein to which an indicator substance is bound.
  • the first and second amyloid proteins may be full-length proteins or protein fragments.
  • the first and second amyloid proteins may be self-assembled.
  • the aggregation inhibitor may be treated simultaneously or sequentially with the second amyloid protein.
  • the aggregation dissolving agent may be used after aggregating the first amyloid protein and the second amyloid protein.
  • the present invention provides a method for searching the action site of an amyloid protein aggregation inhibitor or aggregation dissolving agent using amyloid protein and fragments thereof, for example, in subjects with degenerative brain disease or at risk of developing degenerative brain disease, Provides a method for searching the action site (specifically, the action domain) of an amyloid protein aggregation inhibitor or aggregation dissolving agent to treat or prevent.
  • Another aspect includes (1) providing a plate to which a plurality of first amyloid protein fragments are attached; (2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or (2-2) contacting the plate with a second amyloid protein bound to an indicator material and then treating the candidate material as a protein aggregation dissolving agent; (3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1); or (3-2) screening the site where the amyloid protein aggregation inhibitor candidate or aggregation dissolving agent candidate acts, including the step of measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2). Provides a way to do this.
  • the method may additionally include sorting the degree of inhibition or dissolution of aggregation between the first amyloid fragment and the second amyloid protein by the first amyloid protein fragment.
  • step (2-1) or (2-2) may further include inducing aggregation of the first and second amyloid proteins.
  • the amyloid protein may be amyloid beta, tau protein, or alpha-synuclein.
  • the protein fragment is a contiguous 6 to 15 mer within an amyloid protein
  • the plurality of protein fragments may include two or more overlapping contiguous amino acid residues.
  • the step of measuring the aggregation rate may be done by analyzing the intensity of the indicator material and comparing it for each protein fragment.
  • the method may further include determining a protein fragment for which a high degree of aggregation inhibition or aggregation dissolution is measured as the site of action of the candidate substance. Specifically, one protein fragment for which a high degree of aggregation inhibition or aggregation dissolution is measured is determined as the action site, or an overlapping amino acid residue of a plurality of protein fragments for which aggregation inhibition or aggregation dissolution is measured to be high is determined as the action site. It may include more steps. More specifically, the step of determining a protein fragment as an action site is selecting a protein fragment that shows a higher value when comparing the aggregation inhibition rate (%) or aggregation dissolution rate (%) for each protein fragment through the method shown in the examples. It may include.
  • the action site may be present in two or more amyloid proteins.
  • the amyloid protein may be amyloid beta, tau protein, or alpha-synuclein.
  • the protein fragment may be 6 to 15 mer. Specifically, the protein fragment may be a 6mer.
  • the aggregation inhibitor or aggregation inhibitor candidate may be treated simultaneously or sequentially with the second amyloid protein.
  • the aggregation-dissolving agent or aggregation-dissolving agent candidate may be treated after aggregating the first amyloid protein and the second amyloid protein.
  • the method of screening the site where the amyloid protein aggregation inhibitor candidate or aggregation dissolving agent candidate acts is a molecular docking simulation used to predict and analyze the target site of the A ⁇ aggregation inhibitor or dissolving agent.
  • molecular docking simulation can not only be used to verify research results, but can also be used to develop aggregation inhibitors or solubilizers that specifically inhibit or release aggregation in the A ⁇ sequence using a rational design method.
  • the screening method for candidates for inhibiting or dissolving amyloid protein aggregation not only can the efficacy of existing known amyloid protein aggregation inhibitors or solubilizers be compared, but also the target domains of amyloid protein aggregation inhibitors or solubilizers are analyzed, and further the mechanism is determined. Since it can reveal the identity of amyloid proteins, especially A ⁇ , it can be useful in developing inhibitors or solubilizers that dissolve the aggregation of specific protein sequences.
  • Figure 1 is a schematic diagram illustrating a method for producing amyloid beta and amyloid beta fragments with cysteine attached to the C-terminus.
  • Figure 2 is a schematic diagram showing the types of amyloid hexamer fragments with cysteine attached to the C-terminus.
  • Figure 3 is a graph showing the results of analysis of hexameric fragments of amyloid beta 1-6 to amyloid beta 10-15 by reverse phase-HPLC.
  • Figure 4 is a graph showing the results of analysis of hexameric fragments of amyloid beta 11-16 to amyloid beta 20-25 by reverse phase-HPLC.
  • Figure 5 is a graph showing the results of analysis of hexameric fragments of amyloid beta 21-26 to amyloid beta 30-35 by reverse phase-HPLC.
  • Figure 6 is a graph showing the results of reverse phase-HPLC analysis of hexameric fragments of amyloid beta 31-36 to amyloid beta 37-42 and amyloid beta 1-42 .
  • Figure 7 is a graph confirming the optimal conditions (peptide concentration, fixation time, temperature, pH, and fluorescent label concentration) of A3 and MAP.
  • Figure 8 shows the results of confirming structural, morphological, and cohesive changes in A3 and MAP peptides.
  • Figure 9 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
  • Figure 10 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
  • Figure 11 is a graph confirming the aggregation inhibition effect of amyloid beta aggregation inhibitors (Curcumin and Scyllol-inositol) using A3.
  • Figure 12 is a graph confirming the aggregation-dissolving effect of amyloid beta aggregation-dissolving agents (Necrostantin-1 and Sunitinib) using A3.
  • Figure 13 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
  • Figure 14 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
  • Figure 15 is a graph confirming the aggregation-dissolving action site of the amyloid beta aggregation inhibitor (Curcumin) using MAP.
  • Figure 16 is a graph confirming the aggregation inhibition site of amyloid beta aggregation inhibitor (Scyllol-inositol) using MAP.
  • Figure 17 is a graph confirming the aggregation-dissolving action site of amyloid beta aggregation-dissolving agent (Necrostantin-1) using MAP.
  • Figure 18 is a graph confirming the aggregation inhibitory action site of amyloid beta aggregation dissolving agent (Sunitinib) using MAP.
  • Figure 19 is a schematic diagram showing the A3 manufacturing method.
  • Figure 20 is a schematic diagram showing the MAP manufacturing method.
  • a ⁇ 1-42 hexamer fragment with 37 C-terminal cysteines attached and full-length A ⁇ 1-42 with C-terminal cysteine attached were separated using microwaves.
  • -Fluorenylmethyloxycarbonyl was synthesized through a solid phase peptide synthesis method, and is shown in Figure 2.
  • the first cysteine at the C-terminus was attached using symmetric anhydride activation.
  • 2.2 mmol of Fmoc-Cys(trt)-OH, 1.0 mmol of DIC, and 51 mg of DMAP were dissolved in 2 mL of DMF/DCM (1:1, v/v) solution, and then 0.25 mmol of Wang's solution swollen in DMF. It was reacted with resin LS for 1.5 hours.
  • the remaining protein sequences were sequentially coupled using an automated peptide synthesizer.
  • cleavage cocktail (92.5:2.5:2.5:2.5 TFA/deionized water/TIS/DODT, v/v/v/v) was added and reacted for 4 hours. TFA was evaporated using a rotary evaporator, cooled anhydrous ether was added, and centrifuged for 15 minutes to produce an unrefined peptide, which is shown in Figure 1.
  • the A ⁇ 1-42 hexamer fragment with 37 C-terminal cysteines attached was purified and analyzed using reversed-HPLC, and is shown in Figures 3 to 6.
  • Figure 1 is a schematic diagram illustrating a method for producing amyloid beta and amyloid beta fragments with cysteine attached to the C-terminus.
  • Figure 2 is a schematic diagram showing the types of amyloid hexamer fragments with cysteine attached to the C-terminus.
  • Figures 3 to 6 are graphs showing the results of analysis of amyloid beta hexamer fragments by reversed phase-HPLC.
  • washing buffer 0.1 M Na 3 PO 4 , 0.15 M NaCl, 0.05% Tween 20, pH 7.2
  • the full-length A ⁇ 1-42 -Cys solution was mixed (50 ⁇ g/mL, 5% DMSO) in binding buffer (0.1 M Na 3 PO 4 , 0.15 M NaCl, 10 mM EDTA).
  • binding buffer 0.1 M Na 3 PO 4 , 0.15 M NaCl, 10 mM EDTA
  • 100 ⁇ L of aqueous peptide solution (10 ⁇ M) was added to each well (peptide well) and incubated at 24°C for 24 hours.
  • 200 ⁇ L of washing buffer was added and washing was performed.
  • the cysteine solution was mixed with binding buffer (10 ⁇ g/mL).
  • Figure 19 is a schematic diagram showing the A3 manufacturing method.
  • washing buffer 0.1 M Na 3 PO 4 , 0.15 M NaCl, 0.05% Tween-20, pH 7.2
  • binding buffer 0.1 M Na 3 PO 4 , 0.15 M NaCl, 10 mM EDTA
  • full length A ⁇ 1-42 -Cys solution and binding buffer were mixed (50 ⁇ g/mL, 5% DMSO).
  • aqueous peptide solution (10 ⁇ M) was added to each well (peptide well) and incubated at 24°C for 24 hours. 200 ⁇ L of washing buffer was added and washing was performed.
  • the cysteine solution was mixed with binding buffer (10 ⁇ g/mL). 200 ⁇ L of the cysteine solution (10 ⁇ g/mL) mixed with the binding buffer was added to each well (blank well) and incubated at 24°C for 1 hour. 200 ⁇ L of washing buffer was added and washing was performed, which is shown in Figure 20.
  • Figure 20 is a schematic diagram showing the MAP manufacturing method.
  • fluor-A ⁇ 1-42 concentration conditions 0.1, 1, 10 ⁇ M
  • fluor-A ⁇ 1-42 peptide was incubated at a concentration of 10 ⁇ M for 24 hours under seven pH conditions (1.2, 3.2, 5.2, 7.2, 9.2, 11.2, 13.2), and to find the optimal conditions for generating A ⁇ aggregation in the wells of the plate, fluor-A ⁇ 1-42 peptide was used at a concentration of 10 ⁇ M for 24 hours under six temperature conditions (0, 4 , 24, 34, 54, 74°C). The results are shown in Figure 7.
  • Figure 7 is a graph confirming the optimal conditions (peptide concentration, fixation time, temperature, pH, and fluorescent label concentration) of A3 and MAP.
  • each peptide was treated on the plate at a concentration of 1 ⁇ M and confirmed with the 6E10 antibody, and the results showed that the peptide was effectively attached to the plate in the neutral pH range, especially at pH 6.5- At 7.2, it was confirmed that the peptide was most effectively attached to the plate (Figure 7D).
  • the optimal conditions for immobilizing the purified peptide on the plate are to immobilize the peptide at a concentration of 0.5 to 5 ⁇ M at 28-32°C and pH 6.5-7.2 for 23-25 hours, and then apply a fluorescent label to induce aggregation. It was confirmed that peptides can most effectively induce aggregation with purified peptides under the conditions of 8 to 12 ⁇ M, pH 7-8, and 30-36°C temperature.
  • Figure 8 shows the results of confirming structural, morphological, and cohesive changes in A3 and MAP peptides.
  • Fluor-A ⁇ 1-42 (10 ⁇ M), a conjugate of the fluorescent substance Flamma 552 and A ⁇ 1-42, and an A ⁇ aggregation inhibitor were added to A3 and reacted at room temperature for 24 hours ((+)inhibitor/(+)fluor-A ⁇ 1-42 A3).
  • control wells were treated with only fluor-A ⁇ 1-42 ((-)inhibitor/(+)fluor-A ⁇ 1-42 A3).
  • Substances inhibiting A ⁇ aggregation were searched by comparing the fluorescence intensity of Flamma 552 between the control and wells containing the aggregation inhibitor, and these are shown in Figures 9 and 11.
  • Figure 9 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
  • Figure 11 is a graph confirming the aggregation inhibition effect of amyloid beta aggregation inhibitors (Curcumin and Scyllol-inositol) using A3.
  • aggregation inhibitors were searched through the A ⁇ aggregation inhibitor search method using A3. Curcumin was confirmed to inhibit A ⁇ aggregation by 49.0% and scyllo-inositol by 24.7%.
  • Fluor-A ⁇ 1-42 (10 ⁇ M) was added to A3 and aggregated in the plate at 37°C for 6 hours ((-)dissociator/(+)fluor-A ⁇ 1-42 A3). Afterwards, A ⁇ aggregation dissolving agent was added and treated at room temperature for 24 hours ((+)dissociator/(+)fluor-A ⁇ 1-42 A3). Substances that dissolve aggregation were searched by comparing the fluorescence intensity of Flamma 552 after aggregation for 6 hours and the degree to which the fluorescence intensity of Flamma 552 was reduced after treatment with an aggregation dissolving agent, and these are shown in Figures 10 and 12.
  • Figure 10 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
  • Figure 12 is a graph confirming the aggregation-dissolving effect of amyloid beta aggregation-dissolving agents (Necrostantin-1 and Sunitinib) using A3.
  • agglutination dissolving agents were searched through the A ⁇ aggregation dissolving agent search method using A3. Necrostatin-1 was confirmed to dissolve A ⁇ aggregates by 42.5% and sunitnib by 65.2%.
  • A3 and MAP can provide a platform for effectively screening A ⁇ aggregation inhibitors or dissolution candidates.
  • Fluor-A ⁇ 1-42 (10 ⁇ M), a conjugate of the fluorescent substance Flamma 552 and A ⁇ 1-42, and an A ⁇ aggregation inhibitor were added to the MAP plate and reacted at room temperature for 24 hours ((+)inhibitor/(+)fluor -A ⁇ 1-42 MAP).
  • control wells were treated with only fluor-A ⁇ 1-42 ((-)inhibitor/(+)fluor-A ⁇ 1-42 MAP).
  • Figure 15 is a graph confirming the aggregation-dissolving action site of the amyloid beta aggregation inhibitor (Curcumin) using MAP.
  • Figure 16 is a graph confirming the aggregation inhibition site of amyloid beta aggregation inhibitor (Scyllol-inositol) using MAP.
  • curcumin As shown in Figure 15, through the method of identifying A ⁇ aggregation inhibitors using MAP, curcumin was identified as HQKLVFFA (A ⁇ 14-19 , A ⁇ 15-20 , A ⁇ 16-21 ), GAIIGLM (A ⁇ 29-34 , A ⁇ 30-35 ), and GGVVIA (A ⁇ 37-42 ) domains, and was confirmed to inhibit aggregation by binding to these regions.
  • VHHQKLVFF VHHQKLVFF
  • VGSNKGAIIGL A ⁇ 24-29
  • a ⁇ 25-30 A ⁇ 26-31 , A ⁇ 27-32 , A ⁇ 28-33 , A ⁇ 29-34
  • LMVGGV A ⁇ 34-39
  • GGVVIA A ⁇ 37-42
  • Fluor-A ⁇ 1-42 (10 ⁇ M) was added to MAP and aggregated in the plate at 37°C for 6 hours ((-)dissociator/(+)fluor-A ⁇ 1-42 MAP). Afterwards, A ⁇ aggregation dissolving agent was added and treated at room temperature for 24 hours ((+)dissociator/(+)fluor-A ⁇ 1-42 MAP).
  • the site of action of the substance is identified by confirming which fragments the A ⁇ aggregation dissolving agent dissolves. and this is shown in Figure 14.
  • Figure 17 is a graph confirming the aggregation-dissolving action site of amyloid beta aggregation-dissolving agent (Necrostantin-1) using MAP.
  • Figure 18 is a graph confirming the aggregation inhibitory action site of amyloid beta aggregation dissolving agent (Sunitinib) using MAP.
  • necrostatin-1 was identified as HHQKLVFF (A ⁇ 13-18 , A ⁇ 14-19 , A ⁇ 15-20 ) and GLMVGGVVIA (A ⁇ 33-38 , A ⁇ 34-39 ). , A ⁇ 35-40 , A ⁇ 36-41 , and A ⁇ 37-42 ) domains and can be confirmed to cause aggregation and dissolution. It was confirmed that the A ⁇ 36-41 region was specifically released when compared to the full-length A ⁇ 1-42 .
  • sunitnib was identified as AIIGLM (A ⁇ 30-35 ), and LMVGGVVIA (A ⁇ 34-39 , A ⁇ 35-40 , A ⁇ 36-41 , A ⁇ 37-42 ). It was confirmed that it specifically targets domains and inhibits aggregation.
  • MAP can be useful in analyzing the target domains of A ⁇ aggregation inhibitors or solubilizers and developing inhibitors or solubilizers that unravel the aggregation of specific protein sequences of A ⁇ .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)

Abstract

The present invention relates to methods for screening for a drug that interferes with or modulates binding between amyloid proteins and screening for a target domain of the drug. According to a method for screening for a candidate material for inhibiting or disaggregating amyloid protein aggregation, according to an aspect, the efficacy between previously known amyloid protein aggregation inhibitors or disaggregators can be compared, target domains of amyloid protein aggregation inhibitors or disaggregators can be analyzed, and furthermore, the mechanisms thereof can be verified. Thus, the method can be effectively used in the development of an inhibitor or disaggregator that specifically disaggregates the sequence of an amyloid protein, particularly Aβ protein.

