WO2019134481A1 - Uses of protein in preparing drug for preventing and treating alzheimer's disease - Google Patents

Uses of protein in preparing drug for preventing and treating alzheimer's disease Download PDF

Info

Publication number
WO2019134481A1
WO2019134481A1 PCT/CN2018/120172 CN2018120172W WO2019134481A1 WO 2019134481 A1 WO2019134481 A1 WO 2019134481A1 CN 2018120172 W CN2018120172 W CN 2018120172W WO 2019134481 A1 WO2019134481 A1 WO 2019134481A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
sdss1
modification
drug
amino acid
Prior art date
Application number
PCT/CN2018/120172
Other languages
French (fr)
Chinese (zh)
Inventor
张英豪
付晶鹏
Original Assignee
上海清流生物医药科技有限公司
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 上海清流生物医药科技有限公司 filed Critical 上海清流生物医药科技有限公司
Publication of WO2019134481A1 publication Critical patent/WO2019134481A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes

Definitions

  • the present invention relates to the use of the sDSS1 protein drug for the preparation of a medicament for preventing and treating dementia, comprising a prophylactic medicament for the preparation of degenerative dementia and vascular dementia, or for preparing a pre-, early, intermediate or late stage of these diseases.
  • Therapeutic drugs for the preparation of a medicament for preventing and treating dementia, comprising a prophylactic medicament for the preparation of degenerative dementia and vascular dementia, or for preparing a pre-, early, intermediate or late stage of these diseases.
  • Dementia is a type of brain disease that causes long-term and progressive deterioration of patient thinking and learning memory.
  • the most common form of dementia is Alzheimer's Disease (AD), which accounts for dementia. 50-70% of patients, others include vascular dementia ( 25%), Louise-type dementia ( 15%) and so on.
  • AD Alzheimer's Disease
  • the number of people with dementia worldwide is about 46.8 million, and the number of patients worldwide is expected to reach 75 million in 2030 [1].
  • AD is a typical neurodegenerative disease with clinical manifestations of progressive learning and memory impairment and neurological damage.
  • a typical pathological feature of AD patients is the presence of ⁇ -amyloid (A ⁇ ) plaques and tau deposition in nerve tissue.
  • the ⁇ amyloid hypothesis suggests that the progressive accumulation of toxic proteins caused by the imbalance of A ⁇ protein production and clearance in nerve tissue and the resulting synaptic dysfunction and neuronal death are key mechanisms leading to disease [2] [3].
  • the researchers hope to prevent A ⁇ protein production and aggregation, or promote A ⁇ clearance, in order to prevent or treat diseases [4].
  • the results of AD studies based on animal experiments show that antibodies [5] [6], peptide drugs [7], small molecule drugs [8] can block A ⁇ aggregation or production, reduce plaque formation, and reduce animal disease indications. Therefore, the drug inhibits and clears the pathological deposition of A ⁇ or can be an effective way to treat AD.
  • Shfm1 split hand/split foot malformation type 1 gene is one of the key genes in human crab claw disease. It is highly conserved in evolution and its encoded protein DSS1 is involved in stable genome, homologous gene recombination, DNA damage repair and cell proliferation. Etc. [9-12]. The results of the inventors of the present invention show that the DSS1 protein can be added to the oxidized protein by a energy-consuming enzymatic reaction to help the cells clear the oxidized protein [13]. These results show the important role of DSS1 protein in biological activities.
  • the transient accumulation of A ⁇ protein is one of the key factors in the pathogenesis of AD. It is one of the main ways to treat AD diseases by removing A ⁇ or inhibiting A ⁇ aggregation.
  • the sDSS1 protein provided by the invention can bind to A ⁇ protein to accelerate the clearance of A ⁇ protein, reduce the accumulation of A ⁇ protein, improve the symptoms of the disease, and has the potential for preparing clinical AD and other dementia prevention drugs and therapeutic drugs.
  • a protein for the preparation of a medicament for the prevention and treatment of dementia which is the use of the sDSS1 protein for the preparation of a medicament for the prevention and treatment of dementia.
  • the dementia refers to degenerative dementia, including Alzheimer's disease, Alzheimer's disease, Lewy body dementia, and frontotemporal dementia.
  • the dementia is vascular dementia.
  • the dementia is pre-dementia dementia, mild dementia dementia, moderate dementia dementia, and severe dementia dementia.
  • the sDSS1 protein comprises human, chimpanzee, bonobo, gorilla, orangutan, white-cheeked gibbons, golden monkey, rhesus monkey, golden monkey, East African pheasant, Angora simian, white-tailed white-browed monkey, ghost A basic protein formed by any sDSS1 protein sequence in scorpionfish, porpoise monkey, wherein the amino acid sequence of human sDSS1 is SEQ ID NO: 1, the amino acid sequence of chimpanzee sDSS1 is SEQ ID NO: 2, and the amino acid sequence of porcine chimpanzee sDSS1 is SEQ.
  • the amino acid sequence of gorilla sDSS1 is SEQ ID NO: 4
  • the amino acid sequence of orangutan sDSS1 is SEQ ID NO: 5
  • the amino acid sequence of white buccal gibbon sDSS1 is SEQ ID NO: 6
  • the golden monkey sDSS1 The amino acid sequence of SEQ ID NO: 7, the amino acid sequence of rhesus sDSS1 is SEQ ID NO: 8
  • the amino acid sequence of gilt monkey sDSS1 is SEQ ID NO: 9
  • the amino acid sequence of ssDSS1 is SEQ ID NO: 10, Angola.
  • amino acid sequence of simian sDSS1 is SEQ ID NO: 11
  • amino acid sequence of leucocephalus sDSS1 is SEQ ID NO: 12
  • amino acid sequence of scorpion sDSS1 is SEQ ID NO: 13.
  • the sDSS1 protein is the first protein having a similarity to the basic protein of 70% or more.
  • the sDSS1 protein is based on 58 amino acids of the nitrogen terminus of the basic protein, and fuses other polypeptide fragments at the nitrogen terminal or the carbon terminal, and the structural features or amino acid sequence characteristics of the polypeptide fragment for fusion are The second protein of the same or similar sequence of the carbon end of the basic protein.
  • the sDSS1 protein is based on the basic amino acid of the basic protein of 58 amino acids, and the other amino acid fragments are fused at the nitrogen or carbon end, and the fused protein can realize the transmembrane transport function. .
  • the sDSS1 protein is a fusion formed by using the basic protein, the first protein, the second protein or the third protein to form a protein, a carrier protein, an antibody or other amino acid fragments of any length. protein.
  • the sDSS1 protein is a polypeptide/protein modification produced by modification of the basic protein, the first protein, the second protein, the third protein or the fusion protein.
  • the polypeptide/protein modification is directed to an amino group on an amino acid side chain, a carbonyl group on an amino acid side chain, a nitrogen terminal amino group, a carbon terminal carbonyl group, a cysteine, a tyrosine, a serine, a tryptophan.
  • the method for modifying the polypeptide/protein modification comprises glycosylation modification, fatty acid modification, acylation modification, Fc fragment fusion, albumin fusion, polyethylene glycol modification, dextran modification, heparin modification, polyethylene Pyrrolidone modification, polyamino acid modification, polysialic acid modification, chitosan and its derivative modification, lectin modification, sodium alginate modification, carbomer modification, polyvinylpyrrolidone modification, hydroxypropyl methylcellulose modification, One or more of hydroxypropylcellulose modification, acetylation modification, formylation modification, phosphorylation modification, methylation modification, sulfonation modification, and other pharmaceutically acceptable polypeptide/protein drug modification methods.
  • the sDSS1 protein is an amino acid other than the 20 basic amino acids based on the amino acid sequence of the basic protein, the first protein, the second protein, the third protein or the fusion protein. - 31 non-natural amino acid replacement proteins substituted with any amino acid site.
  • the amino acid substitution of the non-natural amino acid replacement protein comprises replacement with hydroxyproline, hydroxylysine, selenocysteine, D-form amino acid or synthetic non-natural amino acid and derivatives thereof.
  • the sDSS1 protein is pharmaceutically applicable to the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the non-natural amino acid replacement protein. Part or all of the complex formed by the drug carrier.
  • the pharmaceutical carrier of the complex comprises one or one of an enteric coating preparation, a capsule, a microsphere/capsule, a liposome, a microemulsion, a double emulsion, a nanoparticle, a magnetic particle, a gelatin, and a gel. the above.
  • the base of the protein, the first protein, second protein and third protein, fusion protein, polypeptide / protein or complex modifications play operation starting concentration of toxic proteins toxic shield is not less than 0.2 ⁇ g /mL .
  • the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the complex exhibits a molar concentration ratio of shielding toxic protein toxicity of not less than 0.010;
  • the molar concentration ratio refers to the ratio of the molar concentration of the protein, polypeptide/protein modification or complex in the reaction system to the molar concentration of the toxic protein.
  • the sDSS1 protein targets the individual's own sDSS1 protein, and affects the level of the individual's own sDSS1 protein by the exogenous drug.
  • the drug is a drug target of a sDSS1 protein, a gene of sDSS1 protein, a regulatory element of a gene of sDSS1 or a gene of sDSS1.
  • the drug modulates the amount of sDSS1 protein in the blood or cerebrospinal fluid by affecting protease/peptidase activity in blood or cerebrospinal fluid.
  • the drug is the first drug formed by a chemical small molecule drug, an antibody, a polypeptide/protein drug, a nucleic acid drug or a nano drug.
  • the sDSS1 protein is any one of the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification, the complex, and the first drug.
  • the sDSS1 protein is a basic protein, a first protein, a second protein, a third protein, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, A third drug formed by the combination of one, two or more of any one of the first drugs with a pharmaceutically acceptable excipient.
  • the sDSS1 protein is a fourth obtained by introducing a nucleotide sequence encoding the basic protein, the first protein, the second protein, the third protein or the fusion protein into the body and expressing the same by an expression system. Protein.
  • the expression system is eukaryotic expression plasmid vector, adenovirus, adeno-associated virus, lentivirus, retrovirus, baculovirus, herpes virus, pseudorabies virus, ZFN gene editing technology, TALEN gene editing technology, CRISPR/Cas gene editing technology or other medically available gene editing technology or viral vector.
  • the sDSS1 protein is a fifth protein formed by the basic protein, the first protein, the second protein, the third protein or the fusion protein obtained by the transplanted cell in the individual.
  • the cell is a stem cell, a precursor cell or an adult cell of any one of humans.
  • the stem cells are embryonic stem cells, induced pluripotent stem cells, cells obtained by transdifferentiation, or stem cells derived from primary culture, pluripotent or pluripotent stem cells differentiated from mother cells.
  • the sDSS1 protein is a sixth protein formed by the basic protein, the first protein, the second protein, the third protein or the fusion protein obtained by transplanting tissues or organs in an individual.
  • the tissue is a whole organ or part of a tissue block of the brain, liver, kidney, spleen, islet, or blood, fat, muscle, bone marrow, skin.
  • the sDSS1 protein is a seventh protein introduced into the body by serum, cerebrospinal fluid, and interstitial fluid infusion.
  • the prophylactic agent comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, a first drug, and a second Drugs, third drugs, protein drugs in expression systems, cells, tissues, organs, body fluids, tissue fluids, peptide drugs, nucleic acid drugs, chemical small molecule drugs, cell products, commercial transplant tissues, injections, lyophilized powder, One or more of health care products and food additives.
  • the therapeutic agent comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, a first drug, and a second Drugs, third drugs, protein drugs in expression systems, cells, tissues, organs, body fluids, tissue fluids, peptide drugs, nucleic acid drugs, chemical small molecule drugs, cell products, commercial transplant tissues, injections, lyophilized powder, One or more of health care products and food additives.
  • the sDSS1 protein provided by the invention binds to A ⁇ protein, inhibits the aggregation of A ⁇ protein and reduces the binding ability of A ⁇ protein to its receptor protein, and effectively alleviates the cytotoxicity caused by A ⁇ protein.
  • the sDSS1 protein provided by the present invention can accelerate the clearance of A ⁇ protein by microglia, and shields A ⁇ oligomer toxicity and reduces microglia activation induced by A ⁇ oligomer.
  • the sDSS1 protein provided by the present invention significantly improves the disease symptoms of the AD model nematode on the CL4176 nematode and reduces the accumulation of A ⁇ protein in the nematode tissue.
  • the sDSS1 protein provided by the present invention helps mice clear A ⁇ and reduce A ⁇ levels in the blood. Long-term injection of sDSS1 protein into AD model APP/PS1 mice can effectively reduce A ⁇ plaque deposition in brain tissue.
  • the sDSS1 protein provided by the present invention is a protein possessed by humans and other primates, has a relatively small molecular weight, low immunogenicity, and has a natural protein degradation mechanism in vivo, and therefore, clinical application does not cause significant immunity.
  • the reaction or other toxic side effects are safe and reliable.
  • the present invention provides an sDSS1 protein drug for the treatment of dementia.
  • the sDSS1 protein can bind to A ⁇ protein, inhibit A ⁇ aggregation, and reduce A ⁇ protein and receptor by molecular, cell and animal level experiments.
  • the binding efficiency promotes microglia clearance of A ⁇ and reduces microglia activation.
  • sDSS1 protein promotes A ⁇ clearance in the blood, reduces the accumulation of A ⁇ protein in tissues, and alleviates disease symptoms.
  • sDSS1 protein has low immunogenicity and remarkable efficacy, and has the potential to be used for clinical preparation of dementia prevention drugs or for preparing pre-treatment, early, intermediate, and advanced dementia drugs.
  • FIG. 1A-1B Molecular experiments show that A[beta] protein binds to sDSSl protein.
  • a ⁇ 1-42 protein and A ⁇ 1-40 protein were incubated with sDSS1 protein, respectively, and a complex of A ⁇ 1-42 protein and sDSS1 protein was detected by A ⁇ protein antibody (molecular weight 10KD-37KD, Lane 3 and lane 4).
  • a ⁇ protein antibody molecular weight 10KD-37KD, Lane 3 and lane 4
  • the A ⁇ 1-40 protein is incubated alone to form a partial dimer, and incubation with the sDSS1 protein reduces dimer formation.
  • sDSS1 protein and A ⁇ 1-42 protein (lane 3 and lane 4) or sDSS1 protein and A ⁇ 1-40 protein (lane 6 and lane 7) can also be detected by sDSS1 protein antibody ( The molecular weight is between 37 and 150 KD), and the complex formation increases as the concentration of sDSS1 protein in the reaction system increases.
  • ThT assay detects A[beta] 1-42 protein aggregation.
  • the A ⁇ 1-42 protein aggregates at 12 hours and shows an ascending curve.
  • Addition of 5% or 10% sDSS1 protein according to the molecular weight ratio inhibited the ascending curve and prolonged the detection time without observing the rising curve, suggesting that no A ⁇ 1-42 protein aggregation occurred.
  • FIG. 1 D ThT assay detects A ⁇ 1-40 protein aggregation.
  • the A ⁇ 1-40 protein began to show a rising curve at 24 hours, indicating aggregate formation.
  • Addition of 5% or 10% sDSS1 protein according to the molecular weight ratio inhibited the ascending curve, and the prolongation of the detection time did not observe a rising curve, suggesting that no A ⁇ 1-40 protein aggregation occurred.
  • FIG. 1E ThT assay detects sDSS1 protein mutant 4 inhibits A ⁇ 1-40 protein aggregation.
  • the A ⁇ 1-40 protein aggregates at 20-24 hours and shows an ascending curve.
  • 0.667%, 0, 3.33%, or 16.67% sDSS1(M4) protein was added according to the molecular weight ratio, the rise curve was inhibited, and the rise time was not observed, indicating that no A ⁇ 1-40 protein aggregation occurred.