Description

아밀로이드 단백질 응집 억제 또는 용해 후보물질 스크리닝 방법Method for screening candidates for inhibiting or dissolving amyloid protein aggregation
아밀로이드 단백질의 응집 억제제(inhibitor) 또는 용해제(disaggregator)를 스크리닝하는 방법, 키트 및 아밀로이드 단백질 응집 억제제 또는 용해제의 응집 억제 또는 용해 작용 부위를 스크리닝하는 방법에 관한 것이다. It relates to a method for screening an amyloid protein aggregation inhibitor or disaggregator, a kit, and a method for screening the aggregation inhibition or disaggregator site of an amyloid protein aggregation inhibitor or disaggregator.
아밀로이드증(amyloidosis)은 아밀로이드(amyloid)로 불리는 비정상적인 단백질이 조직에 축적되어 나타나는 질환이다. 아밀로이드는 7-13 nm의 직경 및 베타 시트(β-sheet) 구조를 가지고 현미경 하에서 보면 섬유성 형태로 나타나는 단백질 덩어리로, Thioflavin T(ThioT) 및 콩고 레드(congo red)에 의해 염색되는 특징을 가지고 있다. 아밀로이드는 정상적인 체내에서 발견되지 않으며, 현재까지 36가지 단백질이 이를 형성할 수 있음이 보고되어 있다(Picken, Acta Haematol.(2020), 143:322-334). 대표적인 아밀로이드증에는 알츠하이머 병(Alzheimer's disease), 파킨슨 병(Parkinson's disease), 헌팅턴 병(Huntington disease) 및 프라이온 병(prion disease)등의 신경질환이 포함되며, 그 외에도 원인 단백질과 영향받은 장기에 따라 다양한 양상을 가지는 다수의 아밀로이드증이 존재한다.Amyloidosis is a disease that occurs when abnormal proteins called amyloid accumulate in tissues. Amyloid is a protein mass with a diameter of 7-13 nm and a beta-sheet structure that appears in a fibrous form when viewed under a microscope, and is characterized by staining with Thioflavin T (ThioT) and Congo red. there is. Amyloid is not found in the normal body, and to date, it has been reported that 36 proteins can form it (Picken, Acta Haematol. (2020), 143:322-334). Representative amyloidosis includes neurological diseases such as Alzheimer's disease, Parkinson's disease, Huntington disease, and prion disease. In addition, there are various types of amyloidosis depending on the causative protein and affected organ. There are a number of amyloidoses with different presentations.
이러한 아밀로이드증 중 하나인, 알츠하이머병의 중요한 병리적 특징은 노인성 플라크(senile plaque)라고 불리는 펩티드 응집체의 형성이며, 이는 시냅스 기능장애 및 신경세포의 사멸을 유발한다. 이러한 노인성 플라크의 성분 중 하나는 40-42 아미노산 길이의 아밀로이드-베타(amyloid-beta)이다. 아밀로이드-베타 단량체는 올리고머, 원섬유(protofibril) 및 베타-시트가 풍부한 섬유로 자가조립(self-assemble)되기 쉽고, 신경 독성의 발병과 관련이 있다.An important pathological feature of Alzheimer's disease, one of these amyloidoses, is the formation of peptide aggregates called senile plaques, which cause synaptic dysfunction and neuronal death. One of the components of these senile plaques is amyloid-beta, which is 40-42 amino acids long. Amyloid-beta monomers are prone to self-assemble into oligomers, protofibrils, and beta-sheet-rich fibers and are associated with the pathogenesis of neurotoxicity.
아밀로이드-베타 플라크와 신경 독성 사이의 상관관계에 대해서는 아직 명확히 밝혀진 바가 없지만, 아밀로이드-베타의 중간 올리고머 또는 응집체로의 자가 조립은 알츠하이머병과 같은 뇌신경 질환의 발병과 관련된 것으로 여겨지고 있다. Although the correlation between amyloid-beta plaques and neurotoxicity has not yet been clearly elucidated, the self-assembly of amyloid-beta into intermediate oligomers or aggregates is believed to be involved in the pathogenesis of cranial nerve diseases such as Alzheimer's disease.
그러나 현재까지 개발된 아밀로이드성 질병 치료제의 대다수는 환자에게 면역 반응을 일으키는 Aβ 단편(fragment) 또는 Aβ 내의 특정 에피토프(epitope)에 대한 항체를 사용하거나 케미컬 의약품을 적용하는 방식을 채용하고 있다. 케미컬 의약품의 경우에는 현재까지 아밀로이드성 질병에 대한 근본적인 치료제는 부재한 상태이다. However, the majority of amyloid disease treatments developed to date use antibodies against Aβ fragments or specific epitopes within Aβ that cause an immune response in patients, or apply chemical drugs. In the case of chemical drugs, there is currently no fundamental treatment for amyloid disease.
따라서, 본 발명자들은 아밀로이드 단백질, 특히 아밀로이드 베타 플라크 생성을 억제하는 퇴행성 뇌질환 치료제를 스크리닝하는 방법을 제공하기 위해 본 발명을 완성하였다. Accordingly, the present inventors completed the present invention to provide a method of screening for a therapeutic agent for degenerative brain diseases that inhibits the formation of amyloid protein, especially amyloid beta plaques.
일 양상은 아밀로이드 단백질의 응집을 억제, 저해 또는 용해하는 물질을 스크리닝하는 방법을 제공하는 것이다.One aspect is to provide a method of screening for substances that inhibit, inhibit, or dissolve the aggregation of amyloid proteins.
다른 양상은 아밀로이드 단백질의 응집을 억제, 저해 또는 용해하는 물질을 스크리닝하는 키트를 제공하는 것이다.Another aspect is to provide a kit for screening substances that inhibit, inhibit, or dissolve the aggregation of amyloid proteins.
또 다른 양상은 아밀로이드 단백질의 응집을 억제, 저해 또는 용해하는 물질의 응집 억제 또는 용해 작용 부위를 스크리닝하는 방법을 제공하는 것이다.Another aspect is to provide a method of screening for an aggregation inhibition or dissolution site of action of a substance that inhibits, inhibits, or dissolves the aggregation of amyloid protein.
본 발명은 아밀로이드 단백질의 응집 억제제 또는 응집 용해제를 스크리닝하는 방법, 예를 들어 퇴행성 뇌질환을 갖고 있거나 또는 퇴행성 뇌질환이 발생할 위험이 있는 대상체에서 퇴행성 뇌질환을 치료하거나 예방하기 위해 아밀로이드 단백질 응집 억제제 또는 용해제 후보 물질을 스크리닝하는 방법 및 키트를 제공한다. The present invention provides a method for screening for an aggregation inhibitor or an aggregation-dissolving agent of amyloid protein, for example, an amyloid protein aggregation inhibitor or Methods and kits for screening candidate solubilizers are provided.
일 양상은 (1) 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계;One aspect includes (1) providing a plate to which a first amyloid protein is attached;
(2-1) 단백질 응집 억제제 후보물질과 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시키는 단계; 또는 (2-2) 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시킨 후 단백질 응집 용해제 후보물질을 처리하는 단계; 및, (3-1) 상기 (2-1) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하여 무처리 대조군 대비 변화시킨 응집 억제제 후보물질을 선별하는 단계; 또는 (3-2) 상기 (2-2) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하여 무처리 대조군 대비 변화시킨 응집 용해제 후보물질을 선별하는 단계를 포함하는 단백질 응집 억제제 또는 응집 용해제를 스크리닝하는 방법을 제공한다.(2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or (2-2) contacting a second amyloid protein to which an indicator substance is bound to the plate and then treating the candidate substance as a protein aggregation dissolving agent; And, (3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1) and selecting an aggregation inhibitor candidate changed compared to the untreated control group; or (3-2) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2) and selecting an aggregation resolving agent candidate changed compared to the untreated control group. A method for screening solubilizers is provided.
일 구체예에 있어서, 상기 (2-1) 또는 (2-2) 단계는 제1 및 제2 아밀로이드 단백질의 응집을 유도하는 단계를 더 포함하는 것일 수 있다. In one embodiment, step (2-1) or (2-2) may further include inducing aggregation of the first and second amyloid proteins.
일 구체예에 있어서, 상기 단백질 응집 억제제 또는 응집 용해제를 스크리닝하는 방법은 상기 아밀로이드 단백질의 응집률을 변화시킨 물질 또는 무처리 대조군에 비해 아밀로이드 단백질의 응집률을 변화시킨 후보 물질을 아밀로이드 단백질 응집 억제제 또는 응집 용해제로서 선별하는 단계를 더 포함하는 것일 수 있다. In one embodiment, the method of screening for the protein aggregation inhibitor or aggregation dissolving agent is an amyloid protein aggregation inhibitor or a candidate material that changes the aggregation rate of amyloid protein compared to the substance that changed the aggregation rate of the amyloid protein or the untreated control. It may further include a step of selecting as a coagulant dissolving agent.
본 명세서에서 “아밀로이드 단백질”은 베타 평판 구조의 형태로 서로 엉켜 응집되는 단백질로, 아밀로이드 침착 형성이 가능한 단백질 또는 그 절편을 의미한다. 본 발명에서 아밀로이드 단백질은 아밀로이드 단백질 응집체를 구성하는 개별 단백질을 의미할 수 있다. As used herein, “amyloid protein” refers to a protein that tangles and aggregates in the form of a beta plate structure, and refers to a protein or a fragment thereof capable of forming amyloid deposits. In the present invention, amyloid protein may refer to individual proteins constituting amyloid protein aggregates.
일 구체예에 있어서, 상기 아밀로이드 단백질은 아밀로이드 베타, 타우 단백질, 알파-시누클레인, 프리온, 아밀로이드성 단백질, 아밀로이드 트랜스티레틴, 루이소체, 플리글루타민, 토신 A 및 AIMP2 단백질일 수 있다. In one embodiment, the amyloid protein may be amyloid beta, tau protein, alpha-synuclein, prion, amyloidogenic protein, amyloid transthyretin, Lewy body, fliglutamine, Torsin A, and AIMP2 protein.
본 명세서에서 용어 “아밀로이드 베타(amyloid beta)”는 36 내지 43개 아미노산을 포함하는 펩타이드 분자로서, 아밀로이드 플라크의 주된 성분으로, 알츠하이머병의 발병에 관여하는 것으로 알려져 있다. 상기 아밀로이드 베타 펩타이드 분자는 응집하여 신경세포 독성 올리고머를 형성하여, 퇴행성 뇌질환을 유발한다. 상기 아밀로이드 베타 펩타이드 분자는 아밀로이드 전구체 단백질(amyloid precursor protein (APP); UniProtKB P05067)을 베타 세크레타제(beta secretase)와 감마 세크레타제(gamma secretase)로 절단하여 얻어지는 것일 수 있다. As used herein, the term “amyloid beta” is a peptide molecule containing 36 to 43 amino acids, is a major component of amyloid plaques, and is known to be involved in the development of Alzheimer's disease. The amyloid beta peptide molecules aggregate to form neurotoxic oligomers, causing degenerative brain diseases. The amyloid beta peptide molecule may be obtained by chopping amyloid precursor protein (APP); UniProtKB P05067) with beta secretase and gamma secretase.
일 구체예에 있어서, 상기 아밀로이드 단백질은 자가조립(self-assembly)하는 것일 수 있다. In one embodiment, the amyloid protein may self-assemble.
본 명세서에서 용어 “아밀로이드 단백질 응집체”는 질환 상태와 연관되어 있는 아밀로이드 단백질 응집체(aggregate)을 지칭한다. 이러한 질환 상태에는 세포의 사멸, 또는 2개 이상 세포들간 뉴런 신호전달의 부분적이거나 완전한 상실이 포함되나, 이에 제한되는 것은 아니다. 아밀로이드 단백질 응집체는 세포의 내부, 또는 세포의 외부에 위치할 수 있다. 구체적으로, 아밀로이드 단백질 응집체는 신경독성을 유발할 수 있고, 후술하는 퇴행성 뇌질환의 원인이 될 수 있다. As used herein, the term “amyloid protein aggregate” refers to amyloid protein aggregates that are associated with disease states. These disease states include, but are not limited to, cell death, or partial or complete loss of neuronal signaling between two or more cells. Amyloid protein aggregates may be located inside the cell or outside the cell. Specifically, amyloid protein aggregates can cause neurotoxicity and cause degenerative brain diseases described later.
일 구체예에 있어서, 상기 아밀로이드 단백질 응집체는 아밀로이드 단백질의 중합체, 피브릴 전구체, 피브릴(fibril), 플라크(plaque), 동종 단백질과 결합된 단백질 단량체 및 중합체를 포함할 수 있다. 상기 피브릴은 아밀로이드 단백질의 올리고머(oligomer)를 포함할 수 있다. 구체적으로, 상기 자가조립 단백질은 수용성 아밀로이드 베타 응집체 및/또는 불수용성 아밀로이드 베타 응집체, 아밀로이드 베타 올리고머, 아밀로이드 베타 프로토피브릴(protofibril), 아밀로이드 베타 피브릴(fibril), 및 아밀로이드 베타 플라크(plaque)일 수 있다.In one embodiment, the amyloid protein aggregates may include polymers of amyloid proteins, fibril precursors, fibrils, plaques, protein monomers and polymers bound to homologous proteins. The fibrils may include oligomers of amyloid protein. Specifically, the self-assembling protein may be soluble amyloid beta aggregates and/or water-insoluble amyloid beta aggregates, amyloid beta oligomers, amyloid beta protofibrils, amyloid beta fibrils, and amyloid beta plaques. You can.
일 구체예에 있어서, 상기 제1 또는 제2 아밀로이드 단백질은 전장 단백질 (full-length protein) 및/또는 단백질 단편(fragment)인 것일 수 있다. In one embodiment, the first or second amyloid protein may be a full-length protein and/or a protein fragment.
일 구체예에 있어서, 상기 전장 단백질은 아밀로이드 베타1-36, 아밀로이드 베타1-38, 아밀로이드 베타1-40, 또는 아밀로이드 베타1-42 펩타이드인 것일 수 있다. In one embodiment, the full-length protein may be an amyloid beta 1-36, amyloid beta 1-38, amyloid beta 1-40, or amyloid beta 1-42 peptide.
본 명세서에서 용어 “단백질 단편(fragment)”는 단백질의 절단형을 의미한다. 구체적으로, 상기 단백질 단편은 상기 전장 단백질의 절단된 일부분을 의미한다.As used herein, the term “protein fragment” refers to a truncated form of a protein. Specifically, the protein fragment refers to a cleaved portion of the full-length protein.
일 구체예에 있어서, 상기 단백질 단편은 아밀로이드 단백질 내의 연속되는 단백질일 수 있다. 구체적으로, 아밀로이드 단백질 내의 연속되는 6 내지 15mer인 것일 수 있다. In one embodiment, the protein fragment may be a continuous protein within an amyloid protein. Specifically, it may be a continuous 6 to 15 mer within the amyloid protein.
일 구체예에 있어서, 상기 단백질 단편은 6 내지 15mer인 것일 수 있다. In one embodiment, the protein fragment may be 6 to 15 mer.
일 구체예에 있어서, 상기 단백질 단편은 6 내지 14mer인 것일 수 있다. 일 구체예에 있어서, 상기 단백질 단편은 6 내지 13mer인 것일 수 있다. 일 구체예에 있어서, 상기 단백질 단편은 6 내지 12mer인 것일 수 있다. 일 구체예에 있어서, 상기 단백질 단편은 6 내지 11mer인 것일 수 있다. 일 구체예에 있어서, 상기 단백질 단편은 6 내지 10mer인 것일 수 있다. 일 구체예에 있어서, 상기 단백질 단편은 6 내지 9mer인 것일 수 있다. In one embodiment, the protein fragment may be 6 to 14 mer. In one embodiment, the protein fragment may be 6 to 13 mer. In one embodiment, the protein fragment may be 6 to 12 mer. In one embodiment, the protein fragment may be 6 to 11 mer. In one embodiment, the protein fragment may be 6 to 10 mer. In one embodiment, the protein fragment may be 6 to 9 mer.
일 구체예에 있어서, 상기 단백질 단편은 아밀로이드 베타 6mer(이하, 아밀로이드 베타 육량체라고도 함.)인 것일 수 있다. In one embodiment, the protein fragment may be amyloid beta 6mer (hereinafter also referred to as amyloid beta hexamer).
상기 단백질 단편은 DAEFRH일 수 있다. 상기 단백질 단편은 AEFRHD일 수 있다. 상기 단백질 단편은 EFRHDS일 수 있다. 상기 단백질 단편은 FRHDSG일 수 있다. 상기 단백질 단편은 RHDSGY일 수 있다. 상기 단백질 단편은 HDSGYE일 수 있다. 상기 단백질 단편은 DSGYEV일 수 있다. 상기 단백질 단편은 SGYEVH일 수 있다. 상기 단백질 단편은 GYEVHH일 수 있다. 상기 단백질 단편은 YEVHHQ일 수 있다. 상기 단백질 단편은 EVHHQK일 수 있다. 상기 단백질 단편은 VHHQKL일 수 있다. 상기 단백질 단편은 HHQKLV일 수 있다. 상기 단백질 단편은 HQKLVF일 수 있다. 상기 단백질 단편은 QKLVFF일 수 있다. 상기 단백질 단편은 KLVFFA일 수 있다. 상기 단백질 단편은 LVFFAE일 수 있다. 상기 단백질 단편은 VFFAED일 수 있다. 상기 단백질 단편은 FFAEDV일 수 있다. 상기 단백질 단편은 FAEDVG일 수 있다. 상기 단백질 단편은 AEDVGS일 수 있다. 상기 단백질 단편은 EDVGSN일 수 있다. 상기 단백질 단편은 DVGSNK일 수 있다. 상기 단백질 단편은 DVGSNG일 수 있다. 상기 단백질 단편은 VGSNGA일 수 있다. 상기 단백질 단편은 GSNGAI일 수 있다. 상기 단백질 단편은 SNGAII일 수 있다. 상기 단백질 단편은 NGAIIG일 수 있다. 상기 단백질 단편은 GAIIGL일 수 있다. 상기 단백질 단편은 AIIGLM일 수 있다. 상기 단백질 단편은 IIGLMV일 수 있다. 상기 단백질 단편은 IGLMVG일 수 있다. 상기 단백질 단편은 GLMVGG일 수 있다. 상기 단백질 단편은 LMVGGV일 수 있다. 상기 단백질 단편은 MVGGVV일 수 있다. 상기 단백질 단편은 VGGVVI일 수 있다. 상기 단백질 단편은 GGVVIA일 수 있다.The protein fragment may be DAEFRH. The protein fragment may be AEFRHD. The protein fragment may be EFRHDS. The protein fragment may be FRHDSG. The protein fragment may be RHDSGY. The protein fragment may be HDSGYE. The protein fragment may be DSGYEV. The protein fragment may be SGYEVH. The protein fragment may be GYEVHH. The protein fragment may be YEVHHQ. The protein fragment may be EVHHQK. The protein fragment may be VHHQKL. The protein fragment may be HHQKLV. The protein fragment may be HQKLVF. The protein fragment may be QKLVFF. The protein fragment may be KLVFFA. The protein fragment may be LVFFAE. The protein fragment may be VFFAED. The protein fragment may be FFAEDV. The protein fragment may be FAEDVG. The protein fragment may be AEDVGS. The protein fragment may be EDVGSN. The protein fragment may be DVGSNK. The protein fragment may be DVGSNG. The protein fragment may be VGSNGA. The protein fragment may be GSNGAI. The protein fragment may be SNGAII. The protein fragment may be NGAIIG. The protein fragment may be GAIIGL. The protein fragment may be AIIGLM. The protein fragment may be IIGLMV. The protein fragment may be IGLMVG. The protein fragment may be GLMVGG. The protein fragment may be LMVGGV. The protein fragment may be MVGGVV. The protein fragment may be VGGVVI. The protein fragment may be GGVVIA.
일 구체예에 있어서, 상기 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계에 있어서, 제1 아밀로이드 단백질은 C말단에 결합된 시스테인으로 플레이트에 부착되는 것일 수 있다. 구체적으로, 전장 단백질 아밀로이드 베타1-42 또는 아밀로이드 베타 육량체는 C말단에 결합된 시스테인을 통해 플레이트에 부착되는 것일 수 있다.In one embodiment, in the step of providing a plate to which the first amyloid protein is attached, the first amyloid protein may be attached to the plate by cysteine bound to the C-terminus. Specifically, the full-length protein amyloid beta 1-42 or amyloid beta hexamer may be attached to the plate through a cysteine bound to the C-terminus.
일 구체예에 있어서, 상기 시스테인은 대칭 무수화물 활성(systemic anhydride activation)을 통해 제1 아밀로이드 단백질의 C-말단에 결합된 것일 수 있으나 이에 제한되는 것은 아니다. 아밀로이드 단백질의 C- 또는 N-말단에 시스테인의 결합은 통상의 당업자에게 자명한 방법으로 수행될 수 있다. In one embodiment, the cysteine may be bound to the C-terminus of the first amyloid protein through systemic anhydride activation, but is not limited thereto. Binding of cysteine to the C- or N-terminus of amyloid protein can be performed by methods that are obvious to those skilled in the art.
일 구체예에 있어서, 상기 아밀로이드 단백질 C-말단에 시스테인이 결합된 펩타이드는 고체상 펩타이드 합성 방법을 통해 합성되는 것일 수 있다. 구체적으로, 마이크로파를 이용한 9-플루오렌닐메틸옥시카르보닐 고체상 펩타이드 합성에 의한 것일 수 있으나 이에 제한되는 것은 아니다. In one embodiment, the peptide having cysteine bound to the C-terminus of the amyloid protein may be synthesized through a solid-phase peptide synthesis method. Specifically, it may be by synthesizing 9-fluorenylmethyloxycarbonyl solid phase peptide using microwaves, but is not limited thereto.
일 구체예에 있어서, C-말단에 시스테인 결합된 전장 아밀로이드 베타1-42 또는 아밀로이드 베타 육량체는 대칭 무수화물 활성(systemic anhydride activation)을 통해 C-말단에 시스테인(cysteine)을 결합시킨 후, 마이크로파를 이용한 9-플루오렌닐메틸옥시카르보닐 고체상 펩타이드 합성으로 제조될 수 있다.In one embodiment, full-length amyloid beta 1-42 or amyloid beta hexamer with cysteine bound to the C-terminus is formed by binding cysteine to the C-terminus through systemic anhydride activation, and then activating it in the microwave. It can be produced by 9-fluorenylmethyloxycarbonyl solid-phase peptide synthesis using .
일 구체예에 있어서, 상기 C-말단에 시스테인 결합된 전장 아밀로이드 베타1-42 또는 아밀로이드 베타 육량체는 C-말단에 시스테인이 플레이트의 작용기와 반응하여 부착될 수 있고, 구체적으로 말레이미드(maleimide) 및 이의 유도체, 아지리딘 및 이의 유도체, 아크릴로일 및 이의 유도체, 또는 아릴할라이드 및 이의 유도체와 반응하여 부착될 수 있다. In one embodiment, the full-length amyloid beta 1-42 or amyloid beta hexamer with cysteine bound to the C-terminus may be attached to the C-terminus by reacting cysteine with a functional group of the plate, specifically maleimide. and its derivatives, aziridine and its derivatives, acryloyl and its derivatives, or aryl halide and its derivatives.
일 구체예에 있어서, 상기 제1 아밀로이드 단백질은 단량체 형태로 플레이트에 부착되고, 제2 아밀로이드 단백질과 접촉하여 응집체를 이루기 전에 단량체 형태를 유지하도록 플레이트에 부착된 것일 수 있다. In one embodiment, the first amyloid protein may be attached to the plate in monomeric form and may be attached to the plate to maintain the monomeric form before forming aggregates by contact with the second amyloid protein.
일 구체예에 있어서, 상기 방법은 (1) C말단에 결합된 시스테인을 통해 전장 아밀로이드 베타1-42 또는 아밀로이드 베타 육량체가 부착된 플레이트를 제공하는 단계; (2-1) 상기 플레이트에 단백질 응집 억제제 후보물질과 지표물질이 결합된 전장 아밀로이드 베타1-42를 접촉시키는 단계, 또는 (2-2) 지표물질이 결합된 전장 아밀로이드 베타1-42를 접촉시킨 후, 단백질 응집 용해제 후보물질을 처리하는 단계; 및, (3-1) 무처리 대조군 대비 아밀로이드 베타 간의 응집을 억제하는 응집 억제제 후보물질을 선별하는 단계; 또는 무처리 대조군 대비 응집된 아밀로이드 베타 간의 응집을 용해시키는 응집 용해제 후보물질을 선별 단계를 포함하는 단백질 응집 억제제 또는 응집 용해제를 스크리닝하는 방법을 제공하는 것이다.In one embodiment, the method includes the steps of (1) providing a plate to which full-length amyloid beta 1-42 or amyloid beta hexamer is attached through a cysteine bound to the C terminus; (2-1) contacting the plate with full-length amyloid beta 1-42 to which a protein aggregation inhibitor candidate and an indicator are bound, or (2-2) contacting full-length amyloid beta 1-42 to which an indicator is bound. Afterwards, processing the protein aggregation dissolving candidate material; And, (3-1) selecting an aggregation inhibitor candidate that inhibits aggregation between amyloid beta livers compared to the untreated control group; Alternatively, a method of screening a protein aggregation inhibitor or an aggregation-dissolving agent is provided, which includes a step of selecting a candidate for an aggregation-dissolving agent that dissolves aggregation between aggregated amyloid beta compared to an untreated control group.
상기 방법은 아밀로이드 베타 간의 응집률을 측정하는 단계를 더 포함할 수 있다. The method may further include measuring the aggregation rate between amyloid beta.
일 구체예에 있어서, 상기 제2 아밀로이드 단백질을 접촉시키는 단계는 제1 아밀로이드 단백질과 응집을 유도하는 단계를 더 포함하는 것일 수 있다. In one embodiment, the step of contacting the second amyloid protein may further include inducing aggregation with the first amyloid protein.
일 구체예에 있어서, 상기 제2 아밀로이드 단백질은 지표물질과 결합된 것일 수 있다. In one embodiment, the second amyloid protein may be bound to an indicator substance.
상기 지표물질은 제1 아밀로이드 단백질에 제2 아밀로이드 단백질간 결합정도를 측정하기 위한 것; 및 응집 억제제, 응집 용해제, 응집 억제제 후보물질 또는 응집 용해제 후보물질에 의해 제1 아밀로이드 단백질 및 제2 아밀로이드 단백질의 응집 억제능 또는 응집 용해능, 구체적으로 응집 저해정도 또는 응집 용해 정도를 측정하기 위한 것일 수 있다. The indicator material is for measuring the degree of binding between the first amyloid protein and the second amyloid protein; And it may be used to measure the aggregation inhibition or aggregation dissolution ability of the first amyloid protein and the second amyloid protein by the aggregation inhibitor, aggregation dissolving agent, aggregation inhibitor candidate, or aggregation dissolving agent candidate, specifically, the degree of aggregation inhibition or aggregation dissolution. there is.
일 구체예에 있어서, 상기 지표물질은 형광물질 또는 형광 단백질 단편인 것일 수 있다. In one embodiment, the indicator may be a fluorescent substance or a fluorescent protein fragment.
일 구체예에 있어서, 상기 형광물질은 로다민, 쿠마린, 에보블루(EvoBlue), 옥사진, 카보피로닌, 나프탈렌, 비페닐, 안트라센, 페난트렌, 피렌 또는 카바졸을 기본 골격으로 갖는 형광염료 또는 상기 형광염료의 유도체일 수 있다. In one embodiment, the fluorescent material is a fluorescent dye having rhodamine, coumarin, EvoBlue, oxazine, carbopyronine, naphthalene, biphenyl, anthracene, phenanthrene, pyrene, or carbazole as a basic skeleton, or It may be a derivative of the fluorescent dye.
구체적으로, 상기 형광물질는 플루오레세인(Fluorescein), CR110:카복시로다민 110:로다민 그린(상표명), TAMRA:카복시테트라메틸로다민: TMR, 카복시로다민 6G : CR6G, BODIPY FL(상표명) : 4,4-디플루오로-5,7-디메틸-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, BODIPY 493/503(상표명) : 4,4-디플루오로-1,3,5,7-테트라메틸-4-보라-3a,4a-디아자-s-인다센-8-프로피온산, BODIPY R6G(상표명) : 4,4-디플루오로-5-(4-페닐-1,3-부타디에닐)-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, BODIPY 558/568(상표명) : 4,4-디플루오로-5-(2-티에닐)-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, BODIPY 564/570(상표명) : 4,4-디플루오로-5-스티릴-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, BODIPY 576/589(상표명) : 4,4-디플루오로-5-(2-피롤릴)-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, BODIPY 581/591(상표명) : 4,4-디플루오로-5-(4-페닐-1,3-부타디에닐)-4-보라-3a,4a-디아자-s-인다센-3-프로피온산, EvoBlue10(상표명), EvoBlue30(상표명), MR121, ATTO 655(상표명), ATTO 680(상표명), ATTO 700(상표명), ATTO MB2(상표명), Alexa Fluor 350(상표명), Alexa Fluor405(상표명), Alexa Fluor 430(상표명), Alexa Fluor 488(상표명), Alexa Fluor 532(상표명), Alexa Fluor546(상표명), Alexa Fluor 555(상표명), Alexa Fluor 568(상표명), Alexa Fluor 594(상표명), Alexa Fluor633(상표명), Alexa Fluor 680(상표명), Alexa Fluor 700(상표명), Alexa Fluor 750(상표명), Alexa Fluor790(상표명), Flamma 496(상표명), Flamma 507(상표명), Flamma 530(상표명), Flamma 552(상표명), Flamma560(상표명), Flamma 575(상표명), Flamma 581(상표명), Flamma 648(상표명), Flamma 675(상표명), Flamma749(상표명), Flamma 774(상표명), Flamma 775(상표명), Rhodamine Red-X(상표명), Texas Red-X(상표명), 5(6)-TAMRA-X(상표명), 5TAMRA(상표명), Indocyanine green (ICG) 및 2-((E)-2-((E)-2-(4-(2-carboxyethyl)phenoxy)-3-((E)-2-(3,3-dimethyl-5-sulfonato-1-(3-(tri-methyl ammonio)-propyl)indolin-2-ylidene)ethylidene)cyclohex-1-enyl)vinyl)-3,3-dimethyl-1-(3-(trimethyl ammonio)-propyl)-3H-indolium-5-sulfonate disodium bromide (ZW800-1)으로 이루어진 군 중에서 선택되는 것일 수 있다.Specifically, the fluorescent substance is Fluorescein, CR110:Carboxyrhodamine 110:Rhodamine Green (trade name), TAMRA:Carboxytetramethylrhodamine:TMR, Carboxyrhodamine 6G:CR6G, BODIPY FL (trade name): 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-propionic acid, BODIPY 493/503 (trade name): 4,4-difluoro- 1,3,5,7-Tetramethyl-4-bora-3a,4a-diaza-s-indacene-8-propionic acid, BODIPY R6G (trade name): 4,4-difluoro-5-(4- Phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza-s-indacene-3-propionic acid, BODIPY 558/568 (trade name): 4,4-difluoro-5-( 2-thienyl)-4-bora-3a,4a-diaza-s-indacene-3-propionic acid, BODIPY 564/570 (trade name): 4,4-difluoro-5-styryl-4-bora -3a,4a-diaza-s-indacene-3-propionic acid, BODIPY 576/589 (trade name): 4,4-difluoro-5-(2-pyrrolyl)-4-bora-3a,4a- Diaza-s-indacene-3-propionic acid, BODIPY 581/591 (trade name): 4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a, 4a-diaza-s-indacene-3-propionic acid, EvoBlue10 (trade name), EvoBlue30 (trade name), MR121, ATTO 655 (trade name), ATTO 680 (trade name), ATTO 700 (trade name), ATTO MB2 (trade name), Alexa Fluor 350 (trade name), Alexa Fluor405 (trade name), Alexa Fluor 430 (trade name), Alexa Fluor 488 (trade name), Alexa Fluor 532 (trade name), Alexa Fluor546 (trade name), Alexa Fluor 555 (trade name), Alexa Fluor 568 (trade name), Alexa Fluor 594 (trade name), Alexa Fluor633 (trade name), Alexa Fluor 680 (trade name), Alexa Fluor 700 (trade name), Alexa Fluor 750 (trade name), Alexa Fluor790 (trade name), Flamma 496 (trade name), Flamma 507 (brand name), Flamma 530 (brand name), Flamma 552 (brand name), Flamma560 (brand name), Flamma 575 (brand name), Flamma 581 (brand name), Flamma 648 (brand name), Flamma 675 (brand name), Flamma749 (brand name) ), Flamma 774 (trade name), Flamma 775 (trade name), Rhodamine Red-X (trade name), Texas Red-X (trade name), 5(6)-TAMRA-X (trade name), 5TAMRA (trade name), Indocyanine green ( ICG) and 2-((E)-2-((E)-2-(4-(2-carboxyethyl)phenoxy)-3-((E)-2-(3,3-dimethyl-5-sulfonato- 1-(3-(tri-methyl ammonio)-propyl)indolin-2-ylidene)ethylidene)cyclohex-1-enyl)vinyl)-3,3-dimethyl-1-(3-(trimethyl ammonio)-propyl)- It may be selected from the group consisting of 3H-indolium-5-sulfonate disodium bromide (ZW800-1).
일 구체예에 있어서, 상기 형광 단백질 단편은 venus, Cerulean, Citrine, mKate가 있으며 서로 다른 색깔을 띄는 형광 단백질들이거나, 또는 그 일부인 것일 수 있다.In one embodiment, the fluorescent protein fragments include Venus, Cerulean, Citrine, and mKate, and may be fluorescent proteins of different colors, or may be parts thereof.
본 명세서에서 용어 “아밀로이드 단백질 응집 억제제”는 아밀로이드 단백질의 집적을 억제하는 것을 의미하고, “아밀로이드 단백질 응집 용해제”는 이미 형성된 아밀로이드 단백질의 응집을 분해하는 물질을 의미할 수 있다. 상기 응집 억제제 또는 응집 용해제는 아밀로이드 단백질의 자가조립(self-assembly)을 억제하거나 이미 형성된 자가조립 단백질의 응집을 분해하는 물질을 의미할 수 있다. 구체적으로, 수용성 단백질 응집체 및/또는 불수용성 단백질 응집체의 집적을 억제(inhibit aggregation)하거나 용해(disaggregate)하는 물질을 의미할 수 있다. As used herein, the term “amyloid protein aggregation inhibitor” refers to inhibiting the accumulation of amyloid proteins, and the term “amyloid protein aggregation dissolving agent” may refer to a substance that decomposes the aggregation of already formed amyloid proteins. The aggregation inhibitor or aggregation dissolving agent may refer to a substance that inhibits self-assembly of amyloid proteins or breaks down aggregation of already formed self-assembled proteins. Specifically, it may refer to a substance that inhibits aggregation or disaggregates water-soluble protein aggregates and/or water-insoluble protein aggregates.
일 구체예에 있어서, 상기 응집 억제제 또는 응집 용해제는 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인(α-cynuclein)의 응집을 억제 또는 용해하는 것일 수 있다. In one embodiment, the aggregation inhibitor or aggregation dissolving agent may inhibit or dissolve the aggregation of amyloid beta, tau protein, or alpha-synuclein.
일 구체예에 있어서, 상기 응집 억제제는 아밀로이드의 섬유화(amyloid fibrillation) 및 아밀로이드 섬유 응집을 억제하는 것이며, 응집 용해제는 응집된 응집체를 분해(dissociation)의 의미를 포함한다. 구체적으로, 수용성 아밀로이드 베타 응집체 및/또는 불수용성 아밀로이드 베타 응집체, 아밀로이드 베타 올리고머, 아밀로이드 베타 프로토피브릴(protofibril), 아밀로이드 베타 피브릴(fibril), 및 아밀로이드 베타 플라크(plaque) 등을 용해하거나 집적을 억제하는 물질이다. In one embodiment, the aggregation inhibitor inhibits amyloid fibrillation and amyloid fiber aggregation, and the aggregation dissolving agent includes the dissociation of aggregates. Specifically, it dissolves or accumulates soluble amyloid beta aggregates and/or water-insoluble amyloid beta aggregates, amyloid beta oligomers, amyloid beta protofibrils, amyloid beta fibrils, and amyloid beta plaques. It is an inhibitory substance.
일 구체예에 있어서, 상기 아밀로이드 단백질 응집 억제제 또는 응집 용해제는 퇴행성 뇌질환 치료제일 수 있다.In one embodiment, the amyloid protein aggregation inhibitor or aggregation-dissolving agent may be a treatment for degenerative brain disease.
일 구체예에 있어서, 상기 응집 억제제 또는 응집 억제제 후보물질은 제2 아밀로이드 단백질과 동시에 또는 순차적으로 처리되는 것일 수 있다. In one embodiment, the aggregation inhibitor or aggregation inhibitor candidate may be treated simultaneously or sequentially with the second amyloid protein.
일 구체예에 있어서, 상기 응집 억제제는 제2 아밀로이드 단백질과 동시에 처리되는 것일 수 있다. 구체적으로, 아밀로이드 베타 응집 저해제 커큐민(curcumin) 또는 실로-이노시톨(scyllo-inositol)은 fluor-Aβ1-42과 동시에 처리되는 것일 수 있다. In one embodiment, the aggregation inhibitor may be treated simultaneously with the second amyloid protein. Specifically, the amyloid beta aggregation inhibitor curcumin or scyllo-inositol may be treated simultaneously with fluor-Aβ 1-42 .
일 구체예에서, 상기 응집 용해제 또는 응집 용해제 후보물질은 제1 아밀로이드 단백질 및 제2 아밀로이드 단백질을 응집시킨 후, 처리되는 것일 수 있다.In one embodiment, the aggregation-dissolving agent or aggregation-dissolving agent candidate may be treated after aggregating the first amyloid protein and the second amyloid protein.
구체적으로, 아밀로이드 베타 응집 용해제는 네크로스타틴-1(necrostatin-1) 및 수니티닙(sunitnib)은 fluor-Aβ1-42과 순차적으로, 예를 들어 24시간 후에 처리되는 것일 수 있다. Specifically, the amyloid beta aggregation dissolving agent necrostatin-1 and sunitnib may be treated sequentially with fluor-Aβ 1-42 , for example, after 24 hours.
상기 단백질 응집 억제제 또는 응집 용해제는 퇴행성 뇌질환의 예방 및 치료에 사용되어 (1)아밀로이드 베타의 축적, (2) 뇌세포의 노화, (3) 시냅스의 손실, (4)말초 면역 세포 축적과 같은 퇴행성 뇌질환, 구체적으로 알츠하이머 병의 병리적 증상을 완화시키는 것일 수 있다. The protein aggregation inhibitor or aggregation-dissolving agent is used in the prevention and treatment of degenerative brain diseases such as (1) accumulation of amyloid beta, (2) aging of brain cells, (3) loss of synapses, and (4) accumulation of peripheral immune cells. It may alleviate the pathological symptoms of degenerative brain disease, specifically Alzheimer's disease.
본 명세서에서 용어 "퇴행성 뇌질환"은 뇌의 퇴행성 변화와 관련된 모든 질환, 특히, 뇌 및/또는 뇌신경세포에서의 아밀로이드-베타의 응집 등의 요인에 의하여 유발될 수 있는 모든 질환 (뇌질환)을 포괄적으로 기재하기 위하여 사용된다. 일 구체예에 있어서, 상기 퇴행성 뇌질환은 치매, 알츠하이머병(Alzheimer's disease), 전임상 알츠하이머병(preclinical alzheimer's disease), 파킨슨병(Parkinson's disease), 헌팅턴병 (Huntington's disease), 경도인지장애(mild cognitive impairment), 대뇌 아밀로이드 맥관병증, 다운증후근, 아밀로이드성 뇌졸증(stroke), 전신성 아밀로이드병, 더취(Dutch)형 아밀로이드증, 니만-픽병(Niemann-Pick disease), 노인성 치매, 근위축성 측삭 경화증(amyotrophic lateral sclerosis), 척수소뇌성 운동실조증(spinocerebellar atrophy), 뚜렛 증후군(Tourette's syndrome), 프리드리히 보행실조(Friedrich's ataxia), 마차도-조셉병(Machado-Joseph's disease), 루이 소체 치매(Lewy body dementia), 근육긴장이상(dystonia), 진행성 핵상 마비(progressive supranuclear palsy) 및 전두측두엽 치매(frontotemporal dementia)으로 이루어진 군으로부터 선택되는 어느 하나인 것일 수 있다. 바람직하게, 상기 퇴행성 뇌질환은 알츠하이머병일 수 있다. As used herein, the term “degenerative brain disease” refers to all diseases related to degenerative changes in the brain, especially all diseases (brain diseases) that can be caused by factors such as aggregation of amyloid-beta in the brain and/or brain nerve cells. It is used to describe comprehensively. In one embodiment, the degenerative brain disease includes dementia, Alzheimer's disease, preclinical Alzheimer's disease, Parkinson's disease, Huntington's disease, and mild cognitive impairment. , cerebral amyloid angiopathy, Down syndrome, amyloid stroke, systemic amyloid disease, Dutch amyloidosis, Niemann-Pick disease, senile dementia, amyotrophic lateral sclerosis, Spinocerebellar atrophy, Tourette's syndrome, Friedrich's ataxia, Machado-Joseph's disease, Lewy body dementia, dystonia ), progressive supranuclear palsy, and frontotemporal dementia. Preferably, the degenerative brain disease may be Alzheimer's disease.
본 명세서에서 용어 "알츠하이머병"이란 노인성 치매와 호환적으로 사용되고, 노인성 플라크, 신경염증 엉킴(tangles), 및진행성 신경 손실로 특징되는 특정 퇴행성 뇌질환과 관련된 정신적인 퇴화를 수반하는 질병을 의미할 수 있다.As used herein, the term "Alzheimer's disease" is used interchangeably with senile dementia and refers to a disease involving mental deterioration associated with a specific degenerative brain disease characterized by senile plaques, neuroinflammatory tangles, and progressive nerve loss. You can.
본 명세서에서 용어 "파킨슨병"이란 종종 운동 기능 및 언어능력을 손상시키는 중추 신경계의 만성 및 진행성 퇴행성 질환을 의미할 수 있다.As used herein, the term "Parkinson's disease" may refer to a chronic and progressive degenerative disease of the central nervous system that often impairs motor function and language ability.
본 명세서에서 용어 "헌팅턴병"이란 헌팅턴 단백질을 암호화하는 유전자 내 3 염기 반복 팽창에 의해 야기되어, 무도증, 정신이상, 치매 등이 수반되는 신경퇴행성 질환을 의미할 수 있다.As used herein, the term "Huntington's disease" may refer to a neurodegenerative disease that is caused by a 3-base repeat expansion in the gene encoding the huntingtin protein and is accompanied by chorea, psychosis, dementia, etc.
본 명세서에서 용어 "루게릭병"이란 운동신경세포만 선택적으로 사멸되는 질환으로 대뇌 피질의 상위운동신경세포와 뇌간 및 척수의 아래운동신경세포 모두가 점차적으로 파괴되는 질환을 의미할 수 있다.As used herein, the term "Lou Gehrig's disease" refers to a disease in which only motor neurons are selectively killed, and may refer to a disease in which both upper motor neurons in the cerebral cortex and lower motor neurons in the brainstem and spinal cord are gradually destroyed.
본 명에서에서 용어 "픽병(pick's disease)"는 뇌의 신경세포의 진행성 파괴를 나타내는 질환을 의미할 수있다.As used herein, the term “Pick's disease” may refer to a disease that causes progressive destruction of nerve cells in the brain.
본 발명의 용어 "타우병증"이란 타우 단백질(세포 내 마이크로튜불-관련 단백질과 밀접하게 관련된 패밀리)이 뇌조직에 비정상적으로 축적됨에 의하여 뇌신경이 손상되는 신경퇴행성 질환을 의미할 수 있다.The term "tauopathy" of the present invention may refer to a neurodegenerative disease in which cranial nerves are damaged due to abnormal accumulation of tau protein (a family closely related to intracellular microtubul-related proteins) in brain tissue.
일 구체예에 있어서, 상기 방법은 C말단에 결합된 시스테인으로 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계; In one embodiment, the method includes providing a plate to which a first amyloid protein is attached with cysteine bound to the C-terminus;
(2-1) 상기 플레이트에 단백질 응집 억제제 후보물질과 지표물질이 결합된 제2 아밀로이드 단백질을 접촉시키는 단계, 또는 (2-2) 지표물질이 결합된 제2 아밀로이드 단백질을 접촉시킨 후, 단백질 응집 용해제 후보물질을 처리하는 단계; 및, (3-1) 무처리 대조군 대비 제1 아밀로이드 단백질과 제2 아밀로이드 단백질 간의 응집을 억제하는 응집 억제제 후보물질을 선별하는 단계; 또는 무처리 대조군 대비 제1 아밀로이드 단백질과 제2 아밀로이드 단백질 간의 응집을 용해시키는 응집 용해제 후보물질을 선별 단계를 포함하는 단백질 응집 억제제 또는 응집 용해제를 스크리닝하는 방법을 제공하는 것이다.(2-1) contacting the plate with a second amyloid protein to which a protein aggregation inhibitor candidate and an indicator are bound, or (2-2) contacting a second amyloid protein to which an indicator is bound, and then protein aggregation. Processing the solubilizer candidate; And, (3-1) selecting an aggregation inhibitor candidate that inhibits aggregation between the first amyloid protein and the second amyloid protein compared to the untreated control group; Alternatively, a method for screening a protein aggregation inhibitor or an aggregation dissolving agent comprising the step of selecting a candidate for an aggregation dissolving agent that dissolves the aggregation between the first amyloid protein and the second amyloid protein compared to the untreated control group is provided.
상기 방법은 제1 아밀로이드 단백질과 제2 아밀로이드 단백질 간의 응집률을 측정하는 단계를 더 포함할 수 있다. The method may further include measuring the aggregation rate between the first amyloid protein and the second amyloid protein.
일 구체예에 있어서, 상기 제2 아밀로이드 단백질을 접촉시키는 단계는 제1 아밀로이드 단백질과 응집을 유도하는 단계를 더 포함하는 것일 수 있다. In one embodiment, the step of contacting the second amyloid protein may further include inducing aggregation with the first amyloid protein.
구체적으로, 전장 단백질 아밀로이드 베타1-42 또는 단백질 단편 아밀로이드 베타 육량체는 C말단에 결합된 시스테인으로 플레이트에 부착되는 것일 수 있다. Specifically, the full-length protein amyloid beta 1-42 or the protein fragment amyloid beta hexamer may be attached to the plate with cysteine bound to the C-terminus.