  • FIG. 1F ThT assay detects sDSS1 protein mutant 11 inhibiting A ⁇ 1-40 protein aggregation.
  • the A ⁇ 1-40 protein aggregates at 20-24 hours and shows an ascending curve.
  • 0.667%, 0, 3.33%, or 16.67% sDSS1(M11) protein was added according to the molecular weight ratio, the rising curve was inhibited, and the rise time was not observed, indicating that no A ⁇ 1-40 protein aggregation occurred.
  • the A ⁇ 1-42 protein oligomer and the A ⁇ 1-40 protein oligomer were incubated with the sDSS1 protein, respectively, and the complex formed by the A ⁇ 1-42 protein and the sDSS1 protein was detected by the A ⁇ protein antibody (molecular weight 10KD). Between -37KD, lane 3 and lane 4). No significant difference was detected between the A ⁇ 1-42 protein and the sDSS1 protein.
  • a complex of sDSS1 protein and A ⁇ 1-42 oligomer (lane 3) or sDSS1 protein and A ⁇ 1-40 oligomer (lane 5) can be detected by using sDSS1 protein antibody (molecular weight 37-150KD) between).
  • ThT assay detects the reaction of sDSS1 protein with A ⁇ 1-40 oligomer.
  • the addition of 50% and 100% sDSS1 protein in a molecular weight ratio of 30 ⁇ M A ⁇ 1-40 oligomer showed an increase in the fluorescence value of the A ⁇ 1-40 oligomer, and the fluorescence value remained substantially unchanged during the 12-hour detection period.
  • ThT assay detects the reaction of the sDSS1 protein with the A ⁇ 1-42 oligomer.
  • the addition of 50% and 100% sDSS1 protein in a molecular weight ratio of 30 ⁇ M A ⁇ 1-42 oligomer showed an increase in the fluorescence value of the A ⁇ 1-42 oligomer.
  • the fluorescence value of the A ⁇ 1-42 oligomer also increased, and the fluorescence value increased after the addition of the sDSS1 protein.
  • FIG. 3A The sDSS1 protein is unable to interact with the RAGE receptor protein.
  • the results of molecular interaction test showed that on the binding curve, it can be seen that after the injection of sDSS1 protein, no obvious ascending binding curve was observed, and no obvious dissociation curve was observed.
  • sDSS1 protein inhibits the interaction of A ⁇ protein with RAGE receptor protein.
  • a ⁇ 1-42 protein can bind to RAGE receptor protein, and the addition of sDSS1 protein can inhibit the binding efficiency of A ⁇ protein to RAGE, the binding amount of A ⁇ protein decreases, and the effect shows a typical dose-dependent effect.
  • FIGS. 4A-4C Cell experiments demonstrate that sDSS1 protein shields A ⁇ 1-42 oligomer toxicity and protects N2a cell viability.
  • a ⁇ 1-42 oligomers cause N2a cytotoxicity, and the addition of A ⁇ 1-42 oligomers causes abnormal aggregation and rounding of cells, and sDSS1 protein can alleviate these phenomena.
  • FIG. 2B detection of cell viability revealed that sDSS1 protein can reduce cell viability by A ⁇ 1-42 oligomer. After the addition of the same amount of sDSS1 protein, cell viability is almost unaffected by the toxicity of A ⁇ 1-42 oligomer.
  • FIGS 5A-5B Cell experiments demonstrate that sDSS1 protein shields A ⁇ 1-42 oligomer toxicity and protects primary neuronal cell viability.
  • 10 ⁇ M A ⁇ 1-42 oligomers can cause a decrease in neuronal cell viability, and cell viability can be recovered after the addition of 5 ⁇ M and 10 ⁇ M sDSS1 protein.
  • sDSS1 protein can reduce the cytotoxicity level caused by A ⁇ 1-42 protein, and 10 ⁇ M sDSS1 protein can protect cells from the toxicity of A ⁇ 1-42 oligomer.
  • N 10 per group. Data were analyzed by ANOVE, *, p-value ⁇ 0.05; **, p-value ⁇ 0.01.
  • FIGS. 6A-6B The sDSS1 protein promotes microglia clearance of A ⁇ protein.
  • FIG. 6A A ⁇ 1-42 protein and sDSS1 protein were added to the medium.
  • the concentration of A ⁇ protein in microglia was significantly higher than that of the control with only A ⁇ protein added after 24 hours, with the increase of the concentration of sDSS1 protein added.
  • the cytosolic A ⁇ protein concentration is increased.
  • FIGS 6C-6D The sDSS1 protein reduces microglial activation by A ⁇ 1-42 protein oligomers.
  • FIG. 6C A ⁇ 1-42 protein oligomers caused microglia activation, which showed an increase in ROS levels in BV2 cells.
  • ROS levels in BV2 cells gradually decreased, which was close to the control group.
  • FIG. 6D A ⁇ 1-42 protein oligomer stimulates microglia to release inflammatory factor TNF- ⁇ , and sDSS1 protein reduces TNF- ⁇ release.
  • N 3 per group. Data were analyzed by ANOVE, *, p-value ⁇ 0.05; **, p-value ⁇ 0.01.
  • FIG. 7A The sDSS1 protein ameliorate the disease symptoms of CL4176 nematodes.
  • the sputum rate of the AD model CL4176 nematode gradually increased, and the addition of sDSS1 protein to the culture medium significantly slowed down the rate of sputum.
  • the mitigation efficiency showed drug dose dependence, and the mitigation effect increased with the increase of sDSS1 protein concentration.
  • the ratio of the control group to the nematode was only 15.09%, the 500 ⁇ g/mL sDSS1 protein administration group was 39.47%, and the 1000 ⁇ g/mL sDSS1 protein administration group was 65.71%.
  • the nematode in the control group was almost all sputum (the ratio of the nematode was less than 1%), while in the 500 ⁇ g/mL sDSS1 protein-administered group, the proportion of the untreated nematode was 18.42%, and the 1000 ⁇ g/mL sDSS1 protein-administered group was 48.57. %.
  • the number of nematodes per group is 90.
  • the experiment was repeated 3 times and the data was analyzed by Log-rank (Mantel-Cox) test, ***, p-value ⁇ 0.001.
  • FIG. 7B Immunohistochemical staining for the detection of A ⁇ 1-42 deposition in C14176 nematode tissue.
  • AD model 4176 elegans tissues were fixed and immunofluorescent stained, A ⁇ 1-42 specific antibodies were used to display A ⁇ protein (green fluorescence), Hoechst dye was used to stain nuclei (blue fluorescence), and white arrows indicate A ⁇ plaque deposition.
  • Typical A ⁇ 1-42 plaques and diffuse A ⁇ precipitates were seen in the control nematode tissues. After treatment with 500 ⁇ g/mL sDSS1 protein, A ⁇ plaques and diffuse deposition were significantly reduced. The 1000 ⁇ g/mL sDSS1 protein administration group showed almost no plaque and less diffuse deposition.
  • FIG. 7C - Figure 7D 4176 Reduction of A[beta] 1-42 aggregates and total A[beta] protein content in nematode tissue mills.
  • FIG. 7C Western blotting was used to detect A ⁇ 1-42 aggregates in nematode tissue mills. It can be seen that after treatment with sDSS1 protein, most A ⁇ aggregate bands decreased or disappeared completely (black arrow indicates position).
  • Fig. 7D the total A ⁇ protein in the tissue slurry was detected by ELISA, and it was found that the total A ⁇ protein of the nematode tissue of the sDSS1 protein administration group was significantly decreased, and the content of the 1000 ⁇ g/mL sDSS1 protein administration group was only 59.07% of the control group.
  • the number of nematodes used in each group was 400 in the experiment.
  • FIGS 8A-8B sDSS1 protein promotes A ⁇ protein clearance in mouse blood.
  • Figure 8A shows a model of acute A ⁇ rise by intravenous injection of A ⁇ protein into mice. The results show that sDSS1 protein administration accelerates A ⁇ protein clearance. As the amount of sDSS1 protein administered increased, the A ⁇ content decreased significantly.
  • FIG. 8B Injecting sDSS1 protein into AD model APP/PS1 mice, a significant decrease in A ⁇ levels was also detected in the blood.
  • FIGS 9A-9C sDSS1 protein reduces A ⁇ plaque deposition in brain tissue of APP/PS1 mice.
  • Fig. 9A after staining with FSB dye, a large amount of A ⁇ plaque precipitate (blue fluorescence) was observed in the brain tissue of APP/PS1 mice at 11 months of age. After administration of sDSS1 protein, plaque deposition in the hippocampus of brain tissue decreased. The number of plaques is reduced and the brightness is reduced.
  • Fig. 9B after the photo was processed by Phototshop CS software, the plaque area was counted. The results showed that the plaque area of brain tissue after sDSS1 protein administration was significantly lower than that of the control group.
  • FIG 9C the number of small plaques (marked as 1 in Figure 9A), medium-sized plaques (marked as 2 in Figure 9A), and large plaques (marked as 3 in Figure 9A) were counted according to plaque size. In contrast, the number of medium-sized plaques and large plaques in tissues was significantly decreased after administration of sDSS1 protein, and there was no significant difference in small plaques.
  • APP/PS1 mice were 3 in each group. The data was analyzed by ANOVE, n.s., p-value>0.5; **, p-value ⁇ 0.01.
  • the sDSS1 protein used in the following examples is the human sDSS1 protein produced by the company itself, and the protein sequence is shown in SEQ ID NO: 1.
  • the company controls the quality of protein.
  • the purity of the tested protein is greater than 95%.
  • the endotoxin (less than 3EU/mg protein) and other impurities are in compliance with the standard and can be used in animal experiments without causing significant animal toxicity.
  • Example 1 The sDSS1 protein inhibits A ⁇ protein aggregation.
  • sDSS1 protein, sDSS1 mutants 4 (sDSS1 (M4)) (amino acid sequence is shown in SEQ ID 15), sDSS1 mutants 11 (amino acid sequence see SEQ ID 16) (sDSS1 (M11 )) were diluted to 1mg / mL in PBS (100 ⁇ M).
  • the experiment was carried out in a PBS environment. First, a PBS was added in a 1.5 mL centrifuge tube, and then sDSS1 protein or mutant protein, A ⁇ protein and ThT dye were sequentially added, and the sDSS1 protein molar concentration was added according to a preset concentration gradient.
  • the molar concentrations of the A ⁇ protein and the ThT dye were 30 ⁇ M and 5 ⁇ M, respectively; for the A ⁇ 1-40 protein, the molar concentrations of the A ⁇ protein and the ThT dye were 30 ⁇ M and 10 ⁇ M, respectively.
  • the solution was shaken and mixed on the suspension, and then 200 ⁇ L was added to a black fluorescence detection plate (Costar, 3792) for two duplicate wells per group.
  • the black fluorescence detection plate was placed on a multi-function microplate reader (Molecular Devices, SpectraMax i3x) to detect the fluorescence value, and the excitation light was set to 450 nm, the emission light was 485 nm, the detection interval was 30 minutes, and the detection was continued for 48 hours.
  • a multi-function microplate reader Molecular Devices, SpectraMax i3x
  • Protein Interactions A ⁇ 1-42 and A ⁇ 1-40 protein monomers were diluted to 20 ⁇ M with PBS and mixed with 10 ⁇ M or 20 ⁇ M sDSS1 protein, and 20 ⁇ M A ⁇ protein was used as a positive control.
  • the protein solution was mixed and incubated overnight at 4 °C. 5x loading buffer was added to the incubated protein solution, mixed and heated at 100 ° C for 10 minutes, and the prepared samples were used for Western Blotting analysis.
  • a ⁇ 1-42 or A ⁇ 1-40 protein was co-incubated with sDSS1 protein, and the mixture after incubation was detected. It was found that A ⁇ 1-42 protein forms a complex with sDSS1 protein (molecular weight 10KD-37KD). Co-incubation of the A ⁇ 1-40 protein with the sDSS1 protein significantly reduced the formation of dimers (Fig. 1A). The A ⁇ protein and sDSS1 protein complex signals were detected by the same technique using the sDSS1 protein antibody, and the complex signal was enhanced as the sDSS1 protein concentration was increased (Fig. 1B). A ⁇ 1-42 or A ⁇ 1-40 monomeric proteins aggregate to form an amyloid deposit, which is enhanced by binding to ThT dye.
  • Example 2 The sDSS1 protein binds to an A ⁇ protein oligomer.
  • a ⁇ 1-42 protein solubilization and oligomer preparation 1 mg A ⁇ 1-42 or A ⁇ 1-40 protein was added to 500 ⁇ L of 20 mM NaOH solution, shaken and mixed for 10 minutes, and then sonicated for 10 minutes to help dissolve the protein. Dilute to 100 ⁇ M with PBS solution, incubate at 4 ° C for 24 hours, centrifuge at 12000 g for 10 minutes to remove possible precipitates, and obtain A ⁇ 1-42 or A ⁇ 1-40 oligomers. A ⁇ oligomers are labeled according to the concentration of A ⁇ monomer protein prior to incubation.
  • ThT assay The sDSS1 protein was diluted to 1 mg/mL (100 ⁇ M) with PBS. The experiment was carried out under PBS conditions. First, PBS was added in a 1.5 mL centrifuge tube, and then sDSS1 protein, A ⁇ protein oligomer and ThT dye were sequentially added, and the molar concentration of sDSS1 protein was added according to a preset concentration gradient. For the A ⁇ 1-42 protein, the molar concentrations of the A ⁇ protein and the ThT dye were 30 ⁇ M and 5 m ⁇ , respectively; for the A ⁇ 1-40 protein, the molar concentrations of the A ⁇ protein and the ThT dye were 30 ⁇ M and 10 m ⁇ , respectively.
  • the solution was shaken and mixed on the suspension, and then 200 ⁇ L was added to a black fluorescence detection plate (Costar, 3792) for two duplicate wells per group.
  • the black fluorescent detection plate was placed on a multi-function microplate reader to detect the fluorescence value, and the excitation light was set to 450 nm, the emission light was 485 nm, the detection interval was 30 minutes, and the detection was continued for 12 hours.
  • a ⁇ 1-42 or A ⁇ 1-40 protein oligomer was diluted to 20 ⁇ M with PBS and mixed with 10 ⁇ M or 20 ⁇ M sDSS1 protein, and 20 ⁇ M A ⁇ oligomer was used as a positive control.
  • the protein solution was mixed and incubated overnight at 4 °C. 5x loading buffer was added to the incubated protein solution, mixed and heated at 100 ° C for 10 minutes, and the prepared samples were used for Western Blotting analysis.
  • the oligomer of A ⁇ 1-40 or A ⁇ 1-42 is the major form of A ⁇ protein producing cytotoxicity.
  • the A ⁇ protein was first incubated to form an oligomer, and then incubated with sDSS1 protein for western blotting and ThT detection.
  • the results showed that the reaction of sDSS1 protein with A ⁇ 1-42 oligomer form was detected by A ⁇ protein antibody, and the complex content increased with the increase of sDSS1 protein concentration in the reaction system (Fig. 2A).
  • a complex formed by the sDSS1 protein and the A ⁇ protein can also be detected using the sDSS1 protein antibody (Fig. 2B).
  • the sDSS1 protein can react with the amyloid precipitation of A ⁇ 1-40 or A ⁇ 1-42, and the fluorescence intensity increases after the sDSS1 protein reacts with the A ⁇ 1-40 amyloid precipitate (Fig. 2C); After the reaction of the sDSS1 protein with the A ⁇ 1-42 amyloid precipitate, the fluorescence intensity was significantly reduced (Fig. 2D).
  • the above results indicate that the sDSS1 protein can also form a complex with the A ⁇ protein oligomer form, and these interactions may inhibit the cytotoxicity of the A ⁇ protein.
  • Example 3 sDSS1 protein reduces the binding efficiency of A ⁇ protein to receptor protein.
  • the biomacromolecular interaction instrument (GE, Biacore 3000) was used to detect the binding efficiency of RAGE protein to sDSS1 protein.
  • the chip was first rinsed with PBS for 20 minutes until the baseline was stable, and the protein was coupled to the sensor chip NTA chip (GE) with a His tag on 1 ⁇ g/mL RAGE protein with a coupling amount of approximately 50 RU.
  • the sDSS1 protein was diluted to 10 ⁇ g/mL, the loading amount was 20 ⁇ L, and the loading time was 200 seconds. After completion, rinse with PBS solution and measure the elution curve.
  • RAGE protein was purchased from Beijing Yiqiao Shenzhou Technology Co., Ltd. (11629-H02H).
  • the blank plate was washed once with PBS, and 100 ⁇ L of 0.5 ⁇ g/mL RAGE protein diluted with PBS was added to each well overnight at 4 ° C, and washed once with PBS.
  • the PBS diluted 2% BSA in PBST (0.05% Tween 20) was added and blocked at 37 ° C for 1 hour, and then washed once with PBS.
  • a ⁇ 1-42 protein or A ⁇ 1-42 protein and sDSS1 protein mixture set A ⁇ 1-42 protein concentration to 0.625 ⁇ g/mL and 0.3125 ⁇ g/mL, respectively, and sDSS1 protein concentration of 0.2 ⁇ g/mL and 1 ⁇ g/mL, respectively, incubate at room temperature. 2 hours.
  • PBST 0.05% Tween 20
  • 100 ⁇ L of Rabbit-anti-A ⁇ (1:10000 diluted in PBST solution containing 0.5% BSA) was added, and the mixture was incubated at room temperature for 1 hour, and then washed three times with PBST.
  • HRP horseradish peroxidase
  • sDSS1 protein could not bind to the RAGE protein and showed no significant binding peak on BIAcore (Fig. 3A).
  • ELISA experiments showed that A ⁇ 1-42 protein as a ligand can bind to RAGE protein, showing a higher binding amount.
  • the addition of 0.2 ⁇ g/mL and 1 ⁇ g/mL sDSS1 protein reduced the binding amount of A ⁇ 1-42 to RAGE, and this effect exhibited a significant concentration-dependent effect ( FIG. 3B ).
  • Example 4 sDSS1 protein shields cytotoxicity caused by A ⁇ oligomers.
  • N2a Cell Culture Mouse-derived neuroblastoma cells
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal calf serum
  • mice that were pregnant for 16 days were deeply anesthetized with ether and then sacrificed by cervical dislocation.
  • the E16 stage fetus was dissected.
  • the fetal rats were anesthetized with ice
  • the cervical vertebrae were sacrificed, the rat brain was dissected, and immersed in a pre-cooled DMEM solution (containing 2% double-antibody).
  • the pia mater was removed by ophthalmology and repeatedly washed to remove blood.
  • the frontal cortex was dissected and the tissue was cut with an ophthalmic scissors. All tissues were collected into a centrifuge tube and digested with 5 mL of 0.05% trypsin for 15 minutes at room temperature.
  • the digestion was terminated by adding an equal volume of DMEM complete medium, and the tissue was repeatedly beaten and filtered with a 40 ⁇ m cell strainer to obtain a cell suspension. After the cells were counted, they were added to a 96-well plate at a rate of 3.0 x 10 4 cells per well, 200 ⁇ L of DMEM complete medium, and the plate was incubated at 37 °C. After 24 hours, switch to 200 ⁇ L of neuronal medium containing 98% Neurobasal (Gibco, 21103049), 2% B27 supplement (Gibco, 17504044), 1% GlutaMAX-1supplement (Gibco, 35050061). Fresh neuronal medium was changed every two days thereafter. Cultured neurons matured after 8 days and can be used in subsequent experiments.
  • a ⁇ 1-42 protein solubilization and oligomer preparation 1 mg of A ⁇ 1-42 was diluted to 100 ⁇ M with serum-free DMEM-/- medium, and the dilution was incubated at 4 ° C for 24 hours to obtain A ⁇ 1-42 oligomer protein.
  • the A ⁇ oligomer protein is labeled according to the concentration of A ⁇ monomer protein before incubation.
  • Cytotoxicity assay N2a cells were seeded into 96-well plates at 2x10 4 cells per well. After the cells were attached for 12 hours, they were replaced with serum-free DMEM medium, and the cells were starved for 24 hours.
  • the A ⁇ 1-42 oligomer was diluted to a 20 ⁇ M solution in serum-free DMEM.
  • the negative control group was treated with serum-free DMEM, and the positive control group was added with 20 ⁇ M A ⁇ protein solution.
  • the experimental group contained 10 ⁇ M and 20 ⁇ M sDSS1 protein in addition to 20 ⁇ M A ⁇ protein.
  • a ⁇ was diluted with serum-free DMEM according to the N2a cell experiment and the same sDSS1 protein gradient was set. Neurons were processed for 48 hours. The supernatant of the cells in the 96-well plate was collected, centrifuged at 100 g for 10 minutes, and the supernatant was used for cytotoxicity detection; the cells were used for cell viability assay.
  • the lactate dehydrogenase cytotoxicity test kit was purchased from Biyuntian Biotechnology Co., Ltd. (C0016). When detecting the enzyme activity, first configure a sufficient amount of the working fluid according to the kit instructions. The supernatant collected in a 120 ⁇ L cytotoxicity experiment was added to each well of a 96-well plate, then 60 ⁇ L of the test working solution was added, mixed slightly, and incubated at 37 ° C for 30 minutes. The absorbance at 490 nm was measured on a microplate reader, and the absorbance was directly proportional to the cytotoxicity level.
  • the cell proliferation/cytotoxicity assay kit (CCK-8) was purchased from Dongren Chemical Technology (Shanghai) Co., Ltd. (CK04). The working solution was prepared by diluting the CCK-8 solution with 1:20 (volume ratio, v/v) in serum-free DMEM. After completion of the cell treatment in the 96 wells, the old medium was discarded, and 100 ⁇ l of CCK-8 working solution was added to each well. The plate was incubated in an incubator for 1-2 hours, and then the 450 nm absorbance value was measured on a multi-function microplate reader to reflect the cell viability.
  • sDSS1 protein can significantly block the decrease of cell viability caused by A ⁇ 1-42 oligomer, and 20 ⁇ M sDSS1 protein can protect cells from A ⁇ 1-42 oligomer (Fig. 4B). ).
  • the cytotoxicity level was measured by the LDH kit, and the results showed that the cytotoxicity level of the sDSS1 protein group was significantly decreased (Fig. 4C).
  • the sDSS1 protein On mouse primary neuronal cells, the sDSS1 protein also showed a shielding effect on the toxicity of A ⁇ oligomers, the cell viability of neurons was protected by sDSS1 protein (Fig. 5A), and the level of cytotoxicity was decreased (Fig. 5B).
  • Example 5 The sDSS1 protein promotes microglia clearance of A[beta] protein and reduces microglial activation.
  • BV2 Cell Culture Mouse microglia
  • DMEM medium containing 10% FBS
  • a ⁇ clearance assay BV2 cells were seeded into 6-well plates at 3x10 5 per well and cells were adhered for 12 hours.
  • the A ⁇ protein was previously diluted with DMEM complete medium to a 1 ⁇ M solution.
  • the negative control group was not added with protein, and the positive control group was added with 1 ⁇ M A ⁇ protein solution.
  • the experimental group contained 0.5 ⁇ M and 2 ⁇ M sDSS1 protein except 1 ⁇ M A ⁇ protein. After 24 hours of treatment, 500 ⁇ L of the supernatant was collected, centrifuged at 100 g for 10 minutes, and the supernatant was taken for ELISA assay to detect A ⁇ 1-42 levels.
  • BV2 cells were seeded per well in accordance 1.5x10 5 to 6 well plates, DMEM medium was changed to serum-free 12 hours, cells were starved for 24 hours.
  • the A ⁇ 1-42 oligomer was diluted to a 20 ⁇ M solution in serum-free DMEM.
  • the negative control group was not added with protein, and the positive control group was added with 20 ⁇ M A ⁇ protein solution.
  • the experimental group contained 10 ⁇ M and 20 ⁇ M sDSS1 protein except 20 ⁇ M A ⁇ protein.
  • the supernatant was collected, centrifuged at 100 g for 10 minutes, and the supernatant was taken for ELISA assay to detect the TNF ⁇ level of the solution. All cells were collected for ROS level detection.
  • ELISA assay The culture solution obtained in the above A ⁇ scavenging experiment was used in an ELISA experiment, and the culture solution sample was diluted 50 times with a standard diluent buffer provided by the kit for subsequent detection. According to the instructions of ELISA kit (Invitrogen, Human A ⁇ 42 ELISA kit, KHB3441), the standard protein and the nematode protein to be tested were sequentially incubated and washed, and the chromogenic substrate was added for 20 minutes. The stop solution was added and detected on the microplate reader. 450 nm absorbance value. The A ⁇ protein concentration in the protein sample was calculated from the standard curve.
  • ROS level detection Active oxygen detection kit was purchased from Biyuntian Biotechnology Co., Ltd. (S0033). Positive control cells were pretreated with 1 ⁇ M Rosup for 20 minutes at 37 °C. 300 ⁇ L of the cell suspension was taken, centrifuged at 100 g for 5 minutes, and the cells were washed twice with PBS. 300 ⁇ L of serum-free DMEM-diluted fluorescent probe (DCFDA) (1:1000) was added and incubated at 37 ° C for 20 minutes. After centrifugation at 100 g for 5 minutes, the cells were washed three times with PBS and detected by flow cytometry (Millipore, guava Easycyte). Green fluorescence (488 nm) intensity reflects cellular ROS levels.
  • DCFDA serum-free DMEM-diluted fluorescent probe
  • TNF ⁇ level The supernatant collected in the BV2 cell activation experiment was diluted 10-fold with Standard Dilute buffer provided in the kit for ELISA detection. According to the instructions of ELISA kit (Invitrogen, TNF alpha Mouse ELISA Kit, KMC3011C), the standard protein and the nematode protein to be tested were sequentially incubated and washed, incubated with chromogenic substrate for 20 minutes, and the stop solution was added to the microplate reader. The 450 nm absorbance value was measured. The concentration of TNF ⁇ in the supernatant was calculated from the standard curve.
  • Microglia are the main cells in nerve tissue that remove toxic proteins such as A ⁇ protein.
  • 1 ⁇ M A ⁇ 1-42 protein and different concentrations of sDSS1 protein were added to cultured mouse microglia.
  • the results showed that A ⁇ protein was detected in the cytoplasm of microglia, and the concentration of cytosolic A ⁇ protein was significantly increased with the addition of sDSS1 protein (Fig. 6A).
  • the A ⁇ protein residue in the culture solution decreased, and the A ⁇ protein concentration became lower and lower as the concentration of sDSS1 protein in the solution increased (Fig. 6B).
  • sDSS1 protein in the culture medium can significantly reduce the number of activated microglia, and the cell ROS level is significantly reduced (Fig. 6C).
  • the release of the inflammatory factor TNF- ⁇ gradually decreased as the concentration of sDSS1 protein increased (Fig. 6D).
  • Example 6 The sDSS1 protein reduces disease symptoms in the AD model nematode.
  • AD model nematode detection The nematode experimental platform was provided by Shanghai Southern Model Biotechnology Co., Ltd., and the AD nematode model was provided by the nematode experimental platform.
  • the adult CL4176 nematode was picked into a solid medium and placed in a 16 ° C incubator. After 2 hours of culture, all the adults were picked and the eggs were placed in a 16 ° C incubator. After 48 hours, the culture dishes were transferred to a 25 ° C incubator for further culture and treated with different concentrations of sDSS1 protein, 0 ⁇ g/ml in the control group, 500 ⁇ g/ml in the low dose group, and 1000 ⁇ g/ml in the high dose group.
  • the standard for nematodes is that when the wire is lightly nematode, the nematode is stiff and incapable of moving.
  • elegans lysate containing 62 mM Tris-HCl, 2% SDS, 10% glycerol, 4% ⁇ -mercaptoethanol, cocktail protease inhibition) Agent
  • liquid nitrogen freeze-thaw 2 times 60Hz ultrasound for 5 minutes, centrifugation at 12000rpm for 10 minutes, take the supernatant is the total protein of nematodes.
  • the nematode protein was added to a loading buffer and heat treated at 100 ° C for 10 minutes to prepare a protein sample for Wetern blotting analysis.
  • the completed membrane was first blocked with 5% skim milk powder (dissolved in PBST) for 1 hour at room temperature, then sequentially incubated with rabbit-derived A ⁇ antibody dilution at 4 ° C overnight (1:3000 dilution in blocking solution), washed three times with PBST, HRP coupling The conjugated anti-rabbit antibody dilution was incubated for 1 hour at room temperature (1:5000 dilution into PBST). After the ECL color is developed, the tablet is compressed and photographed and saved.
  • the internal reference used tubulin protein (antibody was purchased from Biyuntian Biotechnology Co., Ltd., AF0001).
  • the total nematode protein extracted from the above Western blotting experiment was used in the ELISA experiment, and the protein sample was diluted three times with the standard diluent buffer provided by the kit for subsequent detection, and the protein dilution was added. The volume was adjusted according to the data of the internal reference in the Western blotting experiment to ensure that the total amount of protein added was consistent. According to the instructions of the ELISA kit, the standard protein and the nematode protein to be tested were sequentially incubated and washed, incubated with a chromogenic substrate for 20 minutes, and the stop solution was added to measure the absorbance at 450 nm on a microplate reader. The A ⁇ protein concentration in the protein sample was calculated from the standard curve.
  • C. elegans immunohistochemical staining CL4176 nematodes were treated in different concentrations of drugs for 20 hours, and after completion, nematodes were collected, with 50-100 nematodes in each group. The nematodes were washed once with PBS and placed in a 1.5 mL centrifuge tube, and the supernatant was discarded, and 4% paraformaldehyde (dissolved in PBS, pH 7.2) was added at 4 ° C overnight. The fixed nematode was first rinsed once with wash buffer (containing 10 mm Tris/HCl, 1% Triton-X100, 1 mM EDTA, pH 7.4).
  • wash buffer containing 10 mm Tris/HCl, 1% Triton-X100, 1 mM EDTA, pH 7.4
  • the nematodes were sequentially incubated with a washing solution containing 1% ⁇ -mercaptoethanol for 1 hour at room temperature, boric acid buffer (containing 25 mM H 3 BO 3 , 12.5 mM NaOH, 10 mM DTT, pH 9.2) was incubated for 1 minute at room temperature for 5 minutes. 1% H 2 O 2 wash buffer was incubated for 15 minutes at room temperature. After completion, the nematodes were washed once with wash buffer and blocked by adding blocking solution (containing 1% BSA, 0.5% Triton-X100, 1 mM EDTA, PBS) for two hours at room temperature.
  • blocking solution containing 1% BSA, 0.5% Triton-X100, 1 mM EDTA, PBS
  • the treated nematodes were sequentially incubated overnight at 4 °C with a primary antibody dilution (A ⁇ antibody, diluted 1:100 in blocking solution), and the secondary antibody dilution (added Hoechst at a ratio of 1:1000) (Donkey-anti-Rabbit-Alex Fluor) 488 (purchased from Life Technology, Inc., A-11008) was incubated for 1 hour at room temperature with a dilution of 1:5000 in PBST. The PBST was washed three times, and after completion, it was mounted and observed with a Confocol laser confocal microscope (Nikon, Nikon C1Plus), and photographed.
  • CL4176 nematode was used as an animal model for AD to detect toxicity caused by accumulation of acute A ⁇ 1-42 protein and was used for drug evaluation.