상기 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계에 있어서, 플레이트에 처리되는 제1아밀로이드 단백질의 농도는 0.00001μM 내지 50μM일 수 있다. 일 구체예에 있어서, 상기 제1아밀로이드 단백질의 농도는 0.01μM 내지 20μM일 수 있다. 일 구체예에 있어서, 상기 제1아밀로이드 단백질의 농도는 0.1μM 내지 15μM일 수 있다. 일 구체예에 있어서, 상기 제1아밀로이드 단백질의 농도는 0.1μM 내지 10μM일 수 있다. 일 구체예에 있어서, 상기 제1 아밀로이드 단백질의 농도는 0.5μM 내지 5μM일 수 있다. 구체적으로, 상기 제1아밀로이드 단백질의 농도는 1μM 일 수 있다. 상기 플레이트에 처리되는 제1아밀로이드 단백질의 농도가 0.1μM이하의 경우에는 플레이트에 고정되는 제1아밀로이드 단백질의 고정 효과가 낮아 본 발명의 스크리닝 방법으로 사용하기가 어렵고, 플레이트에 처리되는 제1아밀로이드 단백질의 농도가 10μM초과의 경우에는 플레이트에 고정되는 제1아밀로이드 단백질의 고정 효과가 포화(saturation)되고, 비특이적 결합 등 고정되지 않고 플레이트에 존재하는 제1아밀로이드 단백질의 농도가 높아져 응집율 측정에 오류를 발생시킬 수 있는 바, 본 발명의 제1아밀로이드 단백질의 농도는0.1μM 내지 10μM, 바람직하게는 0.5μM 내지 5μM이여야 한다.In the step of providing the plate to which the first amyloid protein is attached, the concentration of the first amyloid protein treated on the plate may be 0.00001 μM to 50 μM. In one embodiment, the concentration of the first amyloid protein may be 0.01 μM to 20 μM. In one embodiment, the concentration of the first amyloid protein may be 0.1 μM to 15 μM. In one embodiment, the concentration of the first amyloid protein may be 0.1 μM to 10 μM. In one embodiment, the concentration of the first amyloid protein may be 0.5 μM to 5 μM. Specifically, the concentration of the first amyloid protein may be 1 μM. If the concentration of the first amyloid protein processed on the plate is 0.1 μM or less, the fixation effect of the first amyloid protein fixed on the plate is low, making it difficult to use it in the screening method of the present invention. If the concentration exceeds 10 μM, the fixation effect of the first amyloid protein immobilized on the plate is saturated, and the concentration of the first amyloid protein present on the plate without being fixed due to non-specific binding increases, causing an error in measuring the aggregation rate. As it can be produced, the concentration of the first amyloid protein of the present invention should be 0.1 μM to 10 μM, preferably 0.5 μM to 5 μM.
일 구체예에 있어서, 상기 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계에 있어서, 상기 제1아밀로이드 단백질은 플레이트에 처리하고 20℃ 내지 40℃에서 인큐베이션하여 플레이트에 부착된 것일 수 있다. 일 구체예에 있어서, 상기 아밀로이드 단백질은 25 내지 35℃에서 인큐베이션하여 플레이트에 부착된 것일 수 있다. 구체적으로, 상기 아밀로이드 단백질은 28 내지 32℃에서 인큐베이션하여 플레이트에 부착된 것일 수 있다. 상기 제1 아밀로이드 단백질을 25℃ 미만 또는 35℃초과에서 인큐베이션하여 부착시킬 경우, 플레이트에 부착 효율이 현저히 낮아 용집 억제제 또는 응집 용해제 스크리닝 방법에 적용할 수 없는 바, 제1 아밀로이드 단백질은 플레이트에 처리하고 25℃ 내지 35℃, 바람직하게는 28 내지 32℃에서 인큐베이션하여 플레이트에 부착되어야 한다. In one embodiment, in the step of providing a plate to which the first amyloid protein is attached, the first amyloid protein may be attached to the plate by treating the plate and incubating at 20°C to 40°C. In one embodiment, the amyloid protein may be attached to the plate by incubation at 25 to 35°C. Specifically, the amyloid protein may be attached to the plate by incubation at 28 to 32°C. When the first amyloid protein is attached by incubating at less than 25°C or more than 35°C, the attachment efficiency to the plate is significantly low and cannot be applied to the screening method for aggregation inhibitor or aggregation dissolving agent. Therefore, the first amyloid protein is treated on the plate. It should be adhered to the plate by incubation at 25°C to 35°C, preferably 28 to 32°C.
일 구체예에 있어서, 상기 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계에 있어서, 상기 제1아밀로이드 단백질은 플레이트에 처리하고 12내지 36시간동안 인큐베이션하여 플레이트에 부착되는 것일 수 있다. 일 구체예에 있어서, 상기 아밀로이드 단백질은 20 내지 26시간 동안 인큐베이션하여 플레이트에 부착되는 것일 수 있다. 구체적으로, 상기 제1아밀로이드 단백질은 23 내지 25시간 동안 인큐베이션하여 플레이트에 부착되는 것일 수 있다. 상기 아밀로이드 단백질을 20시간 미만으로 인큐베이션할 경우, 플레이트에 고정되는 제1아밀로이드 단백질의 고정 효과가 낮아 본 발명의 스크리닝 방법으로 사용하기가 어렵고, 26시간 이상으로 인큐베이션할 경우 고정 효과가 포화되므로, 실험의 신속성 및 사업성(경제성) 측면에서 상기 제1아밀로이드 단백질은 20 내지 26시간, 바람직하게 23 내지 25시간 동안 인큐베이션하여 플레이트에 부착되어여 한다. In one embodiment, in the step of providing a plate to which the first amyloid protein is attached, the first amyloid protein may be attached to the plate by treating the plate and incubating for 12 to 36 hours. In one embodiment, the amyloid protein may be attached to the plate by incubating for 20 to 26 hours. Specifically, the first amyloid protein may be attached to the plate by incubating for 23 to 25 hours. When the amyloid protein is incubated for less than 20 hours, the fixation effect of the first amyloid protein immobilized on the plate is low, making it difficult to use it in the screening method of the present invention, and when the amyloid protein is incubated for more than 26 hours, the fixation effect is saturated, so the experiment In terms of speed and business feasibility, the first amyloid protein should be attached to the plate by incubating for 20 to 26 hours, preferably 23 to 25 hours.
일 구체예에 있어서, 상기 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계에 있어서, 상기 제1아밀로이드 단백질은 pH 4 내지 9에서 플레이트에 부착된 것일 수 있다. 일 구체예에 있어서, 상기 제1아밀로이드 단백질은 pH 5 내지 8에서 플레이트에 부착된 것일 수 있다. 구체적으로 제1아밀로이드 단백질은 pH 6.5 내지 7.2에서 플레이트에 부착된 것일 수 있다. 상기 제1 아밀로이드 단백질을 pH 5미만, pH 8 초과에서 플레이트에 부착할 경우, 제1아밀로이드 단백질의 고정 효과가 낮아 본 발명의 스크리닝 방법으로 사용시 스크리닝 효율이 떨어지므로, 제1아밀로이드 단백질은 pH 5 내지 pH 8, 바람직하게는 pH 6.5 내지 7.2에서 플레이트에 부착되어야 한다.In one embodiment, in providing a plate to which the first amyloid protein is attached, the first amyloid protein may be attached to the plate at pH 4 to 9. In one embodiment, the first amyloid protein may be attached to the plate at pH 5 to 8. Specifically, the first amyloid protein may be attached to the plate at pH 6.5 to 7.2. When the first amyloid protein is attached to the plate at pH less than 5 or more than pH 8, the fixation effect of the first amyloid protein is low and the screening efficiency is reduced when used in the screening method of the present invention. Therefore, the first amyloid protein is used at pH 5 or higher. It should adhere to the plate at pH 8, preferably pH 6.5 to 7.2.
일 구체예에 있어서, 상기 제2아밀로이드 단백질을 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시키는 단계에 있어서, 상기 제2아밀로이드 단백질은 제1 아밀로이드 단백질이 부착된 플레이트에 1μM 내지 15μM 범위 내의 농도로 처리될 수 있다. 일 구체예에 있어서, 5μM 내지 15μM 범위 내의 농도로 처리될 수 있다. 구체적으로, 8μM 내지 12μM 범위 내의 농도로 처리될 수 있다. 상기 제2아밀로이드 단백질을 5μM 미만으로 처리할 경우, 제1 및 제2아밀로이드 단백질간 응집율이 낮아 본 발명의 스크리닝 방법으로 사용하기가 어렵고, 상기 제2아밀로이드 단백질을 15μM 초과하여 처리할 경우 비특이적 결합 등 제1아밀로이드 단백질과 응집되지 않고 플레이트에 존재하는 제2아밀로이드 단백질의 농도가 높아져 응집율 측정에 오류를 발생시킬 수 있는 바, 제2아밀로이드 단백질은 제1 아밀로이드 단백질이 부착된 플레이트에 5μM 내지 15μM, 바람직하게는 8μM 내지 12μM로 처리되어야 한다.In one embodiment, in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, the second amyloid protein is treated at a concentration in the range of 1 μM to 15 μM to the plate to which the first amyloid protein is attached. It can be. In one embodiment, it can be treated at a concentration ranging from 5 μM to 15 μM. Specifically, it can be treated at a concentration within the range of 8 μM to 12 μM. When the second amyloid protein is treated at less than 5 μM, the aggregation rate between the first and second amyloid proteins is low, making it difficult to use in the screening method of the present invention, and when the second amyloid protein is treated at more than 15 μM, non-specific binding occurs. The concentration of the second amyloid protein that does not aggregate with the first amyloid protein and is present on the plate increases, which may cause errors in measuring the aggregation rate. The second amyloid protein is added to the plate to which the first amyloid protein is attached at 5 μM to 15 μM. , preferably at 8 μM to 12 μM.
일 구체예에 있어서, 상기 제2아밀로이드 단백질을 제1아밀로이드 단백질이 부착된 플레이트에 접촉시키는 단계에 있어서, 상기 제2아밀로이드 단백질은 제1 아밀로이드 단백질이 부착된 플레이트에 처리하고 20℃ 내지 40℃에서 인큐베이션하여 제1 아밀로이드 단백질과 응집시킬 수 있다. 일 구체예에 있어서, 상기 아밀로이드 단백질은 28 내지 38℃에서 인큐베이션하여 제1 아밀로이드 단백질과 응집시킬 수 있다. 구체적으로, 상기 아밀로이드 단백질은 30 내지 36℃에서 인큐베이션하여 제1 아밀로이드 단백질과 응집시킬 수 있다. 상기 제1 아밀로이드 단백질을 28℃ 미만 또는 38℃초과에서 인큐베이션하여 제1 아밀로이드 단백질과 응집시킬 경우, 제1 아밀로이드 단백질과 응집 효율이 현저히 낮아 용집 억제제 또는 응집 용해제 스크리닝 방법에 적용할 수 없는 바, 제2 아밀로이드 단백질은 제1 아밀로이드 단백질이 부착된 플레이트에 처리하고 28 내지 38℃, 바람직하게는 30 내지 36℃에서 인큐베이션하여 제1 아밀로이드 단백질과 응집시켜야 한다.In one embodiment, in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, the second amyloid protein is treated to the plate to which the first amyloid protein is attached and incubated at 20°C to 40°C. By incubation, it can be aggregated with the first amyloid protein. In one embodiment, the amyloid protein can be aggregated with the first amyloid protein by incubating at 28 to 38°C. Specifically, the amyloid protein may be incubated at 30 to 36° C. to aggregate with the first amyloid protein. When the first amyloid protein is incubated at less than 28°C or more than 38°C to cause aggregation with the first amyloid protein, the efficiency of aggregation with the first amyloid protein is significantly low and cannot be applied to the screening method for an aggregation inhibitor or an aggregation dissolving agent. 2 The amyloid protein should be treated on a plate to which the first amyloid protein is attached and incubated at 28 to 38°C, preferably 30 to 36°C to aggregate with the first amyloid protein.
일 구체예에 있어서, 상기 제2아밀로이드 단백질을 제1아밀로이드 단백질이 부착된 플레이트에 접촉시키는 단계에 있어서, 상기 제2아밀로이드 단백질은 pH 4 내지 9에서 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시킬 수 있다. 일 구체예에 있어서, 상기 제2아밀로이드 단백질은 pH 5 내지 8에서 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시킬 수 있다. 구체적으로, 상기 제2아밀로이드 단백질은 pH 7 내지 8에서 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시킬 수 있다. 상기 제2 아밀로이드 단백질을 pH 5미만, pH 8 초과에서 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시킬 경우, 제1아밀로이드 단백질과 응집 효과가 낮아 본 발명의 스크리닝 방법으로 사용하기가 어려운 바, 제2아밀로이드 단백질은 pH 5 내지 pH 8, 바람직하게는 pH 7 내지 pH 8에서 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시켜야 한다.In one embodiment, in the step of contacting the second amyloid protein with the plate to which the first amyloid protein is attached, the second amyloid protein can be brought into contact with the plate to which the first amyloid protein is attached at pH 4 to 9. there is. In one embodiment, the second amyloid protein may be brought into contact with a plate to which the first amyloid protein is attached at pH 5 to 8. Specifically, the second amyloid protein can be brought into contact with the plate to which the first amyloid protein is attached at pH 7 to 8. When the second amyloid protein is brought into contact with the plate to which the first amyloid protein is attached at a pH of less than 5 or more than pH 8, the aggregation effect with the first amyloid protein is low, making it difficult to use the second amyloid protein in the screening method of the present invention. The amyloid protein should be brought into contact with the plate to which the first amyloid protein is attached at pH 5 to pH 8, preferably pH 7 to pH 8.
다른 양상은 제1 아밀로이드 단백질이 부착된 플레이트; 및 지표물질이 결합된 제2 아밀로이드 단백질을 포함하는 단백질 응집 억제제 또는 응집 용해제를 스크리닝 하기 위한 키트를 제공한다.Another aspect includes a plate to which a first amyloid protein is attached; and a kit for screening a protein aggregation inhibitor or aggregation dissolving agent containing a second amyloid protein to which an indicator substance is bound.
일 구체예에 있어서, 상기 제1 및 제2 아밀로이드 단백질은 전장 단백질(full-length protein)또는 단백질 단편인 것일 수 있다. In one embodiment, the first and second amyloid proteins may be full-length proteins or protein fragments.
일 구체예에 있어서, 상기 제1 및 제2 아밀로이드 단백질은 자가조립되는 것일 수 있다.In one embodiment, the first and second amyloid proteins may be self-assembled.
상기 “자가조립 단백질”, “단백질 단편” “응집 억제제”및 “응집 용해제”는 상기한 바와 같다. The “self-assembling protein,” “protein fragment,” “aggregation inhibitor,” and “aggregation dissolving agent” are as described above.
일 구체예에 있어서, 상기 응집 억제제는 제2 아밀로이드 단백질과 동시에 또는 순차적으로 처리되는 것일 수 있다. In one embodiment, the aggregation inhibitor may be treated simultaneously or sequentially with the second amyloid protein.
일 구체예에 있어서, 상기 응집 용해제는 제1 아밀로이드 단백질 및 제2 아밀로이드 단백질을 응집시킨 후, 처리되는 것일 수 있다.In one embodiment, the aggregation dissolving agent may be used after aggregating the first amyloid protein and the second amyloid protein.
본 발명은 아밀로이드 단백질 및 이의 단편을 이용하여, 아밀로이드 단백질 응집 억제제 또는 응집 용해제의 작용부위를 검색하는 방법, 예를 들어 퇴행성 뇌질환을 갖고 있거나 또는 퇴행성 뇌질환이 발생할 위험이 있는 대상체에서 퇴행성 뇌질환을 치료하거나 예방하기 위해 아밀로이드 단백질 응집 억제제 또는 응집 용해제의 작용부위(구체적으로, 작용 도메인)를 검색하는 방법을 제공한다.The present invention provides a method for searching the action site of an amyloid protein aggregation inhibitor or aggregation dissolving agent using amyloid protein and fragments thereof, for example, in subjects with degenerative brain disease or at risk of developing degenerative brain disease, Provides a method for searching the action site (specifically, the action domain) of an amyloid protein aggregation inhibitor or aggregation dissolving agent to treat or prevent.
다른 양상은 (1) 복수 개의 제1 아밀로이드 단백질 단편이 부착된 플레이트를 제공하는 단계; (2-1) 단백질 응집 억제제 후보물질과 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시키는 단계; 또는 (2-2) 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시킨 후 단백질 응집 용해제 후보물질을 처리하는 단계; (3-1) 상기 (2-1) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하는 단계; 또는 (3-2) 상기 (2-2) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하는 단계를 포함하는 아밀로이드 단백질 응집 억제제 후보물질 또는 응집의 용해제 후보물질이 작용하는 부위를 스크리닝하는 방법을 제공한다.Another aspect includes (1) providing a plate to which a plurality of first amyloid protein fragments are attached; (2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or (2-2) contacting the plate with a second amyloid protein bound to an indicator material and then treating the candidate material as a protein aggregation dissolving agent; (3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1); or (3-2) screening the site where the amyloid protein aggregation inhibitor candidate or aggregation dissolving agent candidate acts, including the step of measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2). Provides a way to do this.
상기 제1 아밀로이드 단편 및 제2 아밀로이드 단백질간 응집 저해 또는 용해 정도를 제1 아밀로이드 단백질 단편별로 정렬하는 단계를 추가적으로 포함할 수 있다. The method may additionally include sorting the degree of inhibition or dissolution of aggregation between the first amyloid fragment and the second amyloid protein by the first amyloid protein fragment.
일 구체예에 있어서, 상기 (2-1) 또는 (2-2) 단계는 제1 및 제2 아밀로이드 단백질의 응집을 유도하는 단계를 더 포함하는 것일 수 있다. In one embodiment, step (2-1) or (2-2) may further include inducing aggregation of the first and second amyloid proteins.
일 구체예에 있어서, 상기 아밀로이드 단백질은 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인인 것일 수 있다.In one embodiment, the amyloid protein may be amyloid beta, tau protein, or alpha-synuclein.
일 구체예에 있어서, 상기 단백질 단편은 아밀로이드 단백질 내의 연속되는 6내지 15mer이고, 상기 복수 개의 단백질 단편은 연속되는 2개 이상의 중첩되는 아미노산 잔기를 포함하는 것일 수 있다. 응집률을 측정하는 단계는 지표물질의 세기를 분석하여 단백질 단편별로 비교하여 분석한 것일 수 있다. In one embodiment, the protein fragment is a contiguous 6 to 15 mer within an amyloid protein, and the plurality of protein fragments may include two or more overlapping contiguous amino acid residues. The step of measuring the aggregation rate may be done by analyzing the intensity of the indicator material and comparing it for each protein fragment.
일 구체예에 있어서, 상기 방법은 응집 저해 또는 응집 용해 정도가 높게 측정되는 단백질 단편을 후보물질의 작용부위로 판정하는 단계를 더 포함하는 것일 수 있다. 구체적으로, 응집 저해 또는 응집 용해 정도가 높게 측정되는 하나의 단백질 단편을 작용부위로 판정하거나 또는 응집 저해 또는 응집 용해 정도가 높게 측정되는 복수 개의 단백질 단편의 서로 중첩되는 아미노산 잔기를 작용부위로 판정하는 단계를 더 포함하는 것일 수 있다. 보다 구체적으로, 단백질 단편을 작용부위로 판정하는 단계는 실시예에 나타낸 방법을 통해 단백질 단편별로 응집 저해율(%) 또는 응집 용해율(%)을 비교해보았을 때, 상위 값을 나타내는 단백질 단편을 선택하는 단계를 포함하는 것일 수 있다.In one embodiment, the method may further include determining a protein fragment for which a high degree of aggregation inhibition or aggregation dissolution is measured as the site of action of the candidate substance. Specifically, one protein fragment for which a high degree of aggregation inhibition or aggregation dissolution is measured is determined as the action site, or an overlapping amino acid residue of a plurality of protein fragments for which aggregation inhibition or aggregation dissolution is measured to be high is determined as the action site. It may include more steps. More specifically, the step of determining a protein fragment as an action site is selecting a protein fragment that shows a higher value when comparing the aggregation inhibition rate (%) or aggregation dissolution rate (%) for each protein fragment through the method shown in the examples. It may include.
일 구체에 있어서, 상기 작용부위는 아밀로이드 단백질 내 2개 이상 존재하는 것일 수 있다.In one embodiment, the action site may be present in two or more amyloid proteins.
상기 “자가조립 단백질”, “단백질 단편” “응집 억제제”및 “응집 용해제”는 상기한 바와 같다. The “self-assembling protein,” “protein fragment,” “aggregation inhibitor,” and “aggregation dissolving agent” are the same as described above.
일 구체예에 있어서, 상기 아밀로이드 단백질은 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인(α-cynuclein)인 것일 수 있다.In one embodiment, the amyloid protein may be amyloid beta, tau protein, or alpha-synuclein.
일 구체예에 있어서, 상기 단백질 단편은 6 내지 15mer인 것일 수 있다. 구체적으로, 상기 단백질 단편은 6mer인 것일 수 있다.In one embodiment, the protein fragment may be 6 to 15 mer. Specifically, the protein fragment may be a 6mer.
일 구체예에 있어서, 상기 응집 억제제 또는 응집 억제제 후보물질은 제2 아밀로이드 단백질과 동시에 또는 순차적으로 처리되는 것일 수 있다. In one embodiment, the aggregation inhibitor or aggregation inhibitor candidate may be treated simultaneously or sequentially with the second amyloid protein.
일 구체예에 있어서, 상기 응집 용해제 또는 응집 용해제 후보물질은 제1 아밀로이드 단백질 및 제2 아밀로이드 단백질을 응집시킨 후, 처리되는 것일 수 있다.In one embodiment, the aggregation-dissolving agent or aggregation-dissolving agent candidate may be treated after aggregating the first amyloid protein and the second amyloid protein.
일 구체예에 있어서, 상기 아밀로이드 단백질 응집 억제제 후보물질 또는 응집의 용해제 후보물질이 작용하는 부위를 스크리닝하는 방법은 Aβ 응집 저해제 또는 용해제의 타겟 사이트(target site)를 예측하고 분석하는데 사용되는 분자 도킹 시뮬레이션(molecular docking simulation) 연구 결과들을 검증에 사용될 수 있을 뿐만 아니라, 합리적 디자인(rational design) 방법을 활용하여 Aβ의 서열에 특이적으로 응집을 억제하거나 풀어주는 응집 억제제제 또는 용해제 개발에 사용될 수 있다.In one embodiment, the method of screening the site where the amyloid protein aggregation inhibitor candidate or aggregation dissolving agent candidate acts is a molecular docking simulation used to predict and analyze the target site of the Aβ aggregation inhibitor or dissolving agent. (molecular docking simulation) can not only be used to verify research results, but can also be used to develop aggregation inhibitors or solubilizers that specifically inhibit or release aggregation in the Aβ sequence using a rational design method.