  • the addition of different concentrations of sDSS1 protein significantly reduced the nematode sputum level, and the effect showed a significant concentration dependence (Fig. 7A).
  • the proportion of the non-nematode in the control group was less than 1%, while the nematodes with 18.42% (500 ⁇ g/mL sDSS1 protein) and 48.57% (1000 ⁇ g/mL sDSS1 protein) in the administration group were normal.
  • the median time of occurrence of sputum in half of the nematodes was 22 hours in the control group, 24 hours in the low concentration group, and 42 hours in the high concentration group.
  • sDSS1 protein reduced the content of A ⁇ protein multimer (Fig. 7C).
  • the A ⁇ 1-42 protein content in the tissue lysate was detected by ELISA. It can be seen that the A ⁇ protein in the sDSS1 protein-treated nematode was decreased, and the A ⁇ protein content in the 1000 ⁇ g/mL sDSS1 protein treatment group was only 59.09% of the control group (Fig. 7D). .
  • sDSS1 protein can reduce the formation of A ⁇ 1-42 aggregates, reduce the accumulation of tissue A ⁇ , and alleviate the disease symptoms of AD model nematodes in the AD nematode model.
  • Example 7 The sDSS1 protein accelerates clearance of A ⁇ protein in mice.
  • APP/PS1 mouse blood A ⁇ clearance test APP/PS1 mice (2 months old, male) were purchased from the Model Animal Center of Nanjing University. The APP/PS1 mice were injected with sDSS1 protein at a dose of 25 mg/kg body weight for 8 weeks from the age of 8 months. After completion, the mice were sacrificed, blood was taken and centrifuged to obtain plasma for ELISA to detect A ⁇ 1-42 levels in the blood.
  • a ⁇ protein acute clearance test ICR mice (6-8 weeks old, male) were purchased from Shanghai Slack Laboratory Animals Co., Ltd. The mice were divided into four groups according to their body weight, including a negative control group (injected with normal saline), a positive control group (administered with A ⁇ protein alone), a low-dose group (A ⁇ -administered and 5 mg/kg body weight injected with sDSS1 protein), and high dose.
  • the administration group (A ⁇ administration and 30 mg/kg body weight injection of sDSS1 protein).
  • the mice were first injected with sDSS1 protein according to the tail vein of the group.
  • the negative control group and the positive control group were injected with the same amount of PBS.
  • the positive control group and the drug-administered group were injected with A ⁇ protein at the tail vein of 10 mg/kg body weight.
  • the control group was injected with the same amount of normal saline. After the injection was completed, the mice were normally reared, and the mice were sacrificed 2 hours later. Blood was taken from the heart, and the cells were centrifuged at 3,500 g for 20 minutes to obtain plasma for ELISA to detect A? 1-42 levels.
  • ELISA test Plasma samples obtained in the above experiment were used for the ELISA experiment. According to the instructions of the ELISA kit, the standard protein and the plasma sample to be tested are sequentially incubated and washed, and the chromogenic substrate is added for 20 minutes, the stop solution is added, and the 450 nm absorbance value is detected on the microplate reader. The A ⁇ protein concentration in plasma was calculated from the standard curve.
  • sDSS1 protein can help clear A ⁇ protein in the blood of mice.
  • high concentrations of A ⁇ 1-42 protein were injected through the tail vein while the sDSS1 protein was injected.
  • the results of the experiment showed that different concentrations of sDSS1 protein administration significantly reduced blood A ⁇ protein levels, and the effect was dose-dependent (Fig. 8A).
  • AD model APP/PS1 mice were injected with sDSS1 protein for a long time in the tail vein, and it was found that sDSS1 protein also reduced blood A ⁇ protein levels (Fig. 8B).
  • Example 8 sDSS1 protein reduces plaque deposition in brain tissue of AD model mice.
  • AD model mice administration APP/PS1 mice (2 months old, male) were purchased from the Model Animal Center of Nanjing University. The APP/PS1 mice were injected with sDSS1 protein at a dose of 50 mg/kg body weight for 8 weeks from the age of 9 months. After completion, the mice were used for behavioral experiments. After the behavioral experiment was completed, the mice were sacrificed, and plasma was taken for blood index detection. Brain tissue was taken and fixed in 4% paraformaldehyde for immunohistochemical staining to analyze plaque deposition levels.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Uses of a protein in preparing a drug for preventing and treating Alzheimer's disease, said protein being an sDSS1 protein used in preparing a drug for preventing neurodegenerative and vascular dementias, or a drug for treating said diseases in the prophase, early, middle and later stages. Said sDSS1 protein interacts with an amyloid β-protein (Aβ) to contribute to Aβ removal while protecting against Aβ toxicity. Moreover, the Aβ deposition is effectively reduced in animals, and the concentration of a soluble Aβ is reduced, leading to disease symptom relief.

Description

一种蛋白在制备预防和治疗痴呆症的药物中的应用Application of a protein in preparing medicine for preventing and treating dementia 技术领域Technical field
本发明内容涉及sDSS1蛋白药物在制备预防和治疗痴呆症的药物中的应用,包括用于制备退行性痴呆症和血管性痴呆症的预防药物,或用于制备这些疾病前期、早期、中期或晚期的治疗药物。The present invention relates to the use of the sDSS1 protein drug for the preparation of a medicament for preventing and treating dementia, comprising a prophylactic medicament for the preparation of degenerative dementia and vascular dementia, or for preparing a pre-, early, intermediate or late stage of these diseases. Therapeutic drugs.
背景技术Background technique
痴呆症(Dementia)是是脑部疾病的一类,此症导致患者思维能力和学习记忆力长期而逐渐地退化,最常见的痴呆症是阿尔兹海默病(Alzheimer’s Disease,AD),占痴呆症患者的50-70%,其他还包括血管性痴呆症(
Figure PCTCN2018120172-appb-000001
25%)、路易氏体型痴呆症(
Figure PCTCN2018120172-appb-000002
15%)等。国际阿尔茨海默症协会(Alzheimer’s Disease International)2015年公布的数据显示,全球痴呆症人数大约4680万人,2030年全球患者预计将达到7500万[1]。AD是一种典型的神经退行性疾病,临床表现为进行性学习记忆功能降低和神经系统损伤。AD患者典型病理特征是神经组织中出现β-淀粉样蛋白(β-amyloid,Aβ)斑块和tau蛋白沉积。β淀粉样蛋白假说认为神经组织中Aβ蛋白产生和清除失衡导致的毒性蛋白进行性堆积及由此引起的突触功能紊乱、神经元死亡是导致疾病的关键机制[2][3]。基于β淀粉样蛋白假说,研究人员希望通过药物抑制Aβ蛋白的生成和聚集,或者促进Aβ清除,从而达到预防或治疗疾病的目的[4]。基于动物实验的AD研究结果表明,抗体[5][6]、多肽类药物[7]、小分子类药物[8]可以阻断Aβ聚集或生成,减少斑块形成,减轻动物疾病指征。因此,药物抑制并清除Aβ病理性沉积或可成为治疗AD的有效途径。
Dementia is a type of brain disease that causes long-term and progressive deterioration of patient thinking and learning memory. The most common form of dementia is Alzheimer's Disease (AD), which accounts for dementia. 50-70% of patients, others include vascular dementia (
Figure PCTCN2018120172-appb-000001
25%), Louise-type dementia (
Figure PCTCN2018120172-appb-000002
15%) and so on. According to data released by Alzheimer's Disease International in 2015, the number of people with dementia worldwide is about 46.8 million, and the number of patients worldwide is expected to reach 75 million in 2030 [1]. AD is a typical neurodegenerative disease with clinical manifestations of progressive learning and memory impairment and neurological damage. A typical pathological feature of AD patients is the presence of β-amyloid (Aβ) plaques and tau deposition in nerve tissue. The β amyloid hypothesis suggests that the progressive accumulation of toxic proteins caused by the imbalance of Aβ protein production and clearance in nerve tissue and the resulting synaptic dysfunction and neuronal death are key mechanisms leading to disease [2] [3]. Based on the beta amyloid hypothesis, the researchers hope to prevent Aβ protein production and aggregation, or promote Aβ clearance, in order to prevent or treat diseases [4]. The results of AD studies based on animal experiments show that antibodies [5] [6], peptide drugs [7], small molecule drugs [8] can block Aβ aggregation or production, reduce plaque formation, and reduce animal disease indications. Therefore, the drug inhibits and clears the pathological deposition of Aβ or can be an effective way to treat AD.
过去十几年来,包括礼来、阿斯利康、强生、辉瑞、罗氏在内的数十家医药企业投入数百亿美元研发AD治疗药物,然而进展并不顺 利,可见AD治疗药物开发的困难性与复杂性。礼来公司(Lily)以Aβ蛋白为靶点单抗药物solanezumab,在III期临床实验中显示能够减缓34%中期AD患者的认知能力下降以及18%患者的行为能力下降,但是受广泛的副作用、综合效应评价不具有统计学差异等因素影响,该药物于2016年12月宣布失败。百健(Biogen)2018年7月公布的Ib期临床实验研究表明,针对Aβ蛋白的实验性单抗药物aducanumab能够减少患者大脑中的β淀粉样蛋白,并且能够显著改善患者的认知水平。百健和卫材(Eisai)2018年11月公布的针对Aβ蛋白的另一个单抗BAN2401的Ⅱ期临床实验也取得显著的临床效果。此外,2017-2018年,国际药业巨头,如罗氏、礼来、阿斯利康、默沙东各自宣布了针对AD疾病的新药开发计划,由此可见医药产业界对AD疾病的重点关注。In the past ten years, dozens of pharmaceutical companies, including Lilly, AstraZeneca, Johnson & Johnson, Pfizer, and Roche, have invested tens of billions of dollars in the development of AD treatments. However, the progress has not been smooth, and the difficulty in developing AD treatment drugs can be seen. With complexity. Lily uses the Aβ protein as a target monoclonal antibody, solanezumab, which has been shown to slow down cognitive decline in 34% of patients with mid-term AD and decline in behavioral capacity in 18% of patients in phase III clinical trials, but is subject to extensive side effects. The comprehensive effect evaluation was not affected by statistical differences, and the drug was declared a failure in December 2016. Biogen's Phase Ib clinical trial published in July 2018 showed that the aducanumab, an experimental monoclonal antibody against Aβ protein, can reduce beta amyloid in the brain of patients and significantly improve the cognitive level of patients. Phase II clinical trials of BAN2401, another monoclonal antibody against Aβ protein, released by Baishen and Eisai in November 2018, also achieved significant clinical results. In addition, in 2017-2018, international pharmaceutical giants such as Roche, Eli Lilly, AstraZeneca, and Merck have announced new drug development plans for AD diseases, which shows that the pharmaceutical industry is focusing on AD diseases.
Shfm1(split hand/split foot malformation type 1)基因是人蟹爪病中的关键基因之一,进化上高度保守,其编码的蛋白DSS1参与到稳定基因组、同源基因重组、DNA损伤修复和细胞增殖等过程[9-12]。本专利发明人的研究结果显示DSS1蛋白作为标签可以通过耗能的酶促反应添加到氧化蛋白上,帮助细胞清除氧化蛋白[13]。这些结果显示DSS1蛋白在生物活动中的重要作用。Shfm1 (split hand/split foot malformation type 1) gene is one of the key genes in human crab claw disease. It is highly conserved in evolution and its encoded protein DSS1 is involved in stable genome, homologous gene recombination, DNA damage repair and cell proliferation. Etc. [9-12]. The results of the inventors of the present invention show that the DSS1 protein can be added to the oxidized protein by a energy-consuming enzymatic reaction to help the cells clear the oxidized protein [13]. These results show the important role of DSS1 protein in biological activities.
以上内容的引文如下:The citations for the above are as follows:
1.Alzheimer’s Disease International(2016)World Alzheimer Report 2015:the global impact of dementia,an analysis of prevalence,incidence,cost and trends.1. Alzheimer’s Disease International (2016) World Alzheimer Report 2015: the global impact of dementia, an analysis of prevalence,incidence, cost and trends.
2.Gupta A,Iadecola C(2015)Impaired Aβclearance:a potential link between atherosclerosis and Alzheimer's disease.Front Aging Neurosci 7:115.2. Gupta A, Iadecola C (2015) Impaired Aβclearance: a potential link between atherosclerosis and Alzheimer's disease. Front Aging Neurosci 7:115.
3.Bu XL,Xiang Y,Jin WS,Wang J,Shen LL,Huang ZL,Zhang K,Liu YH,Zeng F,Liu JH,Sun HL,Zhuang ZQ,Chen SH,Yao XQ, Giunta B,Shan YC,Tan J,Chen XW,Dong ZF,Zhou HD,Zhou XF,Song W,Wang YJ(2017)Blood-derived amyloid-β protein induces Alzheimer's disease pathologies.Mol Psychiatry.[Epub ahead of print]3.Bu XL, Xiang Y, Jin WS, Wang J, Shen LL, Huang ZL, Zhang K, Liu YH, Zeng F, Liu JH, Sun HL, Zhuang ZQ, Chen SH, Yao XQ, Giunta B, Shan YC, Tan J, Chen XW, Dong ZF, Zhou HD, Zhou XF, Song W, Wang YJ (2017) Blood-derived amyloid-β protein induces Alzheimer's disease pathologies. Mol Psychiatry. [Epub ahead of print]
4.Citron M.(2010)Alzheimer's disease:strategies for disease modification.Nat Rev Drug Discov 9(5):387-98.4. Citron M. (2010) Alzheimer's disease: strategies for disease modification. Nat Rev Drug Discov 9(5): 387-98.
5.Winblad B,Andreasen N,Minthon L,Floesser A,Imbert G,Dumortier T,Maguire RP,Blennow K,Lundmark J,Staufenbiel M,Orgogozo JM,Graf A(2012)Safety,tolerability,and antibody response of active Aβ immunotherapy with CAD106 in patients with Alzheimer's disease:randomised,double-blind,placebo-controlled,first-in-human study.Lancet Neurol 11(7):597-604.5.Winblad B, Andreasen N, Minthon L, Floesser A, Imbert G, Dumortier T, Maguire RP, Blennow K, Lundmark J, Staufenbiel M, Orgogozo JM, Graf A (2012) Safety, tolerability, and antibody response of active Aβ Immunotherapy with CAD106 in patients with Alzheimer's disease: randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol 11(7): 597-604.
6.Sevigny J,Chiao P,Bussière T,Weinreb PH,Williams L,Maier M,Dunstan R,Salloway S,Chen T,Ling Y,O'Gorman J,Qian F,Arastu M,Li M1,Chollate S,Brennan MS,Quintero-Monzon O,Scannevin RH,Arnold HM,Engber T,Rhodes K,Ferrero J,Hang Y,Mikulskis A,Grimm J,Hock C,Nitsch RM,Sandrock A.The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease.Nature 537:50–56.6. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, O'Gorman J, Qian F, Arastu M, Li M1, Chollate S, Brennan MS, Quintero-Monzon O, Scannevin RH, Arnold HM, Engber T, Rhodes K, Ferrero J, Hang Y, Mikulskis A, Grimm J, Hock C, Nitsch RM, Sandrock A. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature 537: 50–56.
7.Chang L,Cui W,Yang Y,Xu S,Zhou W,Fu H,Hu S,Mak S,Hu J,Wang Q,Ma VP,Choi TC,Ma ED,Tao L,Pang Y,Rowan MJ,Anwyl R,Han Y,Wang Q(2015)Protection against β-amyloid-induced synaptic and memory impairments via altering β-amyloid assembly by bis(heptyl)-cognitin.Sci Rep 5:10256.7.Chang L, Cui W, Yang Y, Xu S, Zhou W, Fu H, Hu S, Mak S, Hu J, Wang Q, Ma VP, Choi TC, Ma ED, Tao L, Pang Y, Rowan MJ, Anwyl R, Han Y, Wang Q (2015) Protection against β-amyloid-induced synaptic and memory impairments via altering β-amyloid assembly by bis(heptyl)-cognitin.Sci Rep 5:10256.