일 양상에 따른 아밀로이드 단백질 응집 억제 또는 용해 후보물질 스크리닝 방법에 의하면 기존의 알려진 아밀로이드 단백질 응집 저해제 또는 용해제 간의 효능을 비교할 수 있을뿐 아니라, 아밀로이드 단백질 응집 저해제 또는 용해제의 타겟 도메인들을 분석하고, 더 나아가 기전 규명까지 밝힐 수 있어 아밀로이드 단백질, 특히 Aβ의 특정 단백질 서열의 응집을 풀어주는 저해제 또는 용해제 개발에 유용할 수 있다.According to an aspect of the screening method for candidates for inhibiting or dissolving amyloid protein aggregation, not only can the efficacy of existing known amyloid protein aggregation inhibitors or solubilizers be compared, but also the target domains of amyloid protein aggregation inhibitors or solubilizers are analyzed, and further the mechanism is determined. Since it can reveal the identity of amyloid proteins, especially Aβ, it can be useful in developing inhibitors or solubilizers that dissolve the aggregation of specific protein sequences.
도 1은 C-말단에 시스테인이 부착된 아밀로이드 베타 및 아밀로이드 베타 단편의 제조방법을 설명하기 위한 모식도이다.Figure 1 is a schematic diagram illustrating a method for producing amyloid beta and amyloid beta fragments with cysteine attached to the C-terminus.
도 2는 C-말단에 시스테인이 부착된 아밀로이드 육량체(hexamer) 단편의 종류를 나타내기 위한 모식도이다.Figure 2 is a schematic diagram showing the types of amyloid hexamer fragments with cysteine attached to the C-terminus.
도 3은 아밀로이드 베타1-6 내지 아밀로이드 베타10-15의 육량체 단편을 역상-HPLC로 분석한 결과를 나타낸 그래프이다.Figure 3 is a graph showing the results of analysis of hexameric fragments of amyloid beta 1-6 to amyloid beta 10-15 by reverse phase-HPLC.
도 4는 아밀로이드 베타11-16 내지 아밀로이드 베타 20-25의 육량체 단편을 역상-HPLC로 분석한 결과를 나타낸 그래프이다.Figure 4 is a graph showing the results of analysis of hexameric fragments of amyloid beta 11-16 to amyloid beta 20-25 by reverse phase-HPLC.
도 5는 아밀로이드 베타21-26 내지 아밀로이드 베타 30-35의 육량체 단편을 역상-HPLC로 분석한 결과를 나타낸 그래프이다.Figure 5 is a graph showing the results of analysis of hexameric fragments of amyloid beta 21-26 to amyloid beta 30-35 by reverse phase-HPLC.
도 6은 아밀로이드 베타31-36 내지 아밀로이드 베타 37-42의 육량체 단편 및 아밀로이드 베타1-42을 역상-HPLC로 분석한 결과를 나타낸 그래프이다.Figure 6 is a graph showing the results of reverse phase-HPLC analysis of hexameric fragments of amyloid beta 31-36 to amyloid beta 37-42 and amyloid beta 1-42 .
도7는 A3 및 MAP의 최적의 조건(펩타이드 농도, 고정시간, 온도, pH 및 형광 표지의 농도)을 확인한 그래프이다.Figure 7 is a graph confirming the optimal conditions (peptide concentration, fixation time, temperature, pH, and fluorescent label concentration) of A3 and MAP.
도 8은 A3 및 MAP 펩타이드의 구조적, 형태적, 응집적 변화를 확인한 결과이다. Figure 8 shows the results of confirming structural, morphological, and cohesive changes in A3 and MAP peptides.
도 9은 아밀로이드 베타 응집 저해제(inhibitor)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 9 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
도 10는 아밀로이드 베타 응집 용해제(dissociator)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 10 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
도 11은 A3를 이용하여 아밀로이드 베타 응집 저해제(Curcumin 및 Scyllol-inositol)의 응집 저해효과를 확인한 그래프이다. Figure 11 is a graph confirming the aggregation inhibition effect of amyloid beta aggregation inhibitors (Curcumin and Scyllol-inositol) using A3.
도 12는 A3를 이용하여 아밀로이드 베타 응집 용해제(Necrostantin-1 및 Sunitinib)의 응집 용해효과를 확인한 그래프이다. Figure 12 is a graph confirming the aggregation-dissolving effect of amyloid beta aggregation-dissolving agents (Necrostantin-1 and Sunitinib) using A3.
도 13는 아밀로이드 베타 응집 저해제(inhibitor)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 13 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
도 14은 아밀로이드 베타 응집 용해제(dissociator)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 14 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
도 15은 MAP을 이용하여 아밀로이드 베타 응집 저해제(Curcumin)의 응집 용해 작용 부위를 확인한 그래프이다.Figure 15 is a graph confirming the aggregation-dissolving action site of the amyloid beta aggregation inhibitor (Curcumin) using MAP.
도 16은 MAP을 이용하여 아밀로이드 베타 응집 저해제(Scyllol-inositol)의 응집 저해 작용 부위를 확인한 그래프이다.Figure 16 is a graph confirming the aggregation inhibition site of amyloid beta aggregation inhibitor (Scyllol-inositol) using MAP.
도 17는 MAP을 이용하여 아밀로이드 베타 응집 용해제(Necrostantin-1)의 응집 용해 작용 부위를 확인한 그래프이다.Figure 17 is a graph confirming the aggregation-dissolving action site of amyloid beta aggregation-dissolving agent (Necrostantin-1) using MAP.
도 18은 MAP을 이용하여 아밀로이드 베타 응집 용해제(Sunitinib)의 응집 저해 작용 부위를 확인한 그래프이다. Figure 18 is a graph confirming the aggregation inhibitory action site of amyloid beta aggregation dissolving agent (Sunitinib) using MAP.
도 19는 A3 제조방법을 나타낸 모식도이다.Figure 19 is a schematic diagram showing the A3 manufacturing method.
도 20은 MAP 제조방법을 나타낸 모식도이다.Figure 20 is a schematic diagram showing the MAP manufacturing method.
이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예를 제시한다. 그러나 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 하기 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다. 실시예들은 다양한 변환을 가할 수 있는 바, 실시예들은 이하에서 개시되는 실시예들에 한정되는 것이 아니라 다양한 형태로 구현될 수 있다.Below, preferred embodiments are presented to aid understanding of the present invention. However, the following examples are provided only for easier understanding of the present invention, and the content of the present invention is not limited by the following examples. The embodiments may be subject to various changes, and the embodiments are not limited to the embodiments disclosed below and may be implemented in various forms.
실시예 1. A3 및 MAP 제작Example 1. Production of A3 and MAP
1.1.1.1. C-말단 시스테인이 결합된 아밀로이드 베타 합성C-terminal cysteine-conjugated amyloid beta synthesis
A3(Aβ aggregation assay) 및 MAP(mapping amyloid plate) 제작을 위해서 37개의 C-말단 시스테인이 부착된 Aβ1- 42 육량체 단편과 C-말단 시스테인이 부착된 전장 Aβ1-42을 마이크로파를 이용한 9-플루오렌닐메틸옥시카르보닐 고체상 펩타이드 합성 방법을 통해 합성하였고, 이를 도 2에 나타내었다.For A3 (Aβ aggregation assay) and MAP (mapping amyloid plate) production, Aβ 1-42 hexamer fragment with 37 C-terminal cysteines attached and full-length Aβ 1-42 with C-terminal cysteine attached were separated using microwaves. -Fluorenylmethyloxycarbonyl was synthesized through a solid phase peptide synthesis method, and is shown in Figure 2.
구체적으로, C-말단의 첫번째 시스테인은 대칭 무수화물 활성(symmetric anhydride activation) 방법으로 붙였다. 2.2 mmol의 Fmoc-Cys(trt)-OH, 1.0 mmol의 DIC, 그리고 51 mg의 DMAP을 2 mL의 DMF/DCM (1:1, v/v) 용액에 녹인 뒤 DMF에 부풀어진 0.25 mmol의 Wang resin LS과 함께 1.5 시간 동안 반응시켰다. 남은 단백질 서열들은 자동화 펩타이드 합성기(automated peptide synthesizer)를 이용하여 순차적으로 커플링 하였다. 합성이 끝난 뒤 절단 칵테일(92.5:2.5:2.5:2.5 TFA/deionized water/TIS/DODT, v/v/v/v)을 넣고 4 시간 동안 반응시켰다. TFA는 회전증발농축기를 사용하여 증발시킨 후 냉각시킨 무수 에테르를 넣고 15 분 동안 원심 분리시켜 정제되지 않은 형태의 펩타이드를 만들었고, 이는 도 1에 나타내었다. Specifically, the first cysteine at the C-terminus was attached using symmetric anhydride activation. 2.2 mmol of Fmoc-Cys(trt)-OH, 1.0 mmol of DIC, and 51 mg of DMAP were dissolved in 2 mL of DMF/DCM (1:1, v/v) solution, and then 0.25 mmol of Wang's solution swollen in DMF. It was reacted with resin LS for 1.5 hours. The remaining protein sequences were sequentially coupled using an automated peptide synthesizer. After synthesis, cleavage cocktail (92.5:2.5:2.5:2.5 TFA/deionized water/TIS/DODT, v/v/v/v) was added and reacted for 4 hours. TFA was evaporated using a rotary evaporator, cooled anhydrous ether was added, and centrifuged for 15 minutes to produce an unrefined peptide, which is shown in Figure 1.
37개의 C-말단 시스테인이 부착된 Aβ1- 42 육량체 단편을 역상-고성능 액체 크로마토그래피(reversed-HPLC) 방법을 통해 정제하고 분석하였고, 이를 도 3 내지 도 6에 나타내었다.The Aβ 1-42 hexamer fragment with 37 C-terminal cysteines attached was purified and analyzed using reversed-HPLC, and is shown in Figures 3 to 6.
도 1은 C-말단에 시스테인이 부착된 아밀로이드 베타 및 아밀로이드 베타 단편의 제조방법을 설명하기 위한 모식도이다.Figure 1 is a schematic diagram illustrating a method for producing amyloid beta and amyloid beta fragments with cysteine attached to the C-terminus.
도 2는 C-말단에 시스테인이 부착된 아밀로이드 육량체(hexamer) 단편의 종류를 나타내기 위한 모식도이다.Figure 2 is a schematic diagram showing the types of amyloid hexamer fragments with cysteine attached to the C-terminus.
도 3 내지 도 6은 아밀로이드 베타 육량체 단편을 역상-HPLC로 분석한 결과를 나타낸 그래프이다. Figures 3 to 6 are graphs showing the results of analysis of amyloid beta hexamer fragments by reversed phase-HPLC.
1.2.1.2. A3의 제작Production of A3
A3를 제작하기 위해, 말레이미드가 부착된 플레이트에 200μL의 세척 완충액(0.1 M Na3PO4, 0.15 M NaCl, 0.05% Tween 20, pH 7.2)를 넣고 세척을 진행하였다. 전장 Aβ1-42-Cys 용액을 결합 완충액(0.1 M Na3PO4, 0.15 M NaCl, 10 mM EDTA)에 혼합(50μg/mL, 5% DMSO)하였다. 100μL의 펩타이드 수용액(10μM)을 각 웰(peptide well)들에 넣고 24℃에서 24 시간동안 인큐베이션하였다. 200μL의 세척 완충액을 넣고 세척을 진행하였다. 시스테인 용액을 결합 완충액과 혼합(10μg/mL)하였다. 200 μL의 상기 결합 완충액과 혼합된 시스테인 용액(10μg/mL)을 각 웰(blank well)넣고 24℃에서 1 시간 동안 인큐베이션하였다. 200μL의 세척 완충액을 넣고 세척을 진행하였고, 이를 도 19에 나타내었다.To prepare A3, 200 μL of washing buffer (0.1 M Na 3 PO 4 , 0.15 M NaCl, 0.05% Tween 20, pH 7.2) was added to the maleimide-attached plate, and washing was performed. The full-length Aβ 1-42 -Cys solution was mixed (50 μg/mL, 5% DMSO) in binding buffer (0.1 M Na 3 PO 4 , 0.15 M NaCl, 10 mM EDTA). 100 μL of aqueous peptide solution (10 μM) was added to each well (peptide well) and incubated at 24°C for 24 hours. 200 μL of washing buffer was added and washing was performed. The cysteine solution was mixed with binding buffer (10 μg/mL). 200 μL of the cysteine solution (10 μg/mL) mixed with the binding buffer was added to each well (blank well) and incubated at 24°C for 1 hour. Washing was performed by adding 200 μL of washing buffer, which is shown in Figure 19.
도 19는 A3 제조방법을 나타낸 모식도이다.Figure 19 is a schematic diagram showing the A3 manufacturing method.
1.3. MAP의 제작1.3. Creation of MAP
MAP을 제작하기 위해, 말레이미드가 부착된 플레이트에 200μL의 세척 완충액(0.1 M Na3PO4, 0.15 M NaCl, 0.05% Tween-20, pH 7.2)을 넣고 세척을 진행하였다. 상기 1.1에서 제조한 37개의 각각의 Aβ1-42 단편-Cys 용액을 결합 완충액(0.1 M Na3PO4, 0.15 M NaCl, 10 mM EDTA)에 혼합(50μg/mL, 5% DMSO)하였고, 전장 Aβ1-42-Cys 용액과 결합 완충액을 혼합(50μg/mL, 5% DMSO)하였다. 100μL의 펩타이드 수용액(10μM)을 각 웰(peptide well)들에 넣고 24℃에서 24 시간동안 인큐베이션하였다. 200μL의 세척 완충액를 넣고 세척을 진행하였다. 시스테인 용액을 결합 완충액과 혼합(10μg/mL)하였다. 200μL의 상기 결합 완충액과 혼합된 시스테인 용액(10μg/mL)을 각 웰(blank well)넣고 24℃에서 1 시간동안 인큐베이션하였다. 200μL의 세척 완충액을 넣고 세척을 진행하였고 이를 도 20에 나타내었다..To prepare MAP, 200 μL of washing buffer (0.1 M Na 3 PO 4 , 0.15 M NaCl, 0.05% Tween-20, pH 7.2) was added to the maleimide-attached plate and washed. Each of the 37 Aβ 1-42 fragment-Cys solutions prepared in 1.1 above was mixed (50 μg/mL, 5% DMSO) in binding buffer (0.1 M Na 3 PO 4 , 0.15 M NaCl, 10 mM EDTA), and the full length Aβ 1-42 -Cys solution and binding buffer were mixed (50 μg/mL, 5% DMSO). 100 μL of aqueous peptide solution (10 μM) was added to each well (peptide well) and incubated at 24°C for 24 hours. 200 μL of washing buffer was added and washing was performed. The cysteine solution was mixed with binding buffer (10 μg/mL). 200 μL of the cysteine solution (10 μg/mL) mixed with the binding buffer was added to each well (blank well) and incubated at 24°C for 1 hour. 200 μL of washing buffer was added and washing was performed, which is shown in Figure 20.
도 20은 MAP 제조방법을 나타낸 모식도이다.Figure 20 is a schematic diagram showing the MAP manufacturing method.
실험예 1. A3/MAP plate의 최적화 조건 Experimental Example 1. Optimization conditions of A3/MAP plate
1.1.1.1. 응집 억제제 또는 응집 용해제 스크리닝을 위한 최적 조건 확립Establishing optimal conditions for screening aggregation inhibitors or aggregation dissolving agents
A3/MAP의 제작 및 활용의 최적화 조건을 찾기 위하여 펩타이드 고정 농도, 펩타이드 고정 시간, 펩타이드 고정 온도, 펩타이드 고정 pH, 및 fluor-Aβ1-42의 pH, 및 온도 실험을 진행하였다.To find the optimal conditions for the production and use of A3/MAP, experiments were conducted on peptide fixation concentration, peptide fixation time, peptide fixation temperature, peptide fixation pH, and the pH and temperature of fluor-Aβ 1-42 .
모든 실험에는 Aβ1-42 펩타이드를 감지하는 anti-Aβ monoclonal 6E10 항체를 이용하여 플레이트에 있는 Aβ1-42 펩타이드의 양을 간접적으로 비교, 분석하였다. 구체적으로, 실시예 1의 정제된 펩타이드를 플레이트에 고정시키기 위한 최적의 펩타이드 농도를 위해 8가지 농도 조건(0, 0.00001, 0.0001, 0.001, 0,01, 0.1, 1, 10 μM)에서 실험을 진행하였고, 정제된 펩타이드를 플레이트에 고정시키기 위한 최적의 시간을 위해 10가지 시간 조건(0, 4, 8, 12, 16, 20, 24, 28, 32, 36 h)에서 진행하였으며, 정제된 펩타이드를 플레이트에 고정시키기 위한 최적의 온도를 위해 6가지 온도 조건(0, 4, 24, 34, 54, 74℃)에서 실험을 진행하였고, 정제된 펩타이드를 플레이트에 고정시키기 위한 최적의 온도를 위해 7가지 pH 조건(1.2, 3.2, 5.2, 7.2, 9.2, 11.2, 13.2)에서 실험을 진행하였다. 또한, A3/MAP 플레이트에 형광 물질이 결합된 Aβ1-42(fluor-Aβ1-42) 펩타이드를 처리하여 Aβ 응집을 일으키므로, 플레이트의 웰에서 Aβ 응집이 생성되는 최적의 조건을 찾기 위해, 3가지의 fluor-Aβ1-42 농도 조건(0.1, 1, 10μM)에서 실험을 진행하였고, fluor-Aβ1-42 펩타이드를 10μM의 농도로 24시간 동안 7가지 pH조건(1.2, 3.2, 5.2, 7.2, 9.2, 11.2, 13.2)에서 실험을 진행하였으며, 플레이트의 웰에서 Aβ 응집이 생성되는 최적의 조건을 찾기 위해 fluor-Aβ1-42 펩타이드를 10μM의 농도로 24시간 동안 6가지 온도 조건(0, 4, 24, 34, 54, 74℃)에서 실험을 진행하였다. 그 결과를 도 7에 나타내었다. In all experiments, the amount of Aβ 1-42 peptide on the plate was indirectly compared and analyzed using anti-Aβ monoclonal 6E10 antibody, which detects Aβ 1-42 peptide. Specifically, experiments were conducted under eight concentration conditions (0, 0.00001, 0.0001, 0.001, 0,01, 0.1, 1, 10 μM) to determine the optimal peptide concentration for immobilizing the purified peptide of Example 1 on the plate. To determine the optimal time for immobilizing the purified peptide on the plate, the process was performed under 10 time conditions (0, 4, 8, 12, 16, 20, 24, 28, 32, 36 h). Experiments were conducted under 6 temperature conditions (0, 4, 24, 34, 54, 74°C) to determine the optimal temperature for immobilizing the purified peptides on the plate, and 7 conditions were used to determine the optimal temperature for immobilizing the purified peptides on the plate. The experiment was conducted under pH conditions (1.2, 3.2, 5.2, 7.2, 9.2, 11.2, 13.2). In addition, since A3/MAP plates are treated with Aβ1-42 (fluor-Aβ1-42) peptide bound to a fluorescent substance to cause Aβ aggregation, in order to find the optimal conditions for Aβ aggregation to occur in the wells of the plate, three methods were used. The experiment was conducted under fluor-Aβ 1-42 concentration conditions (0.1, 1, 10 μM), and fluor-Aβ 1-42 peptide was incubated at a concentration of 10 μM for 24 hours under seven pH conditions (1.2, 3.2, 5.2, 7.2, 9.2, 11.2, 13.2), and to find the optimal conditions for generating Aβ aggregation in the wells of the plate, fluor-Aβ 1-42 peptide was used at a concentration of 10 μM for 24 hours under six temperature conditions (0, 4 , 24, 34, 54, 74°C). The results are shown in Figure 7.
도7은 A3 및 MAP의 최적의 조건(펩타이드 농도, 고정시간, 온도, pH 및 형광 표지의 농도)을 확인한 그래프이다.Figure 7 is a graph confirming the optimal conditions (peptide concentration, fixation time, temperature, pH, and fluorescent label concentration) of A3 and MAP.
도 7에 나타낸 바와 같이, 정제된 펩타이드를 플레이트에 고정하기 위한 펩타이드 농도 조건과 관련하여, Aβ 펩타이드가 농도 의존적으로 플레이트에 결합되는 것을 확인하였다. 하지만 농도가 1μM 이상에서 더 이상 농도의존적으로 증가하지 않는 것을 관찰하였다. 이를 통해 정제된 펩타이드를 플레이트에 고정시키기 위해서는 1μM 농도의 정제된 펩타이드가 최적 농도인 점을 확인하였다(도 7A). 정제된 펩타이드를 플레이트에 고정하기 위한 펩타이드 시간 조건과 관련하여, 각 펩타이드는 1μM 농도에서, 6E10 항체를 통해 23-36시간동안 모든 Aβ 펩타이드를 효과적으로 플레이트에 고정됨을 확인하였다. 다만, 25시간 인큐베이션에서 더 이상 추가로 Aβ 펩타이드가 고정되지 않는 것을 확인하였다(도 7B). 정제된 펩타이드를 플레이트에 고정하기 위한 펩타이드 온도 조건과 관련하여, 각 펩타이드는 1μM 농도로 플레이트에 처리하고, 6E10 항체로 확인한 결과, 온도가 35℃보다 높아지면 펩타이드가 플레이트에 고정되는 효율이 현저이 낮아지는 것을 확인하였다. 이를 통해, 고정 온도가 28-32℃범위에서 가장 효과적인 것을 확인하였다(도 7C). 정제된 펩타이드를 플레이트에 고정하기 위한 펩타이드 pH 조건과 관련하여, 각 펩타이드는 1μM 농도로 플레이트에 처리하고 6E10 항체로 확인한 결과, 중성의 pH 범위에서 펩타이드가 플레이트에 효과적으로 부착되었고, 특히, pH 6.5-7.2일 때, 펩타이드가 플레이트에 가장 효과적으로 붙는 것을 확인하였다(도 7D). As shown in Figure 7, with respect to the peptide concentration conditions for immobilizing the purified peptide on the plate, it was confirmed that the Aβ peptide was bound to the plate in a concentration-dependent manner. However, it was observed that the concentration no longer increased in a concentration-dependent manner above 1 μM. Through this, it was confirmed that 1 μM concentration of purified peptide was the optimal concentration for immobilizing the purified peptide on the plate (Figure 7A). Regarding the peptide time conditions for immobilizing the purified peptides on the plate, it was confirmed that each peptide was effectively immobilized on the plate for 23-36 hours using the 6E10 antibody at a concentration of 1 μM. However, it was confirmed that no additional Aβ peptide was fixed after 25 hours of incubation (Figure 7B). Regarding the peptide temperature conditions for immobilizing the purified peptides on the plate, each peptide was treated on the plate at a concentration of 1 μM, and as confirmed with the 6E10 antibody, the efficiency of immobilizing the peptides on the plate was significantly lower when the temperature was higher than 35°C. It was confirmed that he lost. Through this, it was confirmed that the fixation temperature was most effective in the range of 28-32°C (Figure 7C). Regarding the peptide pH conditions for immobilizing the purified peptides on the plate, each peptide was treated on the plate at a concentration of 1 μM and confirmed with the 6E10 antibody, and the results showed that the peptide was effectively attached to the plate in the neutral pH range, especially at pH 6.5- At 7.2, it was confirmed that the peptide was most effectively attached to the plate (Figure 7D).
플레이트에서 고정된 Aβ1-42와 fluor-Aβ1-42 간의 응집이 생성되는 최적의 조건 관련하여, fluor-Aβ1-42의 농도 조건과 관련하여, Blank well과 형광의 세기를 비교했을 때 0.1과 1μM의 농도에서는 고정된 Aβ1-42와 fluor-Aβ1-42 간의 응집이 잘 형성되지 않았지만, 10μM 농도에서는 유의적으로 응집이 되며 형광세기가 검출되는 것을 확인하였다(도 7G). 또한, fluor-Aβ1-42을10μM 농도로 24시간 동안 다양한 pH 및 다양한 온도 조건에서 고정된 Aβ1-42와 응집을 유도한 결과, pH가 중성의 범위 특히, pH 7-8일 때, 가장 효과적으로 고정된 Aβ1-42와 fluor-Aβ1-42 간의 응집을 확인하였다(도 7E). 또한, 플레이트에서 고정된 Aβ1-42와 fluor-Aβ1-42 간의 응집이 생성되는 최적의 조건 관련하여, 온도가 28-38℃, 특히 30-36℃에서 가장 효과적으로 고정된 Aβ1-42와 fluor-Aβ1-42 간의 응집을 확인하였다. Regarding the optimal conditions for generating aggregation between fixed Aβ 1-42 and fluor-Aβ 1-42 on the plate, regarding the concentration condition of fluor-Aβ 1-42 , 0.1 when comparing the intensity of fluorescence with the blank well. At a concentration of 1 μM, aggregation between fixed Aβ 1-42 and fluor-Aβ 1-42 was not well formed, but at a concentration of 10 μM, significant aggregation occurred and fluorescence intensity was detected (Figure 7G). In addition, as a result of inducing aggregation of fluor-Aβ 1-42 with fixed Aβ 1-42 at a concentration of 10 μM for 24 hours under various pH and temperature conditions, the result showed that the aggregation was most effective when the pH was in the neutral range, especially when pH was 7-8. Aggregation between effectively immobilized Aβ 1-42 and fluor-Aβ 1-42 was confirmed (Figure 7E). In addition, regarding the optimal conditions for generating aggregation between immobilized Aβ 1-42 and fluor-Aβ 1-42 on a plate, the temperature of immobilized Aβ 1-42 and fluor-Aβ 1-42 was most effective at a temperature of 28-38°C, especially 30-36°C. Aggregation between fluor-Aβ 1-42 was confirmed.