8.Kim HY,Kim HV,Jo S,Lee CJ,Choi SY,Kim DJ,Kim Y(2015)EPPS rescues hippocampus-dependent cognitive deficits in APP/PS1 mice by disaggregation of amyloid-β oligomers and plaques.Nat Commun 6:8997.8.Kim HY, Kim HV, Jo S, Lee CJ, Choi SY, Kim DJ, Kim Y (2015) EPPS rescues hippocampus-dependent cognitive deficits in APP/PS1 mice by disaggregation of amyloid-β oligomers and plaques.Nat Commun 6 :8997.
9.Van Silfhout AT,van den Akker PC,Dijkhuizen T,Verheij JB,Olderode-Berends MJ,Kok K,Sikkema-Raddatz B,van Ravenswaaij-Arts CM(2009)Split hand/foot malformation due to chromosome 7q aberrations(SHFM1):additional support for functional haploinsufficiency as the causative mechanism.Eur J Hum Genet 17(11):1432-8.9.Van Silfhout AT, van den Akker PC, Dijkhuizen T, Verheij JB, Olderode-Berends MJ, Kok K, Sikkema-Raddatz B, van Ravenswaaij-Arts CM (2009) Split hand/foot malformation due to chromosome 7q aberrations (SHFM1 ): additional support for functional haploinsufficiency as the causative mechanism. Eur J Hum Genet 17(11): 1432-8.
10.Li J,Zou C,Bai Y,Wazer DE,Band V,Gao Q(2006)DSS1 is required for the stability of BRCA2.Oncogene 25:1186–1194.10.Li J,Zou C,Bai Y,Wazer DE,Band V,Gao Q(2006)DSS1 is required for the stability of BRCA2.Oncogene 25:1186–1194.
11.Liu J,Doty T,Gibson B,Heyer WD(2010)Human BRCA2 protein promotes RAD51 filament formation on RPA-covered singlestranded DNA.Nat Struct Mol Biol 17:1260–1262.11. Liu J, Doty T, Gibson B, Heyer WD (2010) Human BRCA2 protein promotes RAD51 filament formation on RPA-covered single stranded DNA. Nat Struct Mol Biol 17: 1260–1262.
12.Zhou Q,Kojic M,Cao Z,Lisby M,Mazloum NA,Holloman WK(2007)Dss1 interaction with Brh2 as a regulatory mechanism for recombinational repair.Mol Cell Biol 2:2512–2526.12.Zhou Q, Kojic M, Cao Z, Lisby M, Mazloum NA, Holloman WK (2007) Dss1 interaction with Brh2 as a regulatory mechanism for recombinational repair. Mol Cell Biol 2:2512–2526.
13.Zhang Y,Chang FM,Huang J,Junco JJ,Maffi SK,Pridgen HI,Catano G,Dang H,Ding X,Yang F,Kim DJ,Slaga TJ,He R,Wei SJ(2014)DSSylation,a novel protein modification targets proteins induced by oxidative stress,and facilitates their degradation in cells.Protein Cell 5(2):124-40.13.Zhang Y, Chang FM, Huang J, Junco JJ, Maffi SK, Pridgen HI, Catano G, Dang H, Ding X, Yang F, Kim DJ, Slaga TJ, He R, Wei SJ (2014) DSSylation, a novel Protein modification targets proteins induced by oxidative stress, and facilitates their degradation in cells. Protein Cell 5(2): 124-40.
发明内容Summary of the invention
Aβ蛋白过渡蓄积是AD发病的关键因素之一,通过药物清除Aβ或抑制Aβ聚集是治疗AD疾病的主要途径之一。本发明提供的sDSS1蛋白可以与Aβ蛋白结合从而加速Aβ蛋白清除,减少Aβ蛋白过渡蓄积,改善疾病症状,具备用于制备临床上AD及其他痴呆症预防药物和治疗药物的潜力。The transient accumulation of Aβ protein is one of the key factors in the pathogenesis of AD. It is one of the main ways to treat AD diseases by removing Aβ or inhibiting Aβ aggregation. The sDSS1 protein provided by the invention can bind to Aβ protein to accelerate the clearance of Aβ protein, reduce the accumulation of Aβ protein, improve the symptoms of the disease, and has the potential for preparing clinical AD and other dementia prevention drugs and therapeutic drugs.
具体的技术方案如下:The specific technical solutions are as follows:
一种蛋白在制备预防和治疗痴呆症的药物中的应用,所述的应用是把sDSS1蛋白用于制备痴呆症预防药物和治疗药物。The use of a protein for the preparation of a medicament for the prevention and treatment of dementia, which is the use of the sDSS1 protein for the preparation of a medicament for the prevention and treatment of dementia.
优选地,所述的痴呆症是指退行性痴呆症,包括早老性痴呆症、老年性痴呆症、路易氏体型痴呆症、额颞叶型痴呆症。Preferably, the dementia refers to degenerative dementia, including Alzheimer's disease, Alzheimer's disease, Lewy body dementia, and frontotemporal dementia.
优选地,所述的痴呆症是血管性痴呆症。Preferably, the dementia is vascular dementia.
优选地,所述的痴呆症是痴呆前期痴呆症、轻度痴呆期痴呆症、中度痴呆期痴呆症、重度痴呆期痴呆症。Preferably, the dementia is pre-dementia dementia, mild dementia dementia, moderate dementia dementia, and severe dementia dementia.
优选地,所述的sDSS1蛋白包括人、黑猩猩、倭黑猩猩、大猩猩、红毛猩猩、白颊长臂猿、川金丝猴、恒河猴、滇金丝猴、东非狒狒、安哥拉疣猴、白顶白眉猴、鬼狒、豚尾猴中的任一sDSS1蛋白序列形成的基础蛋白,其中人sDSS1的氨基酸序列如SEQ ID NO:1,黑猩猩sDSS1的氨基酸序列如SEQ ID NO:2,倭黑猩猩sDSS1的氨基酸序列如SEQ ID NO:3,大猩猩sDSS1的氨基酸序列如SEQ ID NO:4,红毛猩猩sDSS1的氨基酸序列如SEQ ID NO:5,白颊长臂猿sDSS1的氨基酸序列如SEQ ID NO:6,川金丝猴sDSS1的氨基酸序列如SEQ ID NO:7,恒河猴sDSS1的氨基酸序列如SEQ ID NO:8,滇金丝猴sDSS1的氨基酸序列如SEQ ID NO:9,东非狒狒sDSS1的氨基酸序列如SEQ ID NO:10,安哥拉疣猴sDSS1的氨基酸序列如SEQ ID NO:11,白顶白眉猴sDSS1的氨基酸序列如SEQ ID NO:12,鬼狒sDSS1的氨基酸序列如SEQ ID NO:13,豚尾猴sDSS1的氨基酸序列如SEQ ID NO:14。Preferably, the sDSS1 protein comprises human, chimpanzee, bonobo, gorilla, orangutan, white-cheeked gibbons, golden monkey, rhesus monkey, golden monkey, East African pheasant, Angora simian, white-tailed white-browed monkey, ghost A basic protein formed by any sDSS1 protein sequence in scorpionfish, porpoise monkey, wherein the amino acid sequence of human sDSS1 is SEQ ID NO: 1, the amino acid sequence of chimpanzee sDSS1 is SEQ ID NO: 2, and the amino acid sequence of porcine chimpanzee sDSS1 is SEQ. ID NO: 3, the amino acid sequence of gorilla sDSS1 is SEQ ID NO: 4, the amino acid sequence of orangutan sDSS1 is SEQ ID NO: 5, the amino acid sequence of white buccal gibbon sDSS1 is SEQ ID NO: 6, and the golden monkey sDSS1 The amino acid sequence of SEQ ID NO: 7, the amino acid sequence of rhesus sDSS1 is SEQ ID NO: 8, the amino acid sequence of gilt monkey sDSS1 is SEQ ID NO: 9, and the amino acid sequence of ssDSS1 is SEQ ID NO: 10, Angola. The amino acid sequence of simian sDSS1 is SEQ ID NO: 11, the amino acid sequence of leucocephalus sDSS1 is SEQ ID NO: 12, and the amino acid sequence of scorpion sDSS1 is SEQ ID NO: 13. M.nemestrina sDSS1 amino acid sequence as SEQ ID NO: 14.
优选地,所述的sDSS1蛋白是与所述的基础蛋白相似度达到70%以上的第一种蛋白。Preferably, the sDSS1 protein is the first protein having a similarity to the basic protein of 70% or more.
优选地,所述的sDSS1蛋白是以所述的基础蛋白的氮端58个氨基酸为基础,在氮端或碳端融合其他多肽片段,用于融合的多肽片段的结构特征或氨基酸序列特征与所述的基础蛋白的碳端31个序列相同或相似的第二种蛋白。Preferably, the sDSS1 protein is based on 58 amino acids of the nitrogen terminus of the basic protein, and fuses other polypeptide fragments at the nitrogen terminal or the carbon terminal, and the structural features or amino acid sequence characteristics of the polypeptide fragment for fusion are The second protein of the same or similar sequence of the carbon end of the basic protein.
优选地,所述的sDSS1蛋白是以所述的基础蛋白的氮端58个氨基酸为基础,在氮端或碳端融合其他氨基酸片段,融合后的蛋白能实现跨膜转运功能的第三种蛋白。Preferably, the sDSS1 protein is based on the basic amino acid of the basic protein of 58 amino acids, and the other amino acid fragments are fused at the nitrogen or carbon end, and the fused protein can realize the transmembrane transport function. .
优选地,所述的sDSS1蛋白是利用所述的基础蛋白、第一种蛋白、第二种蛋白或第三种蛋白与该种蛋白自身、载体蛋白、抗体或其他任意长度氨基酸片段连接形成的融合蛋白。Preferably, the sDSS1 protein is a fusion formed by using the basic protein, the first protein, the second protein or the third protein to form a protein, a carrier protein, an antibody or other amino acid fragments of any length. protein.
优选地,所述的sDSS1蛋白是所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白进行的修饰产生的多肽/蛋白修饰物。Preferably, the sDSS1 protein is a polypeptide/protein modification produced by modification of the basic protein, the first protein, the second protein, the third protein or the fusion protein.
优选地,所述多肽/蛋白修饰物是针对氨基酸侧链上的氨基、氨基酸侧链上的羰基、氮端末端氨基、碳端末端羰基、半胱氨酸、酪氨酸、丝氨酸、色氨酸进行的特异性或非特异性的1-20个位点的化学修饰。Preferably, the polypeptide/protein modification is directed to an amino group on an amino acid side chain, a carbonyl group on an amino acid side chain, a nitrogen terminal amino group, a carbon terminal carbonyl group, a cysteine, a tyrosine, a serine, a tryptophan. Specific or non-specific chemical modification of 1-20 sites.
优选地,所述多肽/蛋白修饰物的修饰方法包括糖基化修饰、脂肪酸修饰、酰基化修饰、Fc片段融合、白蛋白融合、聚乙二醇修饰、右旋糖苷修饰、肝素修饰、聚乙烯吡咯烷酮修饰、聚氨基酸修饰、多聚唾液酸修饰、壳聚糖及其衍生物修饰、凝集素修饰、海藻酸钠修饰、卡波姆修饰、聚乙烯吡咯烷酮修饰、羟丙基甲基纤维素修饰、羟丙基纤维素修饰、乙酰化修饰、甲酰化修饰、磷酸化修饰、甲基化修饰、磺酸化修饰以及其他医药上可用的多肽/蛋白药物修饰方法的一种或一种以上。Preferably, the method for modifying the polypeptide/protein modification comprises glycosylation modification, fatty acid modification, acylation modification, Fc fragment fusion, albumin fusion, polyethylene glycol modification, dextran modification, heparin modification, polyethylene Pyrrolidone modification, polyamino acid modification, polysialic acid modification, chitosan and its derivative modification, lectin modification, sodium alginate modification, carbomer modification, polyvinylpyrrolidone modification, hydroxypropyl methylcellulose modification, One or more of hydroxypropylcellulose modification, acetylation modification, formylation modification, phosphorylation modification, methylation modification, sulfonation modification, and other pharmaceutically acceptable polypeptide/protein drug modification methods.
优选地,所述的sDSS1蛋白是利用所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白的氨基酸序列为基础进行的20种基本氨基酸以外的氨基酸进行的1-31个任意氨基酸位点替换的非天然氨基酸替代蛋白。Preferably, the sDSS1 protein is an amino acid other than the 20 basic amino acids based on the amino acid sequence of the basic protein, the first protein, the second protein, the third protein or the fusion protein. - 31 non-natural amino acid replacement proteins substituted with any amino acid site.
优选地,所述的非天然氨基酸替代蛋白的氨基酸替换包括替换成羟脯氨酸、羟赖氨酸、硒代半胱氨酸、D-型氨基酸或者人工合成的非天然氨基酸及其衍生物。Preferably, the amino acid substitution of the non-natural amino acid replacement protein comprises replacement with hydroxyproline, hydroxylysine, selenocysteine, D-form amino acid or synthetic non-natural amino acid and derivatives thereof.
优选地,所述的sDSS1蛋白是把所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或非天然氨基酸替代蛋白与医药上可应用的药物载体形成的部分或全部复合体。Preferably, the sDSS1 protein is pharmaceutically applicable to the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the non-natural amino acid replacement protein. Part or all of the complex formed by the drug carrier.
优选地,所述复合体的药物载体包含肠溶衣制剂、胶囊、微球/囊、脂质体、微乳液、复乳液、纳米颗粒、磁颗粒、明胶和凝胶中的一种或一种以上。Preferably, the pharmaceutical carrier of the complex comprises one or one of an enteric coating preparation, a capsule, a microsphere/capsule, a liposome, a microemulsion, a double emulsion, a nanoparticle, a magnetic particle, a gelatin, and a gel. the above.
优选地,所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或复合体发挥屏蔽毒性蛋白毒性的 起始工作浓度为不小于0.2μg/mLPreferably, the base of the protein, the first protein, second protein and third protein, fusion protein, polypeptide / protein or complex modifications play operation starting concentration of toxic proteins toxic shield is not less than 0.2μg /mL .
优选地,所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或复合体发挥屏蔽毒性蛋白毒性的摩尔浓度比率为不小于0.010;所述摩尔浓度比率是指反应体系中蛋白、多肽/蛋白修饰物或复合体的摩尔浓度与毒性蛋白摩尔浓度的比率。Preferably, the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the complex exhibits a molar concentration ratio of shielding toxic protein toxicity of not less than 0.010; The molar concentration ratio refers to the ratio of the molar concentration of the protein, polypeptide/protein modification or complex in the reaction system to the molar concentration of the toxic protein.
优选地,所述的sDSS1蛋白是以个体自身sDSS1蛋白为靶点,通过外源药物影响个体自身sDSS1蛋白的水平。Preferably, the sDSS1 protein targets the individual's own sDSS1 protein, and affects the level of the individual's own sDSS1 protein by the exogenous drug.
优选地,所述的药物是以sDSS1蛋白、sDSS1蛋白的基因、sDSS1的基因的调控元件或sDSS1的基因的转录产物为药物作用靶点。Preferably, the drug is a drug target of a sDSS1 protein, a gene of sDSS1 protein, a regulatory element of a gene of sDSS1 or a gene of sDSS1.
优选地,所述的药物是通过影响血液或脑脊液中蛋白酶/肽酶活性从而调节sDSS1蛋白在血液或脑脊液中的含量。Preferably, the drug modulates the amount of sDSS1 protein in the blood or cerebrospinal fluid by affecting protease/peptidase activity in blood or cerebrospinal fluid.
优选地,所述的药物是化学小分子药物、抗体、多肽/蛋白药物、核酸药物或纳米药物形成的第一种药物。Preferably, the drug is the first drug formed by a chemical small molecule drug, an antibody, a polypeptide/protein drug, a nucleic acid drug or a nano drug.
优选地,所述的sDSS1蛋白是以所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物、复合体、第一种药物中的任一一种成分的两种或多种的组合形成的第二种药物。Preferably, the sDSS1 protein is any one of the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification, the complex, and the first drug. A second drug formed by a combination of two or more of one component.
优选地,所述的sDSS1蛋白是以所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物、、非天然氨基酸替代蛋白、复合体、第一种药物中的任一一种成分的一种、两种或多种与医药上可用的赋形剂的组合形成的第三种药物。Preferably, the sDSS1 protein is a basic protein, a first protein, a second protein, a third protein, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, A third drug formed by the combination of one, two or more of any one of the first drugs with a pharmaceutically acceptable excipient.
优选地,所述的sDSS1蛋白是通过表达体系把编码所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白的核苷酸序列导入体内并表达获得的第四种蛋白。Preferably, the sDSS1 protein is a fourth obtained by introducing a nucleotide sequence encoding the basic protein, the first protein, the second protein, the third protein or the fusion protein into the body and expressing the same by an expression system. Protein.
优选地,所述的表达体系是真核表达质粒载体、腺病毒、腺相关病毒、慢病毒、逆转录病毒、杆状病毒、疱疹病毒、伪狂犬病毒、ZFN基因编辑技术、TALEN基因编辑技术、CRISPR/Cas基因编辑技术或其他医疗上可用的基因编辑技术或病毒载体。Preferably, the expression system is eukaryotic expression plasmid vector, adenovirus, adeno-associated virus, lentivirus, retrovirus, baculovirus, herpes virus, pseudorabies virus, ZFN gene editing technology, TALEN gene editing technology, CRISPR/Cas gene editing technology or other medically available gene editing technology or viral vector.
优选地,所述的sDSS1蛋白是通过移植细胞在个体体内获得的所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白形成的第五种蛋白。Preferably, the sDSS1 protein is a fifth protein formed by the basic protein, the first protein, the second protein, the third protein or the fusion protein obtained by the transplanted cell in the individual.
优选地,所述的细胞是任意一种人的干细胞、前体细胞或成体细胞。Preferably, the cell is a stem cell, a precursor cell or an adult cell of any one of humans.
优选地,所述的干细胞是胚胎干细胞、诱导多能干细胞、转分化得到的细胞,或者来源于原代培养的干细胞、由母细胞分化得到的多能或单能干细胞。Preferably, the stem cells are embryonic stem cells, induced pluripotent stem cells, cells obtained by transdifferentiation, or stem cells derived from primary culture, pluripotent or pluripotent stem cells differentiated from mother cells.
优选地,所述的sDSS1蛋白是通过移植组织或器官在个体体内获得的所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白形成的第六种蛋白。Preferably, the sDSS1 protein is a sixth protein formed by the basic protein, the first protein, the second protein, the third protein or the fusion protein obtained by transplanting tissues or organs in an individual.
优选地,所述的组织是脑、肝、肾、脾、胰岛的完整器官或部分组织块,或血液、脂肪、肌肉、骨髓、皮肤。Preferably, the tissue is a whole organ or part of a tissue block of the brain, liver, kidney, spleen, islet, or blood, fat, muscle, bone marrow, skin.
优选地,所述的sDSS1蛋白是通过血清、脑脊液、组织间液输注引入个体体内的第七种蛋白。Preferably, the sDSS1 protein is a seventh protein introduced into the body by serum, cerebrospinal fluid, and interstitial fluid infusion.
优选地,所述的预防药物是包含基础蛋白、第一种至第七种任一蛋白、融合蛋白、多肽/蛋白修饰物、非天然氨基酸替代蛋白、复合体、第一种药物、第二种药物、第三种药物、在表达体系、细胞、组织、器官、体液、组织液的蛋白药物、多肽药物、核酸药物、化学小分子药物、细胞产品、商业化移植组织、注射液、冻干粉、保健品、食品添加剂中的一种或多种。Preferably, the prophylactic agent comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, a first drug, and a second Drugs, third drugs, protein drugs in expression systems, cells, tissues, organs, body fluids, tissue fluids, peptide drugs, nucleic acid drugs, chemical small molecule drugs, cell products, commercial transplant tissues, injections, lyophilized powder, One or more of health care products and food additives.
优选地,所述的治疗药物是包含基础蛋白、第一种至第七种任一蛋白、融合蛋白、多肽/蛋白修饰物、非天然氨基酸替代蛋白、复合体、第一种药物、第二种药物、第三种药物、在表达体系、细胞、组织、器官、体液、组织液的蛋白药物、多肽药物、核酸药物、化学小分子药物、细胞产品、商业化移植组织、注射液、冻干粉、保健品、食品添加剂中的一种或多种。Preferably, the therapeutic agent comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, a non-natural amino acid replacement protein, a complex, a first drug, and a second Drugs, third drugs, protein drugs in expression systems, cells, tissues, organs, body fluids, tissue fluids, peptide drugs, nucleic acid drugs, chemical small molecule drugs, cell products, commercial transplant tissues, injections, lyophilized powder, One or more of health care products and food additives.
本发明的特点和/或有益效果有:Features and/or benefits of the present invention are:
1.本发明提供的sDSS1蛋白与Aβ蛋白结合,抑制Aβ蛋白聚集并降低Aβ蛋白与其受体蛋白的结合能力,有效缓解Aβ蛋白引起的细胞毒性。1. The sDSS1 protein provided by the invention binds to Aβ protein, inhibits the aggregation of Aβ protein and reduces the binding ability of Aβ protein to its receptor protein, and effectively alleviates the cytotoxicity caused by Aβ protein.
2.本发明提供的sDSS1蛋白可以加速小胶质细胞清除Aβ蛋白,并且屏蔽Aβ寡聚物毒性,减少Aβ寡聚物引起的小胶质细胞激活。2. The sDSS1 protein provided by the present invention can accelerate the clearance of Aβ protein by microglia, and shields Aβ oligomer toxicity and reduces microglia activation induced by Aβ oligomer.
3.本发明提供的sDSS1蛋白在CL4176线虫上显著改善AD模型线虫的疾病症状,降低线虫组织中Aβ蛋白的蓄积。3. The sDSS1 protein provided by the present invention significantly improves the disease symptoms of the AD model nematode on the CL4176 nematode and reduces the accumulation of Aβ protein in the nematode tissue.
4.本发明提供的sDSS1蛋白帮助小鼠清除Aβ,减少血液中Aβ水平。给AD模型APP/PS1小鼠长时间注射sDSS1蛋白可以有效减少脑组织中Aβ斑块沉积。4. The sDSS1 protein provided by the present invention helps mice clear Aβ and reduce Aβ levels in the blood. Long-term injection of sDSS1 protein into AD model APP/PS1 mice can effectively reduce Aβ plaque deposition in brain tissue.
5.本发明提供的sDSS1蛋白是人和其他灵长类动物所具有的 蛋白,分子量相对较小,免疫原性低,并且体内存在天然的蛋白降解机制,因此,临床应用不会引起明显的免疫反应或其他的毒副效应,安全可靠。5. The sDSS1 protein provided by the present invention is a protein possessed by humans and other primates, has a relatively small molecular weight, low immunogenicity, and has a natural protein degradation mechanism in vivo, and therefore, clinical application does not cause significant immunity. The reaction or other toxic side effects are safe and reliable.
综上,本发明提供了一种用于痴呆症治疗的sDSS1蛋白药物,通过分子水平、细胞水平和动物水平的实验验证,sDSS1蛋白可以与Aβ蛋白结合,抑制Aβ聚集,降低Aβ蛋白与受体的结合效率,促进小胶质细胞清除Aβ并减少小胶质细胞激活。在动物实验中,sDSS1蛋白促进血液中Aβ清除,减少组织中Aβ蛋白的过渡蓄积,缓解疾病症状。sDSS1蛋白免疫原性较低,药效显著,具备用于临床上制备痴呆症预防药物,或者用于制备治疗前期、早期、中期、晚期痴呆症药物的潜力。In summary, the present invention provides an sDSS1 protein drug for the treatment of dementia. The sDSS1 protein can bind to Aβ protein, inhibit Aβ aggregation, and reduce Aβ protein and receptor by molecular, cell and animal level experiments. The binding efficiency promotes microglia clearance of Aβ and reduces microglia activation. In animal experiments, sDSS1 protein promotes Aβ clearance in the blood, reduces the accumulation of Aβ protein in tissues, and alleviates disease symptoms. sDSS1 protein has low immunogenicity and remarkable efficacy, and has the potential to be used for clinical preparation of dementia prevention drugs or for preparing pre-treatment, early, intermediate, and advanced dementia drugs.
附图说明DRAWINGS
下面结合附图,对本发明做进一步详细的阐述,以使本发明能够清楚、完整,但不是为了限制本发明的保护范围。The invention is further described in detail below with reference to the accompanying drawings, in which
图1A-图1B.分子实验显示Aβ蛋白与sDSS1蛋白结合。Figure 1A-1B. Molecular experiments show that A[beta] protein binds to sDSSl protein.
图1A,分别把Aβ 1-42蛋白和Aβ 1-40蛋白与sDSS1蛋白进行孵育,用Aβ蛋白抗体可以检测到Aβ 1-42蛋白与sDSS1蛋白形成的复合物(分子量10KD-37KD之间,第3泳道和第4泳道)。Aβ 1-40蛋白单独孵育形成部分二聚体,与sDSS1蛋白孵育可以减少二聚体形成。1A, Aβ 1-42 protein and Aβ 1-40 protein were incubated with sDSS1 protein, respectively, and a complex of Aβ 1-42 protein and sDSS1 protein was detected by Aβ protein antibody (molecular weight 10KD-37KD, Lane 3 and lane 4). The Aβ 1-40 protein is incubated alone to form a partial dimer, and incubation with the sDSS1 protein reduces dimer formation.
图1B,用sDSS1蛋白抗体同样可以检测到sDSS1蛋白与Aβ 1-42蛋白(第3泳道和第4泳道)或sDSS1蛋白与Aβ 1-40蛋白(第6泳道和第7泳道)形成复合物(分子量37-150KD之间),并且复合物形成随着反应体系中sDSS1蛋白浓度增加而增加。Figure 1B, sDSS1 protein and Aβ 1-42 protein (lane 3 and lane 4) or sDSS1 protein and Aβ 1-40 protein (lane 6 and lane 7) can also be detected by sDSS1 protein antibody ( The molecular weight is between 37 and 150 KD), and the complex formation increases as the concentration of sDSS1 protein in the reaction system increases.
图1C.ThT实验检测Aβ1-42蛋白聚集。Aβ1-42蛋白在12小时就发生聚集,显示出上升曲线。按照分子量比率加入5%或10% sDSS1蛋白后,能抑制上升曲线,延长检测时间也没有观察到上升曲线,提示没有Aβ 1-42蛋白聚集发生。Figure 1C. ThT assay detects A[beta] 1-42 protein aggregation. The Aβ1-42 protein aggregates at 12 hours and shows an ascending curve. Addition of 5% or 10% sDSS1 protein according to the molecular weight ratio inhibited the ascending curve and prolonged the detection time without observing the rising curve, suggesting that no Aβ 1-42 protein aggregation occurred.
图1D.ThT实验检测Aβ1-40蛋白聚集。Aβ 1-40蛋白在24小时开始出现上升曲线,显示聚集体形成。按照分子量比率加入5%或10%sDSS1蛋白能抑制上升曲线,延长检测时间也没有观察到上升曲线,提示没有Aβ 1-40蛋白聚集发生。Figure 1 D. ThT assay detects Aβ1-40 protein aggregation. The Aβ 1-40 protein began to show a rising curve at 24 hours, indicating aggregate formation. Addition of 5% or 10% sDSS1 protein according to the molecular weight ratio inhibited the ascending curve, and the prolongation of the detection time did not observe a rising curve, suggesting that no Aβ 1-40 protein aggregation occurred.
图1E.ThT实验检测sDSS1蛋白突变体4抑制Aβ1-40蛋白聚集。Aβ1-40蛋白在20-24小时发生聚集,显示出上升曲线。按照分子量比率加入0.667%、0、3.33%或16.67%sDSS1(M4)蛋白后,能抑制上升曲线,延长检测时间也没有观察到上升曲线,提示没有Aβ 1-40蛋白聚集发生。Figure 1E. ThT assay detects sDSS1 protein mutant 4 inhibits Aβ1-40 protein aggregation. The Aβ1-40 protein aggregates at 20-24 hours and shows an ascending curve. When 0.667%, 0, 3.33%, or 16.67% sDSS1(M4) protein was added according to the molecular weight ratio, the rise curve was inhibited, and the rise time was not observed, indicating that no Aβ 1-40 protein aggregation occurred.
图1F.ThT实验检测sDSS1蛋白突变体11抑制Aβ1-40蛋白聚集。Aβ1-40蛋白在20-24小时发生聚集,显示出上升曲线。按照分子量比率加入0.667%、0、3.33%或16.67%sDSS1(M11)蛋白后,能抑制上升曲线,延长检测时间也没有观察到上升曲线,提示没有Aβ 1-40蛋白聚集发生。Figure 1F. ThT assay detects sDSS1 protein mutant 11 inhibiting Aβ1-40 protein aggregation. The Aβ1-40 protein aggregates at 20-24 hours and shows an ascending curve. When 0.667%, 0, 3.33%, or 16.67% sDSS1(M11) protein was added according to the molecular weight ratio, the rising curve was inhibited, and the rise time was not observed, indicating that no Aβ 1-40 protein aggregation occurred.
图2A-图2B.分子实验显示Aβ蛋白寡聚物与sDSS1蛋白结合。2A-2B. Molecular experiments show that Aβ protein oligomers bind to sDSS1 protein.
图2A,分别把Aβ 1-42蛋白寡聚物和Aβ 1-40蛋白寡聚物与sDSS1蛋白进行孵育,用Aβ蛋白抗体可以检测到Aβ 1-42蛋白与sDSS1蛋白形成的复合物(分子量10KD-37KD之间,第3泳道和第4泳道)。Aβ 1-42蛋白与sDSS1蛋白的反应没有检测到明显的差异。2A, the Aβ 1-42 protein oligomer and the Aβ 1-40 protein oligomer were incubated with the sDSS1 protein, respectively, and the complex formed by the Aβ 1-42 protein and the sDSS1 protein was detected by the Aβ protein antibody (molecular weight 10KD). Between -37KD, lane 3 and lane 4). No significant difference was detected between the Aβ 1-42 protein and the sDSS1 protein.
图2B,利用sDSS1蛋白抗体可以检测到sDSS1蛋白与Aβ 1-42寡聚物(第3泳道)或sDSS1蛋白与Aβ 1-40寡聚物(第5泳道)形成的复合物(分子量37-150KD之间)。2B, a complex of sDSS1 protein and Aβ 1-42 oligomer (lane 3) or sDSS1 protein and Aβ 1-40 oligomer (lane 5) can be detected by using sDSS1 protein antibody (molecular weight 37-150KD) between).
图2C.ThT实验检测sDSS1蛋白与Aβ1-40寡聚物反应。在30μM Aβ1-40寡聚物中按照分子量比率加入50%和100%sDSS1蛋白,可以见到Aβ 1-40寡聚物荧光值提高,在12小时检测时间内荧 光值基本保持不变。Figure 2C. ThT assay detects the reaction of sDSS1 protein with Aβ1-40 oligomer. The addition of 50% and 100% sDSS1 protein in a molecular weight ratio of 30 μM Aβ1-40 oligomer showed an increase in the fluorescence value of the Aβ 1-40 oligomer, and the fluorescence value remained substantially unchanged during the 12-hour detection period.