이상의 결과를 통해, 정제된 펩타이드를 플레이트에 고정시키 위한 최적한 조건은 0.5 내지 5μM 농도의 펩타이드를 23-25시간동안 28-32℃, pH 6.5-7.2에서 고정시킨 후, 응집을 유도하는 형광표지 펩타이드도 8 내지 12μM, pH 7-8, 및 30-36℃온도 조건에서 정제된 펩타이드와 가장 효과적으로 응집 유도할 수 있음을 확인하였다.From the above results, the optimal conditions for immobilizing the purified peptide on the plate are to immobilize the peptide at a concentration of 0.5 to 5 μM at 28-32°C and pH 6.5-7.2 for 23-25 hours, and then apply a fluorescent label to induce aggregation. It was confirmed that peptides can most effectively induce aggregation with purified peptides under the conditions of 8 to 12 μM, pH 7-8, and 30-36°C temperature.
1.2. 펩타이드의 구조적/형태적/응집적 변화1.2. Structural/conformational/aggregative changes in peptides
항-Aβ 단일 클론 6E10항체와 항-oligomer 다클론 A11 항체들을 이용하여 플레이트에 고정된 전장 Aβ1-42들이 안정한 단량체 상태를 유지하는지를 확인하였다. 6E10 항체를 통해 전장 Aβ1-42 펩타이드들이 동일한 양으로 안정하게 플레이트에 고정되는지 확인하였고, A11 항체는 웰 안에서 전장 Aβ1-42 펩타이드들의 부착과정 동안 응집되는지 확인하는데 사용되었다. 비교 분석을 하기 위해 1,1,1,3,3,3-헥사플루오로-2-프로파놀(HFIP) 또는 로릴 황산 나트륨(SDS)를 2 시간 동안 처리해서 단량체화시킨 대조군들을 사용하였다.Using anti-Aβ monoclonal 6E10 antibody and anti-oligomer polyclonal A11 antibody, it was confirmed whether the full-length Aβ 1-42 immobilized on the plate maintained a stable monomeric state. The 6E10 antibody was used to confirm whether the full-length Aβ 1-42 peptides were stably immobilized on the plate in equal amounts, and the A11 antibody was used to confirm whether the full-length Aβ 1-42 peptides aggregated during the attachment process in the well. For comparative analysis, control groups monomerized by treatment with 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) or sodium lauryl sulfate (SDS) for 2 hours were used.
또한, 항-Aβ18-23 단일클론 4G8항체와 항-Aβ4-10 다클론 6E10 항체들을 이용하여 플레이트에 고정된 Aβ1-42 단편들 또는 전장 Aβ1-42 펩타이드들이 부착과정동안 구조적/형태적 형질을 유지하는지 확인하였다. 4G8의 에피토프는 Aβ1-42의 Aβ18-23 서열이며 6E10의 에피토프는 Aβ1-42의 Aβ4-10 서열로 알려져 있다. 따라서 이 항체들을 이용해 부착된 Aβ1-42 단편들 또는 전장 Aβ1-42를 타켓하는지 확인하였다.In addition, using anti-Aβ 18-23 monoclonal 4G8 antibody and anti-Aβ 4-10 polyclonal 6E10 antibody, the structural/morphological changes of Aβ 1-42 fragments or full-length Aβ 1-42 peptides immobilized on plates were observed during the attachment process. It was confirmed whether the original traits were maintained. The epitope of 4G8 is known to be the Aβ 18-23 sequence of Aβ 1-42 , and the epitope of 6E10 is known to be the Aβ 4-10 sequence of Aβ 1-42 . Therefore, these antibodies were used to confirm whether they targeted attached Aβ 1-42 fragments or full-length Aβ 1-42 .
추가적으로 10μM 전장 Aβ1-42 펩타이드를 처리하여 부착된 전장Aβ1-42들이 고유의 응집 기전을 유지하는지 확인하였다. 티오플라빈-T(ThT) 형광 물질은 Aβ1-42 응집체의 β시트의 층 간에 삽입되면 형광의 세기가 증가하고 적색-편향되는데, 이러한 ThT의 특성을 이용하여 부착된 전장 Aβ1-42 펩타이드와 추가적으로 넣어준 10μM 전장Aβ1-42 펩타이드가 서로 응집하는지를 확인하였다.Additionally, 10 μM full-length Aβ 1-42 peptide was treated to confirm whether the attached full-length Aβ 1-42 maintained its unique aggregation mechanism. When thioflavin-T (ThT) fluorescent substance is inserted between the layers of the β-sheet of Aβ 1-42 aggregates, the fluorescence intensity increases and is red-biased. Using these properties of ThT, the attached full-length Aβ 1-42 peptide It was confirmed whether the additionally added 10 μM full-length Aβ 1-42 peptide coagulate with each other.
도 8은 A3 및 MAP 펩타이드의 구조적, 형태적, 응집적 변화를 확인한 결과이다. Figure 8 shows the results of confirming structural, morphological, and cohesive changes in A3 and MAP peptides.
도 8에 나타낸 바와 같이, 6E10 항체 및 A11 항체를 이용하였을 때 플레이트 안에 응집된 전장 Aβ1-42 펩타이드들을 발견하지 못하였고, 이는 대조군 그룹들과 비교하였을 때도 차이가 없는 것을 확인하였다. 4G8 항체가 전장 Aβ1-42 그리고 Aβ18-23를 타겟하는 것을 확인하였다. 6E10 항체 역시도 전장 Aβ1-42, Aβ4-9, 그리고 Aβ5-10 펩타이드들을 타켓하는 것을 확인하였다. ThT 형광 물질을 통해, 인큐베이션 시간 동안 점차적으로 응집이 관찰되었으며, 24 시간 이후부터 안정한 β시트 형태를 갖는 아밀로이드 피브릴(fibril)의 형성을 확인하였다.As shown in Figure 8, when using the 6E10 antibody and A11 antibody, full-length Aβ 1-42 peptides were not found aggregated in the plate, and there was no difference when compared with the control group. It was confirmed that the 4G8 antibody targets full-length Aβ 1-42 and Aβ 18-23 . The 6E10 antibody was also confirmed to target full-length Aβ 1-42 , Aβ 4-9 , and Aβ 5-10 peptides. Through ThT fluorescence, aggregation was gradually observed during the incubation time, and the formation of amyloid fibrils with a stable β-sheet shape was confirmed after 24 hours.
이상의 결과를 통해, 플레이트에 부착된 모든 전장 Aβ1-42 펩타이드들은 안정한 단량체 형태를 유지하는 것을 확인하였고, 부착과정 이후에도 단편들과 전장 Aβ1-42 펩타이드들이 고유의 구조적/형태적 형질을 유지하는 것을 확인하였다. Through the above results, it was confirmed that all full-length Aβ 1-42 peptides attached to the plate maintained a stable monomeric form, and that fragments and full-length Aβ 1-42 peptides maintained their unique structural/morphological characteristics even after the attachment process. confirmed.
실험예 2. 아밀로이드 베타 응집 억제제의 스크리닝 Experimental Example 2. Screening of amyloid beta aggregation inhibitors
2.1. 아밀로이드 베타 응집 저해제(inhibitor)의 스크리닝2.1. Screening of amyloid beta aggregation inhibitors
A3에 형광 물질인 Flamma 552와 Aβ1-42를 컨쥬게이션한 fluor-Aβ1-42(10μM) 와 Aβ 응집 저해제를 넣고 24 시간 동안 상온에서 반응시켰다((+)inhibitor/(+)fluor-Aβ1-42 A3). 비교 분석을 위해 대조군 웰에는 fluor-Aβ1-42만 처리하였다((-)inhibitor/(+)fluor-Aβ1-42 A3). 대조군과 응집 저해제를 넣은 웰들 간 Flamma 552 형광 세기를 비교하여 Aβ 응집 저해 물질들을 검색하였고, 이를 도 9및 도 11에 나타내었다.Fluor-Aβ 1-42 (10μM), a conjugate of the fluorescent substance Flamma 552 and Aβ 1-42, and an Aβ aggregation inhibitor were added to A3 and reacted at room temperature for 24 hours ((+)inhibitor/(+)fluor-Aβ 1-42 A3). For comparative analysis, control wells were treated with only fluor-Aβ 1-42 ((-)inhibitor/(+)fluor-Aβ 1-42 A3). Substances inhibiting Aβ aggregation were searched by comparing the fluorescence intensity of Flamma 552 between the control and wells containing the aggregation inhibitor, and these are shown in Figures 9 and 11.
실험 데이터를 정규화하기위해 Zi = (Xi - min(x)) / (max (x) - min (x))* 100의 공식을 이용하였다.To normalize the experimental data, the formula Zi = (Xi - min(x)) / (max (x) - min (x))* 100 was used.
Zi: The normalized value in the datasetZi: The normalized value in the dataset
Xi: The value in the dataset, (+)inhibitor/(+)fluor-Aβ1-42 A3 Xi: The value in the dataset, (+)inhibitor/(+)fluor-Aβ 1-42 A3
min(x): (-)inhibitor/(+)fluor-Aβ1-42 A3min(x): (-)inhibitor/(+)fluor-Aβ 1-42 A3
Max(x): (-)inhibitor/(-)fluor-Aβ1-42 A3 Max(x): (-)inhibitor/(-)fluor-Aβ 1-42 A3
도 9는 아밀로이드 베타 응집 저해제(inhibitor)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 9 is a schematic diagram showing a method for screening candidate substances as amyloid beta aggregation inhibitors.
도 11은 A3를 이용하여 아밀로이드 베타 응집 저해제(Curcumin 및 Scyllol-inositol)의 응집 저해효과를 확인한 그래프이다. Figure 11 is a graph confirming the aggregation inhibition effect of amyloid beta aggregation inhibitors (Curcumin and Scyllol-inositol) using A3.
도 11에 나타낸 바와 같이, A3를 이용한 Aβ 응집 저해제 검색법을 통하여 응집 저해제들을 검색하였다. Curcumin은 49.0%, scyllo-inositol은 24.7% 정도로 Aβ 응집을 저해하는 것을 확인하였다.As shown in Figure 11, aggregation inhibitors were searched through the Aβ aggregation inhibitor search method using A3. Curcumin was confirmed to inhibit Aβ aggregation by 49.0% and scyllo-inositol by 24.7%.
2.2. 아밀로이드 베타 응집 용해제(dissociator)의 스크리닝2.2. Screening of amyloid beta dissociators
A3에 fluor-Aβ1-42(10μM)를 넣고 6 시간 동안 37℃에서 플레이트 안에서 응집시켰다((-)dissociator/(+)fluor-Aβ1-42 A3). 그 후 Aβ 응집 용해제를 넣고 상온에서 24 시간 동안 처리하였다((+)dissociator/(+)fluor-Aβ1-42 A3). 6 시간 동안 응집시킨 후의 Flamma 552 형광 세기와 응집 용해제를 처리한 후의 Flamma 552의 형광 세기가 감소되는 정도를 비교하여 응집을 용해시키는 물질들을 검색하였고, 이를 도 10 및 도 12에 나타내었다.Fluor-Aβ 1-42 (10 μM) was added to A3 and aggregated in the plate at 37°C for 6 hours ((-)dissociator/(+)fluor-Aβ 1-42 A3). Afterwards, Aβ aggregation dissolving agent was added and treated at room temperature for 24 hours ((+)dissociator/(+)fluor-Aβ 1-42 A3). Substances that dissolve aggregation were searched by comparing the fluorescence intensity of Flamma 552 after aggregation for 6 hours and the degree to which the fluorescence intensity of Flamma 552 was reduced after treatment with an aggregation dissolving agent, and these are shown in Figures 10 and 12.
데이터를 정규화하기위해 Zi = (Xi - min(x)) / (max (x) - min (x))* 100의 공식을 이용하였다.To normalize the data, the formula Zi = (Xi - min(x)) / (max (x) - min (x))* 100 was used.
Zi: The normalized value in the datasetZi: The normalized value in the dataset
Xi: The value in the dataset, (+)dissociator/(+)fluor-Aβ1-42 A3 Xi: The value in the dataset, (+)dissociator/(+)fluor-Aβ 1-42 A3
min(x): (-)dissociator/(+)fluor-Aβ1-42 A3min(x): (-)dissociator/(+)fluor-Aβ 1-42 A3
Max(x): (-)dissociator/(-)fluor-Aβ1-42 A3 Max(x): (-)dissociator/(-)fluor-Aβ 1-42 A3
도 10는 아밀로이드 베타 응집 용해제(dissociator)로 후보 물질의 스크리닝 방법을 나타낸 모식도이다. Figure 10 is a schematic diagram showing a method for screening candidate substances as an amyloid beta aggregation dissociator.
도 12는 A3를 이용하여 아밀로이드 베타 응집 용해제(Necrostantin-1 및 Sunitinib)의 응집 용해효과를 확인한 그래프이다. Figure 12 is a graph confirming the aggregation-dissolving effect of amyloid beta aggregation-dissolving agents (Necrostantin-1 and Sunitinib) using A3.
도 12에 나타낸 바와 같이, A3를 이용한 Aβ 응집 용해제 검색법을 통하여 응집 용해제들을 검색하였다. Necrostatin-1은 42.5%, sunitnib은 65.2% 정도로 Aβ 응집을 용해하는 것을 확인하였다.As shown in Figure 12, agglutination dissolving agents were searched through the Aβ aggregation dissolving agent search method using A3. Necrostatin-1 was confirmed to dissolve Aβ aggregates by 42.5% and sunitnib by 65.2%.
이상의 결과를 통해, A3 및 MAP은 Aβ 응집 저해제 또는 용해 후보 물질들을 효과적으로 스크리닝할 수 있는 플랫폼을 제공할 수 있음을 확인하였다. Through the above results, it was confirmed that A3 and MAP can provide a platform for effectively screening Aβ aggregation inhibitors or dissolution candidates.
실험예 3. 아밀로이드 베타 응집 억제제의 활성 부위 매핑Experimental Example 3. Active site mapping of amyloid beta aggregation inhibitor
3.1. 아밀로이드 베타 응집 저해제(inhibitor)의 활성 부위 매핑3.1. Active site mapping of amyloid beta aggregation inhibitor
MAP 플레이트에 형광 물질인 Flamma 552와 Aβ1-42를 컨쥬게이션한 fluor-Aβ1-42(10 μM)와 Aβ 응집 저해제를 넣고 24 시간 동안 상온에서 반응시켰다 ((+)inhibitor/(+)fluor-Aβ1-42 MAP). 비교 분석을 위해 대조군 웰에는 fluor-Aβ1-42만 처리하였다((-)inhibitor/(+)fluor-Aβ1-42 MAP). 대조군과 응집 저해제를 넣은 웰들 간 Flamma 552 형광 세기를 비교하여 Aβ 응집 저해 물질이 어떤 단편들의 응집을 저해시키는지 확인함으로써 물질의 작용 부위를 규명하였고, 이를 도 13에 나타내었다.Fluor-Aβ 1-42 (10 μM), a conjugate of the fluorescent substance Flamma 552 and Aβ 1-42, and an Aβ aggregation inhibitor were added to the MAP plate and reacted at room temperature for 24 hours ((+)inhibitor/(+)fluor -Aβ 1-42 MAP). For comparative analysis, control wells were treated with only fluor-Aβ 1-42 ((-)inhibitor/(+)fluor-Aβ 1-42 MAP). By comparing the fluorescence intensity of Flamma 552 between the control and the wells containing the aggregation inhibitor, it was confirmed which fragments the Aβ aggregation inhibitor inhibits aggregation, thereby identifying the site of action of the substance, which is shown in Figure 13.
실험 데이터를 정규화하기 위해 실험예 2.1과 동일한 방법인 Zi = (Xi - min(x)) / (max (x) - min (x))* 100의 공식을 이용하였다. 또한 응집 저해 정도를 단편별로 분석한 뒤 서로 겹치는 서열이 있는 단편들을 반영한 평균 값을 구하였다. 또한, 그 중 가장 높은 값을 기준으로 정규화하여 응집 저해제가 Aβ의 어느 특정한 서열을 타겟하는지 확인할 수 있는 color-coded heatmap(초록색으로 갈수록 강하게 Aβ 응집 저해)을 제작했다고, 이를 도 15 및 도 16에 나타내었다.To normalize the experimental data, the formula Zi = (Xi - min(x)) / (max (x) - min (x))* 100, the same method as in Experimental Example 2.1, was used. In addition, the degree of aggregation inhibition was analyzed for each fragment, and an average value reflecting fragments with overlapping sequences was calculated. In addition, by normalizing based on the highest value, a color-coded heatmap was created to confirm which specific sequence of Aβ the aggregation inhibitor targets (the stronger the inhibition of Aβ aggregation in green), which is shown in Figures 15 and 16. indicated.
도 15은 MAP을 이용하여 아밀로이드 베타 응집 저해제(Curcumin)의 응집 용해 작용 부위를 확인한 그래프이다.Figure 15 is a graph confirming the aggregation-dissolving action site of the amyloid beta aggregation inhibitor (Curcumin) using MAP.
도 16은 MAP을 이용하여 아밀로이드 베타 응집 저해제(Scyllol-inositol)의 응집 저해 작용 부위를 확인한 그래프이다.Figure 16 is a graph confirming the aggregation inhibition site of amyloid beta aggregation inhibitor (Scyllol-inositol) using MAP.
도 15에 나타낸 바와 같이, MAP를 이용한 Aβ 응집 저해제 규명 방법을 통하여 curcumin이 HQKLVFFA(Aβ14-19, Aβ15-20, Aβ16-21), GAIIGLM (Aβ29-34, Aβ30-35), 및 GGVVIA(Aβ37-42) 도메인들을 특이적으로 타겟하며, 이 부위와 결합함으로써 응집을 저해하는 것을 확인하였다. As shown in Figure 15, through the method of identifying Aβ aggregation inhibitors using MAP, curcumin was identified as HQKLVFFA (Aβ 14-19 , Aβ 15-20 , Aβ 16-21 ), GAIIGLM (Aβ 29-34 , Aβ 30-35 ), and GGVVIA (Aβ 37-42 ) domains, and was confirmed to inhibit aggregation by binding to these regions.
도 16에 나타낸 바와 같이, MAP를 이용한 Aβ 응집 저해제 규명 방법을 통하여 scyllo-inositol이 VHHQKLVFF(Aβ12-17, Aβ13-18, Aβ14-19, Aβ15-20), VGSNKGAIIGL(Aβ24-29, Aβ25-30, Aβ26-31, Aβ27-32, Aβ28-33, Aβ29-34), LMVGGV (Aβ34-39), 및 GGVVIA(Aβ37-42) 도메인들을 특이적으로 타겟하며, 이 부위와 결합함으로써 응집을 저해하는 것을 확인하였다.As shown in Figure 16, through the method of identifying Aβ aggregation inhibitors using MAP, scyllo-inositol was identified as VHHQKLVFF (Aβ 12-17 , Aβ 13-18 , Aβ 14-19 , Aβ 15-20 ), VGSNKGAIIGL (Aβ 24-29 ) , Aβ 25-30 , Aβ 26-31 , Aβ 27-32 , Aβ 28-33 , Aβ 29-34 ), LMVGGV (Aβ 34-39 ), and GGVVIA (Aβ 37-42 ) domains. , it was confirmed that aggregation was inhibited by binding to this site.
3.2. 아밀로이드 베타 응집 용해제(dissociator)의 활성 부위 매핑3.2. Mapping the active site of the amyloid beta dissociator.
MAP에 fluor-Aβ1-42(10 μM)를 넣고 6 시간 동안 37℃에서 플레이트 안에 응집시켰다((-)dissociator/(+)fluor-Aβ1-42 MAP). 그 후 Aβ 응집 용해제를 넣고 24 시간 동안 상온에서 처리하였다((+)dissociator/(+)fluor-Aβ1-42 MAP). 6 시간 동안 응집시킨 후의 Flamma 552 형광 세기와 응집 용해제를 처리 후의 Flamma 552의 형광 세기가 감소되는 정도를 비교하여, Aβ 응집 용해 물질이 어떤 단편들의 응집을 용해시켜주는지 확인함으로써 물질의 작용 부위를 규명하였고, 이를 도 14에 나타내었다.Fluor-Aβ 1-42 (10 μM) was added to MAP and aggregated in the plate at 37°C for 6 hours ((-)dissociator/(+)fluor-Aβ 1-42 MAP). Afterwards, Aβ aggregation dissolving agent was added and treated at room temperature for 24 hours ((+)dissociator/(+)fluor-Aβ 1-42 MAP). By comparing the fluorescence intensity of Flamma 552 after aggregation for 6 hours and the degree to which the fluorescence intensity of Flamma 552 is reduced after treatment with an aggregation dissolving agent, the site of action of the substance is identified by confirming which fragments the Aβ aggregation dissolving agent dissolves. and this is shown in Figure 14.
실험 데이터를 정규화하기 위해 실험예 2.2와 동일한 방법인 Zi = (Xi - min(x)) / (max (x) - min (x))* 100의 공식을 이용하였다. 또한 응집 저해 정도를 단편별로 분석한 뒤 서로 겹치는 서열이 있는 단편들을 반영한 평균 값을 구하였다. 또한, 그 중 가장 높은 값을 기준으로 정규화하여 응집 용해제가 Aβ의 어느 특정한 서열을 타겟하는지 확인할 수 있는 color-coded heatmap(파란색으로 갈수록 강하게 Aβ 응집 용해)을 제작했고, 이를 도 17 및 도 18에 나타내었다.To normalize the experimental data, the same formula as in Experimental Example 2.2, Zi = (Xi - min(x)) / (max (x) - min (x))* 100, was used. In addition, the degree of aggregation inhibition was analyzed for each fragment, and an average value reflecting fragments with overlapping sequences was calculated. In addition, by normalizing based on the highest value, a color-coded heatmap was created to confirm which specific sequence of Aβ the aggregation lytic agent targets (Aβ aggregation dissolution becomes stronger as the color increases in blue), which is shown in Figures 17 and 18. indicated.
도 17은 MAP을 이용하여 아밀로이드 베타 응집 용해제(Necrostantin-1)의 응집 용해 작용 부위를 확인한 그래프이다.Figure 17 is a graph confirming the aggregation-dissolving action site of amyloid beta aggregation-dissolving agent (Necrostantin-1) using MAP.
도 18은 MAP을 이용하여 아밀로이드 베타 응집 용해제(Sunitinib)의 응집 저해 작용 부위를 확인한 그래프이다. Figure 18 is a graph confirming the aggregation inhibitory action site of amyloid beta aggregation dissolving agent (Sunitinib) using MAP.
도 17에 나타낸 바와 같이, MAP를 이용한 Aβ 응집 용해제 규명 방법을 통하여 necrostatin-1이 HHQKLVFF(Aβ13-18, Aβ14-19, Aβ15-20)과 GLMVGGVVIA(Aβ33-38, Aβ34-39, Aβ35-40, Aβ36-41, Aβ37-42) 도메인들을 특이적으로 타겟하며 응집 용해를 일으킨다는 것을 확인할 수 있다. 전장 Aβ1-42와 비교할 때 Aβ36-41 부위를 특이적으로 풀어준다는 것을 확인하였다. As shown in Figure 17, through the method of identifying Aβ aggregation dissolving agents using MAP, necrostatin-1 was identified as HHQKLVFF (Aβ 13-18 , Aβ 14-19 , Aβ 15-20 ) and GLMVGGVVIA (Aβ 33-38 , Aβ 34-39 ). , Aβ 35-40 , Aβ 36-41 , and Aβ 37-42 ) domains and can be confirmed to cause aggregation and dissolution. It was confirmed that the Aβ 36-41 region was specifically released when compared to the full-length Aβ 1-42 .
도 18에 나타낸 바와 같이, MAP를 이용한 Aβ 응집 저해제 규명 방법을 통하여 sunitnib이 AIIGLM(Aβ30-35), 및 LMVGGVVIA(Aβ34-39, Aβ35-40, Aβ36-41, Aβ37-42) 도메인들을 특이적으로 타겟하며 응집 저해를 일으킨다는 것을 확인하였다.As shown in Figure 18, through the method of identifying Aβ aggregation inhibitors using MAP, sunitnib was identified as AIIGLM (Aβ 30-35 ), and LMVGGVVIA (Aβ 34-39 , Aβ 35-40 , Aβ 36-41 , Aβ 37-42 ). It was confirmed that it specifically targets domains and inhibits aggregation.
이상의 결과를 통해, MAP는 Aβ 응집 저해제 또는 용해제의 타겟 도메인들을 분석하고, Aβ의 특정 단백질 서열의 응집을 풀어주는 저해제 또는 용해제 개발에 유용할 수 있다.Based on the above results, MAP can be useful in analyzing the target domains of Aβ aggregation inhibitors or solubilizers and developing inhibitors or solubilizers that unravel the aggregation of specific protein sequences of Aβ.

Claims (24)

  1. (1) 제1 아밀로이드 단백질이 부착된 플레이트를 제공하는 단계;(1) providing a plate to which the first amyloid protein is attached;
    (2-1) 단백질 응집 억제제 후보물질과 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시키는 단계; 또는(2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or
    (2-2) 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시킨 후 단백질 응집 용해제 후보물질을 처리하는 단계; 및,(2-2) contacting the second amyloid protein bound with an indicator substance to the plate and then treating the candidate substance with a protein aggregation dissolving agent; and,
    (3-1) 상기 (2-1) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하여 무처리 대조군 대비 변화시킨 응집 억제제 후보물질을 선별하는 단계; 또는(3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1) and selecting aggregation inhibitor candidates changed compared to the untreated control group; or
    (3-2) 상기 (2-2) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하여 무처리 대조군 대비 변화시킨 응집 용해제 후보물질을 선별하는 단계를 포함하는 단백질 응집 억제제 또는 응집 용해제를 스크리닝하는 방법.(3-2) A protein aggregation inhibitor or aggregation-dissolving agent comprising the step of measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2) and selecting candidates for aggregation-dissolving agent changed compared to the untreated control group. How to screen.
  2. 청구항 1에 있어서, 상기 (2-1) 또는 (2-2) 단계는 제1 및 제2 아밀로이드 단백질의 응집을 유도하는 단계를 더 포함하는 것인 방법.The method according to claim 1, wherein step (2-1) or (2-2) further comprises the step of inducing aggregation of the first and second amyloid proteins.
  3. 청구항 1에 있어서, 상기 아밀로이드 단백질은 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인(α-cynuclein)인 것인 방법.The method according to claim 1, wherein the amyloid protein is amyloid beta, tau protein, or alpha-synuclein (α-cynuclein).
  4. 청구항 1에 있어서, 상기 제1 및 제2 아밀로이드 단백질은 자가조립(self-assembly)하는 것인 방법.The method according to claim 1, wherein the first and second amyloid proteins self-assemble.
  5. 청구항 1에 있어서, 상기 제1 또는 제2 아밀로이드 단백질은 전장 단백질 (full-length protein) 또는 단백질 단편인 것인 방법.The method according to claim 1, wherein the first or second amyloid protein is a full-length protein or a protein fragment.
  6. 청구항 5에 있어서, 상기 단백질 단편은 아밀로이드 단백질 내의 연속되는 6 내지 15mer인 것인 방법.The method of claim 5, wherein the protein fragment is a contiguous 6 to 15 mer within an amyloid protein.
  7. 청구항 1에 있어서, 상기 제1 아밀로이드 단백질은 시스테인을 통해 플레이트에 부착되는 것인 방법.The method of claim 1, wherein the first amyloid protein is attached to the plate through cysteine.
  8. 청구항 1에 있어서, 상기 제1 아밀로이드 단백질은 단량체 형태를 유지하도록 플레이트에 부착되는 것인 방법.The method of claim 1, wherein the first amyloid protein is attached to the plate to maintain its monomeric form.
  9. 청구항 1에 있어서, 상기 지표물질은 형광물질인 것인 방법.The method according to claim 1, wherein the indicator substance is a fluorescent substance.
  10. 청구항 1에 있어서, 상기 응집 억제제 후보물질은 제2 아밀로이드 단백질과 동시에 또는 순차적으로 처리되는 것인 방법.The method of claim 1, wherein the aggregation inhibitor candidate is treated simultaneously or sequentially with the second amyloid protein.
  11. 청구항 1에 있어서, 상기 응집 용해제 후보물질은 제1 아밀로이드 단백질과 제2 아밀로이드 단백질이 응집된 후 처리되는 것인 방법.The method according to claim 1, wherein the aggregation-dissolving agent candidate is treated after the first amyloid protein and the second amyloid protein are aggregated.
  12. 청구항 1에 있어서, 상기 (1)의 제1 아밀로이드 단백질은 0.1μM 내지 10μM 범위 내의 농도 또는 20시간 내지 26시간 범위 내의 시간으로 플레이트에 처리되는 것인 방법.The method according to claim 1, wherein the first amyloid protein of (1) is treated on the plate at a concentration in the range of 0.1 μM to 10 μM or for a time in the range of 20 hours to 26 hours.
  13. 청구항 1에 있어서, 상기 (1)의 제1 아밀로이드 단백질은 25℃ 내지 35℃ 범위 내의 온도 또는 pH 5 내지 pH 8 범위 내 pH 조건으로 플레이트에 처리되는 것인 방법.The method according to claim 1, wherein the first amyloid protein of (1) is treated on a plate at a temperature in the range of 25°C to 35°C or pH conditions in the range of pH 5 to pH 8.
  14. 청구항 1에 있어서, 상기 (2-1) 또는 (2-2)의 제2 아밀로이드 단백질은 5μM 내지 15μM 범위 내의 농도, 28℃ 내지 38℃ 범위 내의 온도 또는 pH 5 내지 pH 8 범위 내 pH 조건으로 제1 아밀로이드 단백질이 부착된 플레이트에 접촉시키는 것인 방법.The method according to claim 1, wherein the second amyloid protein of (2-1) or (2-2) is prepared at a concentration in the range of 5 μM to 15 μM, a temperature in the range of 28 ℃ to 38 ℃, or pH conditions in the range of pH 5 to pH 8. 1 A method of contacting a plate to which amyloid protein is attached.
  15. 청구항 1에 있어서, 상기 응집 억제제 또는 응집 용해제는 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인의 응집을 억제 또는 용해하는 것인 방법.The method of claim 1, wherein the aggregation inhibitor or aggregation dissolving agent inhibits or dissolves aggregation of amyloid beta, tau protein, or alpha-synuclein.
  16. 청구항 1에 있어서, 상기 응집 억제제 또는 응집 용해제는 퇴행성 뇌질환 치료제인 것인 방법.The method according to claim 1, wherein the aggregation inhibitor or aggregation-dissolving agent is a treatment for degenerative brain disease.
  17. 청구항 14에 있어서, 상기 퇴행성 뇌질환은 치매, 알츠하이머병(Alzheimer's disease), 전임상 알츠하이머병(preclinical alzheimer's disease), 파킨슨병(Parkinson's disease), 헌팅턴병 (Huntington's disease), 경도인지장애(mild cognitive impairment), 대뇌 아밀로이드 맥관병증, 다운증후근, 아밀로이드성 뇌졸증(stroke), 전신성 아밀로이드병, 더취(Dutch)형 아밀로이드증, 니만-픽병(Niemann-Pick disease), 노인성 치매, 근위축성 측삭 경화증(amyotrophic lateral sclerosis), 척수소뇌성 운동실조증(spinocerebellar atrophy), 뚜렛 증후군(Tourette's syndrome), 프리드리히 보행실조(Friedrich's ataxia), 마차도-조셉병(Machado-Joseph's disease), 루이 소체 치매(Lewy body dementia), 근육긴장이상(dystonia), 진행성 핵상 마비(progressive supranuclear palsy) 및 전두측두엽 치매(frontotemporal dementia)으로 이루어진 군으로부터 선택되는 어느 하나인 것인 방법.The method of claim 14, wherein the degenerative brain disease includes dementia, Alzheimer's disease, preclinical Alzheimer's disease, Parkinson's disease, Huntington's disease, mild cognitive impairment, Cerebral amyloid angiopathy, Down syndrome, amyloid stroke, systemic amyloid disease, Dutch amyloidosis, Niemann-Pick disease, senile dementia, amyotrophic lateral sclerosis, spinal cord Spinocerebellar atrophy, Tourette's syndrome, Friedrich's ataxia, Machado-Joseph's disease, Lewy body dementia, dystonia , progressive supranuclear palsy, and frontotemporal dementia.
  18. 제1 아밀로이드 단백질이 부착된 플레이트; 및 A plate to which the first amyloid protein is attached; and
    지표물질이 결합된 제2 아밀로이드 단백질을 포함하고,Contains a second amyloid protein to which an indicator substance is bound,
    상기 제1 및 제2 아밀로이드 단백질은 자가조립되는 것인, 단백질 응집 억제제 또는 응집 용해제를 스크리닝 하기 위한 키트.A kit for screening a protein aggregation inhibitor or aggregation dissolving agent, wherein the first and second amyloid proteins are self-assembled.
  19. 청구항 18에 있어서, 상기 제1 아밀로이드 단백질은 전장 단백질 또는 단백질 단편인 것인 키트.The kit according to claim 18, wherein the first amyloid protein is a full-length protein or a protein fragment.
  20. (1) 복수 개의 제1 아밀로이드 단백질 단편이 부착된 플레이트를 제공하는 단계;(1) providing a plate to which a plurality of first amyloid protein fragments are attached;
    (2-1) 단백질 응집 억제제 후보물질과 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시키는 단계; 또는(2-1) contacting the plate with a second amyloid protein bound to a protein aggregation inhibitor candidate and an indicator; or
    (2-2) 지표물질이 결합된 제2 아밀로이드 단백질을 상기 플레이트에 접촉시킨 후 단백질 응집 용해제 후보물질을 처리하는 단계; (2-2) contacting the second amyloid protein bound with an indicator substance to the plate and then treating the candidate substance with a protein aggregation dissolving agent;
    (3-1) 상기 (2-1) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하는 단계; 또는(3-1) measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-1); or
    (3-2) 상기 (2-2) 단계에서 제1 아밀로이드 단백질과 제2 아밀로이드의 응집률을 측정하는 단계를 포함하는 아밀로이드 단백질 응집 억제제 후보물질 또는 응집의 용해제 후보물질이 작용하는 부위를 스크리닝하는 방법.(3-2) Screening the site where the amyloid protein aggregation inhibitor candidate or aggregation dissolver candidate acts, including the step of measuring the aggregation rate of the first amyloid protein and the second amyloid in step (2-2). method.
  21. 청구항 20에 있어서, 상기 (2-1) 또는 (2-2) 단계는 제1 및 제2 아밀로이드 단백질의 응집을 유도하는 단계를 더 포함하는 것인 방법.The method of claim 20, wherein step (2-1) or (2-2) further comprises inducing aggregation of the first and second amyloid proteins.
  22. 청구항 20에 있어서, 상기 아밀로이드 단백질은 아밀로이드 베타, 타우 단백질, 또는 알파-시누클레인인 것인 방법.The method of claim 20, wherein the amyloid protein is amyloid beta, tau protein, or alpha-synuclein.
  23. 청구항 20에 있어서, 상기 단백질 단편은 아밀로이드 단백질 내의 연속되는 6 내지 15mer이고, 상기 복수개의 단백질 단편은 연속되는 2개 이상의 중첩되는 아미노산 잔기를 포함하는 것인 방법.The method of claim 20, wherein the protein fragment is a contiguous 6 to 15 mer in an amyloid protein, and the plurality of protein fragments include two or more consecutive overlapping amino acid residues.
  24. 청구항 23에 있어서, 응집 저해 또는 응집 용해 정도가 높게 측정되는 단백질 단편을 후보물질의 작용부위로 판정하는 단계를 더 포함하는 방법.The method of claim 23, further comprising determining a protein fragment for which a high degree of aggregation inhibition or aggregation dissolution is measured as the site of action of the candidate substance.
PCT/KR2023/015443 2022-10-07 2023-10-06 Method for screening for candidate material for inhibiting or disaggregating amyloid protein aggregation WO2024076210A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20220129004 2022-10-07
KR10-2022-0129004 2022-10-07