图2D.ThT实验检测sDSS1蛋白与Aβ1-42寡聚物反应。在30μM Aβ1-42寡聚物中按照分子量比率加入50%和100%sDSS1蛋白,可以见到Aβ 1-42寡聚物荧光值提高。在12小时检测限内,Aβ1-42寡聚物的荧光值还有上升,加入sDSS1蛋白后,荧光值上升减慢。Figure 2D. ThT assay detects the reaction of the sDSS1 protein with the Aβ 1-42 oligomer. The addition of 50% and 100% sDSS1 protein in a molecular weight ratio of 30 μM Aβ 1-42 oligomer showed an increase in the fluorescence value of the Aβ 1-42 oligomer. Within the 12-hour detection limit, the fluorescence value of the Aβ1-42 oligomer also increased, and the fluorescence value increased after the addition of the sDSS1 protein.
图3A.sDSS1蛋白不能与RAGE受体蛋白相互作用。分子互作仪检测结果显示,在结合曲线上,可以看到sDSS1蛋白进样后,看不到明显的上升结合曲线,也看不到明显的解离曲线。Figure 3A. The sDSS1 protein is unable to interact with the RAGE receptor protein. The results of molecular interaction test showed that on the binding curve, it can be seen that after the injection of sDSS1 protein, no obvious ascending binding curve was observed, and no obvious dissociation curve was observed.
图3B.sDSS1蛋白抑制Aβ蛋白与RAGE受体蛋白的相互作用。Aβ 1-42蛋白可以结合到RAGE受体蛋白上,加入sDSS1蛋白后可以抑制Aβ蛋白与RAGE结合效率,Aβ蛋白结合量下降,并且该效应呈现典型的剂量依赖效应。sDSS1蛋白浓度越大,Aβ蛋白结合效率越低。Figure 3B. sDSS1 protein inhibits the interaction of Aβ protein with RAGE receptor protein. Aβ 1-42 protein can bind to RAGE receptor protein, and the addition of sDSS1 protein can inhibit the binding efficiency of Aβ protein to RAGE, the binding amount of Aβ protein decreases, and the effect shows a typical dose-dependent effect. The greater the concentration of sDSS1 protein, the lower the binding efficiency of Aβ protein.
图4A-图4C.细胞实验验证sDSS1蛋白屏蔽Aβ1-42寡聚物毒性并保护N2a细胞活力。Figures 4A-4C. Cell experiments demonstrate that sDSS1 protein shields Aβ 1-42 oligomer toxicity and protects N2a cell viability.
图1A,Aβ1-42寡聚物引起N2a细胞毒性,加入Aβ 1-42寡聚物引起细胞异常聚集和变圆,sDSS1蛋白可以缓解这些现象。1A, Aβ1-42 oligomers cause N2a cytotoxicity, and the addition of Aβ 1-42 oligomers causes abnormal aggregation and rounding of cells, and sDSS1 protein can alleviate these phenomena.
图2B,检测细胞活力发现sDSS1蛋白可以减少细胞活力受Aβ 1-42寡聚物影响,等量的sDSS1蛋白加入后,细胞活力几乎不受Aβ 1-42寡聚物毒性的影响。Figure 2B, detection of cell viability revealed that sDSS1 protein can reduce cell viability by Aβ 1-42 oligomer. After the addition of the same amount of sDSS1 protein, cell viability is almost unaffected by the toxicity of Aβ 1-42 oligomer.
图2C,Aβ 1-42引起细胞毒性,显示为溶液LDH含量上升,加入sDSS1蛋白后,溶液LDH含量显著降低,显示细胞毒性水平下降。每组N=5。数据经ANOVE分析,*,p-value<0.05;**,p-value<0.01。In Fig. 2C, Aβ 1-42 caused cytotoxicity, which showed an increase in the LDH content of the solution. After the addition of sDSS1 protein, the LDH content of the solution was significantly decreased, indicating a decrease in cytotoxicity. N=5 per group. Data were analyzed by ANOVE, *, p-value < 0.05; **, p-value < 0.01.
图5A-图5B.细胞实验验证sDSS1蛋白屏蔽Aβ1-42寡聚物毒性并保护原代神经元细胞活力。Figures 5A-5B. Cell experiments demonstrate that sDSS1 protein shields Aβ 1-42 oligomer toxicity and protects primary neuronal cell viability.
图5A,10μM Aβ1-42寡聚物可以引起神经元细胞活力下降,加入5μM和10μM sDSS1蛋白后,细胞活力可以被挽回。Figure 5A, 10 μM Aβ 1-42 oligomers can cause a decrease in neuronal cell viability, and cell viability can be recovered after the addition of 5 μM and 10 μM sDSS1 protein.
图5B,sDSS1蛋白能减少Aβ 1-42蛋白引起的细胞毒性水平上升,10μM sDSS1蛋白可以保护细胞几乎不受Aβ 1-42寡聚物毒性的影响。每组N=10。数据经ANOVE分析,*,p-value<0.05;**,p-value<0.01。Figure 5B, sDSS1 protein can reduce the cytotoxicity level caused by Aβ 1-42 protein, and 10 μM sDSS1 protein can protect cells from the toxicity of Aβ 1-42 oligomer. N=10 per group. Data were analyzed by ANOVE, *, p-value < 0.05; **, p-value < 0.01.
图6A-图6B.sDSS1蛋白促进小胶质细胞清除Aβ蛋白。Figures 6A-6B. The sDSS1 protein promotes microglia clearance of Aβ protein.
图6A,培养基中同时添加Aβ 1-42蛋白和sDSS1蛋白,24小时后检测到小胶质细胞胞浆Aβ蛋白的浓度显著高于只添加Aβ蛋白的对照,随着添加的sDSS1蛋白浓度增加,胞浆Aβ蛋白浓度增加。Figure 6A, Aβ 1-42 protein and sDSS1 protein were added to the medium. The concentration of Aβ protein in microglia was significantly higher than that of the control with only Aβ protein added after 24 hours, with the increase of the concentration of sDSS1 protein added. The cytosolic Aβ protein concentration is increased.
图6B,相应的,在添加sDSS1蛋白的培养液中的Aβ 1-42蛋白的含量显著低于只添加Aβ的对照样品,且呈现剂量依赖效应。每组N=3。数据经ANOVE分析,*,p-value<0.05;**,p-value<0.01。Fig. 6B, correspondingly, the content of A? 1-42 protein in the culture medium to which sDSS1 protein was added was significantly lower than that of the control sample to which only A? was added, and showed a dose-dependent effect. N=3 per group. Data were analyzed by ANOVE, *, p-value < 0.05; **, p-value < 0.01.
图6C-图6D.sDSS1蛋白降低Aβ 1-42蛋白寡聚物引起的小胶质细胞激活。Figures 6C-6D. The sDSS1 protein reduces microglial activation by Aβ 1-42 protein oligomers.
图6C,Aβ 1-42蛋白寡聚物引起小胶质细胞的激活,表现为BV2细胞ROS水平提高,添加不同浓度的sDSS1蛋白后,BV2细胞ROS水平逐渐下降,接近对照组细胞水平。In Fig. 6C, Aβ 1-42 protein oligomers caused microglia activation, which showed an increase in ROS levels in BV2 cells. After adding different concentrations of sDSS1 protein, ROS levels in BV2 cells gradually decreased, which was close to the control group.
图6D,Aβ 1-42蛋白寡聚物刺激小胶质细胞释放炎症因子TNF-α,sDSS1蛋白添加后减少TNF-α释放。每组N=3。数据经ANOVE分析,*,p-value<0.05;**,p-value<0.01。Figure 6D, Aβ 1-42 protein oligomer stimulates microglia to release inflammatory factor TNF-α, and sDSS1 protein reduces TNF-α release. N=3 per group. Data were analyzed by ANOVE, *, p-value < 0.05; **, p-value < 0.01.
图7A.sDSS1蛋白缓解CL4176线虫的疾病症状。随着培养时间延长,AD模型CL4176线虫的瘫痪率逐渐上升,在培养液中加入sDSS1蛋白可以显著减慢瘫痪率上升。且缓解效率呈现药物剂量依赖性,随着sDSS1蛋白浓度增加,缓解效应增强。在26小时,对照组未瘫痪线虫的比率仅仅为15.09%,500μg/mL sDSS1蛋白给药组为39.47%,1000μg/mL sDSS1蛋白给药组为65.71%。到了43小时,对照组线虫几乎全部瘫痪(未瘫痪线虫比率小于1%),而500μg/mL sDSS1蛋白给药组中,未瘫痪的线虫比例为18.42%,1000μg/mL sDSS1蛋白给药组为48.57%。每组线虫数量为90条。 该实验重复3次,数据经Log-rank(Mantel-Cox)test分析,***,p-value<0.001。Figure 7A. The sDSS1 protein ameliorate the disease symptoms of CL4176 nematodes. As the culture time prolonged, the sputum rate of the AD model CL4176 nematode gradually increased, and the addition of sDSS1 protein to the culture medium significantly slowed down the rate of sputum. And the mitigation efficiency showed drug dose dependence, and the mitigation effect increased with the increase of sDSS1 protein concentration. At 26 hours, the ratio of the control group to the nematode was only 15.09%, the 500 μg/mL sDSS1 protein administration group was 39.47%, and the 1000 μg/mL sDSS1 protein administration group was 65.71%. At 43 hours, the nematode in the control group was almost all sputum (the ratio of the nematode was less than 1%), while in the 500 μg/mL sDSS1 protein-administered group, the proportion of the untreated nematode was 18.42%, and the 1000 μg/mL sDSS1 protein-administered group was 48.57. %. The number of nematodes per group is 90. The experiment was repeated 3 times and the data was analyzed by Log-rank (Mantel-Cox) test, ***, p-value < 0.001.
图7B.免疫组织荧光染色检测C14176线虫组织Aβ 1-42沉积。AD模型4176线虫组织经过固定和免疫荧光染色,Aβ 1-42特异性抗体用于显示Aβ蛋白(绿色荧光),Hoechst染料用于染色细胞核(蓝色荧光),白色箭头指示Aβ斑块沉积。在对照组线虫组织中可见典型的Aβ 1-42斑块和弥散的Aβ沉淀。经过500μg/mL sDSS1蛋白处理,Aβ斑块和弥散性沉积显著减少。1000μg/mL sDSS1蛋白给药组几乎看不到斑块,弥散性沉积较少。Figure 7B. Immunohistochemical staining for the detection of Aβ 1-42 deposition in C14176 nematode tissue. AD model 4176 elegans tissues were fixed and immunofluorescent stained, Aβ 1-42 specific antibodies were used to display Aβ protein (green fluorescence), Hoechst dye was used to stain nuclei (blue fluorescence), and white arrows indicate Aβ plaque deposition. Typical Aβ 1-42 plaques and diffuse Aβ precipitates were seen in the control nematode tissues. After treatment with 500 μg/mL sDSS1 protein, Aβ plaques and diffuse deposition were significantly reduced. The 1000 μg/mL sDSS1 protein administration group showed almost no plaque and less diffuse deposition.
图7C-图7D.4176线虫组织研磨液中Aβ 1-42聚集体和总Aβ蛋白含量减少。Figure 7C - Figure 7D. 4176 Reduction of A[beta] 1-42 aggregates and total A[beta] protein content in nematode tissue mills.
图7C,用Western blotting方法检测线虫组织研磨液中Aβ 1-42聚集体,可见经过sDSS1蛋白处理后,多数Aβ聚集体条带减少或完全消失(黑色箭头指示位置)。Figure 7C, Western blotting was used to detect Aβ 1-42 aggregates in nematode tissue mills. It can be seen that after treatment with sDSS1 protein, most Aβ aggregate bands decreased or disappeared completely (black arrow indicates position).
图7D,经过ELISA检测组织研磨液中总Aβ蛋白量,发现sDSS1蛋白给药组线虫组织总Aβ蛋白显著降低,1000μg/mL sDSS1蛋白给药组含量仅仅是对照组的59.07%。实验中每组用的线虫数量为400条。In Fig. 7D, the total Aβ protein in the tissue slurry was detected by ELISA, and it was found that the total Aβ protein of the nematode tissue of the sDSS1 protein administration group was significantly decreased, and the content of the 1000 μg/mL sDSS1 protein administration group was only 59.07% of the control group. The number of nematodes used in each group was 400 in the experiment.
图8A-图8B.sDSS1蛋白促进小鼠血液中Aβ蛋白清除。Figures 8A-8B. sDSS1 protein promotes Aβ protein clearance in mouse blood.
图8A,给小鼠静脉注射Aβ蛋白制造急性Aβ上升模型,结果显示sDSS1蛋白给药能加速Aβ蛋白清除。随着sDSS1蛋白给药量增加,Aβ含量明显下降。Figure 8A shows a model of acute Aβ rise by intravenous injection of Aβ protein into mice. The results show that sDSS1 protein administration accelerates Aβ protein clearance. As the amount of sDSS1 protein administered increased, the Aβ content decreased significantly.
图8B,给AD模型APP/PS1小鼠注射sDSS1蛋白,在血液中也可以检测到Aβ水平显著降低。急性模型每组5只小鼠,APP/PS1小鼠每组3只。数据经ANOVE分析,**,p-value<0.01;***,p-value<0.001。Figure 8B. Injecting sDSS1 protein into AD model APP/PS1 mice, a significant decrease in Aβ levels was also detected in the blood. Acute model 5 mice per group, APP/PS1 mice 3 per group. Data were analyzed by ANOVE, **, p-value < 0.01; ***, p-value < 0.001.
图9A-图9C.sDSS1蛋白降低APP/PS1小鼠脑组织Aβ斑块沉积。Figures 9A-9C. sDSS1 protein reduces Aβ plaque deposition in brain tissue of APP/PS1 mice.
图9A,经FSB染料染色后,在11月龄APP/PS1小鼠脑组织中可以看到大量Aβ斑块沉淀(蓝色荧光),sDSS1蛋白给药后,脑组织海马区斑块沉积下降,显示斑块数量减少,亮度降低。Fig. 9A, after staining with FSB dye, a large amount of Aβ plaque precipitate (blue fluorescence) was observed in the brain tissue of APP/PS1 mice at 11 months of age. After administration of sDSS1 protein, plaque deposition in the hippocampus of brain tissue decreased. The number of plaques is reduced and the brightness is reduced.
图9B,图片用Phototshop CS软件处理后,统计斑块面积,结果显示sDSS1蛋白给药后脑组织斑块面积显著低于对照组。Fig. 9B, after the photo was processed by Phototshop CS software, the plaque area was counted. The results showed that the plaque area of brain tissue after sDSS1 protein administration was significantly lower than that of the control group.
图9C,按照斑块大小统计小型斑块(图9A中标记为1)、中型斑块(图9A中标记为2)、大型斑块(图9A中标记为3)数量,数据显示与对照组相比,sDSS1蛋白给药后组织中中型斑块以及大型斑块数量显著下降,小型斑块没有显著差异。APP/PS1小鼠每组3只。数据经ANOVE分析,n.s.,p-value>0.5;**,p-value<0.01。Figure 9C, the number of small plaques (marked as 1 in Figure 9A), medium-sized plaques (marked as 2 in Figure 9A), and large plaques (marked as 3 in Figure 9A) were counted according to plaque size. In contrast, the number of medium-sized plaques and large plaques in tissues was significantly decreased after administration of sDSS1 protein, and there was no significant difference in small plaques. APP/PS1 mice were 3 in each group. The data was analyzed by ANOVE, n.s., p-value>0.5; **, p-value<0.01.