Publications (1)

Publication Number Publication Date
WO2024076210A1 true WO2024076210A1 (en) 2024-04-11

Family

ID=89859169

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/015443 WO2024076210A1 (en) 2022-10-07 2023-10-06 Method for screening for candidate material for inhibiting or disaggregating amyloid protein aggregation

Country Status (2)

Country Link
KR (1) KR102631987B1 (en)
WO (1) WO2024076210A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20040070221A (en) * 2001-12-13 2004-08-06 주식회사 포스코 Beta-amyloid binding factors and inhibitors thereof
KR20070051342A (en) * 2003-06-12 2007-05-17 에자이 가부시키가이샤 Neurocyte protective agent
US20070135334A1 (en) * 2002-09-06 2007-06-14 Tel Aviv University Future Technology Development L.P. Peptides and methods using same for diagnosing and treating amyloid-associated diseases
US20080268549A1 (en) * 2007-04-17 2008-10-30 Nicotera Thomas M Thioflavin t method for detection of amyloid polypeptide fibril aggregation
KR20110109128A (en) * 2010-03-30 2011-10-06 호서대학교 산학협력단 A METHOD FOR SCREENING THE INHIBITOR OF β-AMYLOID COAGULATION AND DISSOCIATION

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20040070221A (en) * 2001-12-13 2004-08-06 주식회사 포스코 Beta-amyloid binding factors and inhibitors thereof
US20070135334A1 (en) * 2002-09-06 2007-06-14 Tel Aviv University Future Technology Development L.P. Peptides and methods using same for diagnosing and treating amyloid-associated diseases
KR20070051342A (en) * 2003-06-12 2007-05-17 에자이 가부시키가이샤 Neurocyte protective agent
US20080268549A1 (en) * 2007-04-17 2008-10-30 Nicotera Thomas M Thioflavin t method for detection of amyloid polypeptide fibril aggregation
KR20110109128A (en) * 2010-03-30 2011-10-06 호서대학교 산학협력단 A METHOD FOR SCREENING THE INHIBITOR OF β-AMYLOID COAGULATION AND DISSOCIATION

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MERCEDES NOVO: "Critical aggregation concentration for the formation of early Amyloid-β (1–42) oligomers", SCIENTIFIC REPORTS, NATURE PUBLISHING GROUP, US, vol. 8, no. 1, 2018, US , XP093156686, ISSN: 2045-2322, DOI: 10.1038/s41598-018-19961-3 *
SHENG ZHANG: "Expression of N-Terminal Cysteine Aβ 42 and Conjugation to Generate Fluorescent and Biotinylated Aβ 42", BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY, vol. 60, no. 15, 20 April 2021 (2021-04-20), pages 1191 - 1200, XP093156687, ISSN: 0006-2960, DOI: 10.1021/acs.biochem.1c00105 *

Also Published As

Publication number Publication date
KR102631987B9 (en) 2024-04-08
KR102631987B1 (en) 2024-02-01

Similar Documents

Publication Publication Date Title
Ohhashi et al. The intrachain disulfide bond of β2-microglobulin is not essential for the immunoglobulin fold at neutral pH, but is essential for amyloid fibril formation at acidic pH
WO2017030292A1 (en) Prevention and treatment of neurodegenerative diseases through autophagy activity mediated by ligand or arginylated bip binding to p62 zz domain
WO2024076210A1 (en) Method for screening for candidate material for inhibiting or disaggregating amyloid protein aggregation
WO2021015544A1 (en) Benzoquinolizinium-based compound, preparation method therefor, and contrast medium composition comprising same
Patel et al. Localization of Locusta-DP in locust CNS and hemolymph satisfies initial hormonal criteria
WO2018194253A1 (en) Composition for biological tissue transparency and method for biological tissue transparency using same
WO2012050402A2 (en) Cell-permeable recombinant parkin protein and a pharmaceutical composition for treating degenerative brain diseases containing the same
WO2019182371A1 (en) Composition for diagnosis of degenerative neurological diseases
WO2019143125A1 (en) Composition for screening ongoing progress of alzheimer's disease by using beta amyloid oligomer in nasal discharge specimen and method for screening ongoing progress of alzheimer's disease by using same
WO2016144136A1 (en) Method for separating nucleic acids from fepe tissue
WO2018208011A2 (en) BIOCOMPATIBLE PEPTIDE SUPPRESSIVE OF AGGREGATION OF β-AMYLOID PROTEIN
Silva et al. Antiplasmodial activity study of angiotensin II via Ala scan analogs
WO2014137158A1 (en) Kit for diagnosing kidney diseases
Ramachandran et al. Skeletal muscle myosin cross‐bridge cycling is necessary for myofibrillogenesis
WO2020009551A1 (en) Graphene quantum dot as therapeutic agent for disease associated with abnormal fibrillation or aggregation of neuroprotein
Gimenez-Bonafé et al. Chromatin condensation, cysteine-rich protamine, and establishment of disulphide interprotamine bonds during spermiogenesis of Eledone cirrhosa (Cephalopoda)
WO2016153282A1 (en) Screening method for discovering autophagy-specific inhibitor
Dickerson et al. Structure–activity and immunochemical data provide evidence of developmental-and tissue-specific myosuppressin signaling
WO2018203613A1 (en) Peptide for toll-like receptor (tlr) inhibition and pharmaceutical composition comprising same
WO2010016706A9 (en) Novel use of idbf
WO2022055334A1 (en) Composition for preventing or treating pulmonary fibrosis disease
WO2021221444A1 (en) Compound as ubr-box domain ligand
WO2018062588A1 (en) Curcumin derivative, method for producing same, and photo-acoustic imaging agent comprising same for detecting beta-amyloid plaque
EP4208542A1 (en) Improved cell-permeable modified parkin recombinant protein for treatment of neurodegenerative diseases and use thereof
WO2024029995A1 (en) Amyloid beta-specific peptide cbrv1-04369 and composition for treating alzheimer's disease comprising same

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: 23875271

Country of ref document: EP

Kind code of ref document: A1