具体实施方式Detailed ways
以下内容将结合实例对本发明中的优选方案进行说明和验证,不是对本发明的范围进行限定。本发明的所有范围限定以权利要求书中的限定为准。The preferred embodiments of the present invention are described and illustrated in the following examples, which are not intended to limit the scope of the invention. All ranges of the invention are defined by the claims.
下述实施案例中所用的实验方法如无特殊说明,均为常规实验方法。The experimental methods used in the following examples are routine experimental methods unless otherwise specified.
下述实施案例中所用的sDSS1蛋白为本公司自行生产的人源sDSS1蛋白,蛋白序列见SEQ ID NO:1。本公司对蛋白品质进行质量控制,经检测蛋白纯度大于95%,内毒素(小于3EU/mg蛋白)和其他杂质残留符合标准,可用于动物实验而不引起明显的动物毒性反应。The sDSS1 protein used in the following examples is the human sDSS1 protein produced by the company itself, and the protein sequence is shown in SEQ ID NO: 1. The company controls the quality of protein. The purity of the tested protein is greater than 95%. The endotoxin (less than 3EU/mg protein) and other impurities are in compliance with the standard and can be used in animal experiments without causing significant animal toxicity.
下述实施案例中材料和试剂,除了sDSS1蛋白其他均可以通过商业途径获取。The materials and reagents in the following examples, except for the sDSS1 protein, are commercially available.
实施例1、sDSS1蛋白抑制Aβ蛋白聚集。Example 1. The sDSS1 protein inhibits Aβ protein aggregation.
实验方法experimental method
1.ThT实验 Aβ1-42和Aβ1-40蛋白委托苏州强耀生物科技有 限公司合成并制成冻干粉,蛋白纯度经检测大于95%。硫代黄素T染料(ThT,Sigam-Aldrich,T3516)首先用甲醇溶解为1mg/mL(3.1mM),继续用PBS稀释为1mM。Aβ蛋白用20mM NaOH溶解为2mg/mL并用PBS稀释为100μΜ。 sDSS1蛋白、sDSS1蛋白 突变体4(sDSS1(M4))(氨基酸序列见SEQ ID 15)、sDSS1蛋白 突变体11(sDSS1(M11))(氨基酸序列见SEQ ID 16)分别用PBS稀释为1mg/mL(100μM)。实验在PBS环境中进行,首先在1.5mL离心管中补充需要添加PBS,然后依次加入sDSS1蛋白或突变体蛋白、Aβ蛋白和ThT染料,sDSS1蛋白摩尔浓度根据预设的浓度梯度添加。对于Aβ1-42蛋白,Aβ蛋白和ThT染料的摩尔浓度分别为30μM和5μM;对于Aβ 1-40蛋白,Aβ蛋白和ThT染料的摩尔浓度分别为30μM和10μM。完成后,溶液在混悬仪上震荡混匀,然后取200μL加入到黑色荧光检测板(Costar,3792)中,每组两个复孔。黑色荧光检测板放入多功能酶标仪(Molecular Devices,SpectraMax i3x)上检测荧光值,设置激发光450nm,发射光485nm,检测间隔30分钟,连续检测48个小时。 1.ThT experiment Aβ1-42 and Aβ1-40 protein were entrusted to Suzhou Qiang Yao Biotechnology Co., Ltd. to synthesize and make lyophilized powder, and the protein purity was detected to be greater than 95%. The thioflavin T dye (ThT, Sigam-Aldrich, T3516) was first dissolved in methanol to 1 mg/mL (3.1 mM) and continued to be diluted to 1 mM with PBS. The Aβ protein was dissolved to 2 mg/mL with 20 mM NaOH and diluted to 100 μM with PBS. sDSS1 protein, sDSS1 mutants 4 (sDSS1 (M4)) (amino acid sequence is shown in SEQ ID 15), sDSS1 mutants 11 (amino acid sequence see SEQ ID 16) (sDSS1 (M11 )) were diluted to 1mg / mL in PBS (100 μM). The experiment was carried out in a PBS environment. First, a PBS was added in a 1.5 mL centrifuge tube, and then sDSS1 protein or mutant protein, Aβ protein and ThT dye were sequentially added, and the sDSS1 protein molar concentration was added according to a preset concentration gradient. For the Aβ 1-42 protein, the molar concentrations of the Aβ protein and the ThT dye were 30 μM and 5 μM, respectively; for the Aβ 1-40 protein, the molar concentrations of the Aβ protein and the ThT dye were 30 μM and 10 μM, respectively. After completion, the solution was shaken and mixed on the suspension, and then 200 μL was added to a black fluorescence detection plate (Costar, 3792) for two duplicate wells per group. The black fluorescence detection plate was placed on a multi-function microplate reader (Molecular Devices, SpectraMax i3x) to detect the fluorescence value, and the excitation light was set to 450 nm, the emission light was 485 nm, the detection interval was 30 minutes, and the detection was continued for 48 hours.
2.蛋白质相互作用 Aβ 1-42和Aβ 1-40蛋白单体用PBS稀释到20μM并与10μM或20μM sDSS1蛋白混合,20μM Aβ蛋白作为阳性对照。蛋白液混匀后放在4℃孵育过夜。孵育的蛋白液中加入5ⅹ上样缓冲液,混匀并在100℃加热10分钟,制备的样品用于Western Blotting分析。2. Protein Interactions Aβ 1-42 and Aβ 1-40 protein monomers were diluted to 20 μM with PBS and mixed with 10 μM or 20 μM sDSS1 protein, and 20 μM Aβ protein was used as a positive control. The protein solution was mixed and incubated overnight at 4 °C. 5x loading buffer was added to the incubated protein solution, mixed and heated at 100 ° C for 10 minutes, and the prepared samples were used for Western Blotting analysis.
3.蛋白免疫印迹实验(Western Blotting) 15μL制备的上样样品加入上样孔,10%预制胶(Life technology公司C#NP0321BOX)分离蛋白后转移到PVDF膜上。膜依次经过5%脱脂牛奶封闭1小时,第一级抗体Rabbit-anti-Aβ(Cell Signaling Technology公司,#8243)或Rabbit-anti-sDSS1(发明人委托上海睿智化学研究有限公司制备)4℃孵育过夜,TBST溶液清洗三遍;第二级抗体(Goat-anti-rabbit HRP抗体)室温孵育2小时。用TBST清洗三遍,完成后用发光液 显影并用X光片显示条带。3. Western Blotting 15 μL of the prepared sample was added to the well, and 10% of the pre-formed gel (Life Technology C#NP0321BOX) was used to separate the protein and transferred to the PVDF membrane. The membrane was blocked by 5% skim milk for 1 hour, and the first-stage antibody Rabbit-anti-Aβ (Cell Signaling Technology, #8243) or Rabbit-anti-sDSS1 (invented by Shanghai Ruizhi Chemical Research Co., Ltd.) was incubated at 4 °C. Overnight, the TBST solution was washed three times; the second antibody (Goat-anti-rabbit HRP antibody) was incubated for 2 hours at room temperature. It was washed three times with TBST, and after completion, it was developed with a luminescent liquid and the strip was displayed with an X-ray film.
实验结果Experimental result
在缓冲液中,把Aβ 1-42或Aβ 1-40蛋白与sDSS1蛋白进行共孵育,检测孵育后的混合物,可以看到Aβ 1-42蛋白与sDSS1蛋白形成复合物(分子量10KD-37KD)。Aβ 1-40蛋白与sDSS1蛋白共孵育可以显著减少二聚体的形成(图1A)。用sDSS1蛋白抗体同样科技检测到Aβ蛋白与sDSS1蛋白复合物信号,并且随着sDSS1蛋白浓度增加,复合物信号增强(图1B)。Aβ 1-42或Aβ 1-40单体蛋白发生聚集,形成淀粉样沉淀,与ThT染料结合后荧光强度增强。在对照组,可以看到随着孵育时间延长,荧光亮度逐渐增强,显示Aβ 1-40和Aβ 1-42蛋白发生聚集。但是,在反应体系中加入不同浓度的sDSS1蛋白或sDSS1突变体蛋白后,荧光亮度不增加并持续保持在初始值附近,说明sDSS1蛋白显著抑制了Aβ蛋白聚集。而且,最低仅需要在反应体系中添加5%的sDSS1蛋白即可抑制Aβ蛋白聚集反应(图1C,图1D); 在反应体系中添加 0.667%或3.33%的sDSS1(M4)蛋白或sDSS1(M11)蛋白即可抑制 Aβ蛋白聚集反应(图1E,图1F)。这些结果说明,sDSS1蛋白可以与Aβ蛋白发生相互作用形成复合物,并抑制Aβ蛋白聚集体形成过程。 In the buffer, Aβ 1-42 or Aβ 1-40 protein was co-incubated with sDSS1 protein, and the mixture after incubation was detected. It was found that Aβ 1-42 protein forms a complex with sDSS1 protein (molecular weight 10KD-37KD). Co-incubation of the Aβ 1-40 protein with the sDSS1 protein significantly reduced the formation of dimers (Fig. 1A). The Aβ protein and sDSS1 protein complex signals were detected by the same technique using the sDSS1 protein antibody, and the complex signal was enhanced as the sDSS1 protein concentration was increased (Fig. 1B). Aβ 1-42 or Aβ 1-40 monomeric proteins aggregate to form an amyloid deposit, which is enhanced by binding to ThT dye. In the control group, it can be seen that as the incubation time is prolonged, the fluorescence brightness gradually increases, indicating that Aβ 1-40 and Aβ 1-42 proteins are aggregated. However, after adding different concentrations of sDSS1 protein or sDSS1 mutant protein in the reaction system, the fluorescence brightness did not increase and remained near the initial value, indicating that sDSS1 protein significantly inhibited Aβ protein aggregation. Moreover, it is only necessary to add 5% of sDSS1 protein to the reaction system to inhibit the Aβ protein aggregation reaction (Fig. 1C, Fig. 1D); 0.667% or 3.33% of sDSS1(M4) protein or sDSS1 (M11) is added to the reaction system. The protein can inhibit the Aβ protein aggregation reaction (Fig. 1E, Fig. 1F) . These results indicate that the sDSS1 protein can interact with the Aβ protein to form a complex and inhibit the formation of Aβ protein aggregates.
实施例2、sDSS1蛋白与Aβ蛋白寡聚物结合。Example 2. The sDSS1 protein binds to an Aβ protein oligomer.
实验方法experimental method
1.Aβ 1-42蛋白溶解及寡聚物制作 1mg Aβ1-42或Aβ1-40蛋白加入500μL 20mM NaOH溶液,震荡混匀10分钟后,超声10分钟帮助蛋白溶解。用PBS溶液稀释到100μM,稀释液放在4℃孵育24小时,12000g离心10分钟除去可能的沉淀,获得Aβ 1-42或Aβ 1-40寡聚物。Aβ寡聚物按照孵育前Aβ单体蛋白浓度来标注。1. Aβ 1-42 protein solubilization and oligomer preparation 1 mg Aβ1-42 or Aβ1-40 protein was added to 500 μL of 20 mM NaOH solution, shaken and mixed for 10 minutes, and then sonicated for 10 minutes to help dissolve the protein. Dilute to 100 μM with PBS solution, incubate at 4 ° C for 24 hours, centrifuge at 12000 g for 10 minutes to remove possible precipitates, and obtain Aβ 1-42 or Aβ 1-40 oligomers. Aβ oligomers are labeled according to the concentration of Aβ monomer protein prior to incubation.
2.ThT实验 sDSS1蛋白用PBS稀释为1mg/mL(100μM)。 实验在PBS条件下进行,首先在1.5mL离心管中补充需要添加PBS,然后依次加入sDSS1蛋白、Aβ蛋白寡聚物和ThT染料,sDSS1蛋白摩尔浓度根据预设的浓度梯度添加。对于Aβ 1-42蛋白,Aβ蛋白和ThT染料的摩尔浓度分别为30μM和5mΜ;对于Aβ 1-40蛋白,Aβ蛋白和ThT染料的摩尔浓度分别为30μM和10mΜ。完成后,溶液在混悬仪上震荡混匀,然后取200μL加入到黑色荧光检测板(Costar,3792)中,每组两个复孔。黑色荧光检测板放入多功能酶标仪上检测荧光值,设置激发光450nm,发射光485nm,检测间隔30分钟,连续检测12个小时。2. ThT assay The sDSS1 protein was diluted to 1 mg/mL (100 μM) with PBS. The experiment was carried out under PBS conditions. First, PBS was added in a 1.5 mL centrifuge tube, and then sDSS1 protein, Aβ protein oligomer and ThT dye were sequentially added, and the molar concentration of sDSS1 protein was added according to a preset concentration gradient. For the Aβ 1-42 protein, the molar concentrations of the Aβ protein and the ThT dye were 30 μM and 5 mΜ, respectively; for the Aβ 1-40 protein, the molar concentrations of the Aβ protein and the ThT dye were 30 μM and 10 mΜ, respectively. After completion, the solution was shaken and mixed on the suspension, and then 200 μL was added to a black fluorescence detection plate (Costar, 3792) for two duplicate wells per group. The black fluorescent detection plate was placed on a multi-function microplate reader to detect the fluorescence value, and the excitation light was set to 450 nm, the emission light was 485 nm, the detection interval was 30 minutes, and the detection was continued for 12 hours.
3.蛋白质相互作用 Aβ 1-42或Aβ 1-40蛋白寡聚物用PBS稀释到20μM并与10μM或20μM sDSS1蛋白混合,20μM Aβ寡聚物作为阳性对照。蛋白液混匀后放在4℃孵育过夜。孵育的蛋白液中加入5ⅹ上样缓冲液,混匀并在100℃加热10分钟,制备的样品用于Western Blotting分析。3. Protein Interaction The Aβ 1-42 or Aβ 1-40 protein oligomer was diluted to 20 μM with PBS and mixed with 10 μM or 20 μM sDSS1 protein, and 20 μM Aβ oligomer was used as a positive control. The protein solution was mixed and incubated overnight at 4 °C. 5x loading buffer was added to the incubated protein solution, mixed and heated at 100 ° C for 10 minutes, and the prepared samples were used for Western Blotting analysis.
4.蛋白免疫印迹实验(Western Blotting) 15μL制备的上样样品加入上样孔,4-12%预制胶(Life technology公司C#NP0321BOX)分离蛋白后转移到PVDF膜上。膜依次经过5%脱脂牛奶封闭1小时,第一级抗体Rabbit-anti-Aβ(Cell Signaling Technology公司,#8243)或Rabbit-anti-sDSS1(发明人委托上海睿智化学研究有限公司制备)4℃孵育过夜,TBST溶液清洗三遍;第二级抗体(Goat-anti-rabbit HRP抗体)室温孵育2小时。用TBST清洗三遍,完成后用发光液显影并用X光片显示条带。4. Western Blotting 15 μL of the prepared sample was added to the well, and 4-12% pre-formed (Life Technology C#NP0321BOX) was used to separate the protein and transferred to the PVDF membrane. The membrane was blocked by 5% skim milk for 1 hour, and the first-stage antibody Rabbit-anti-Aβ (Cell Signaling Technology, #8243) or Rabbit-anti-sDSS1 (invented by Shanghai Ruizhi Chemical Research Co., Ltd.) was incubated at 4 °C. Overnight, the TBST solution was washed three times; the second antibody (Goat-anti-rabbit HRP antibody) was incubated for 2 hours at room temperature. It was washed three times with TBST, and after completion, it was developed with a luminescent liquid and the strip was displayed with an X-ray film.
实验结果Experimental result
Aβ 1-40或Aβ 1-42的寡聚物是Aβ蛋白产生细胞毒性的主要形式。为了检测sDSS1蛋白与Aβ蛋白寡聚物的反应,首先孵育Aβ蛋白形成寡聚物形式,然后与sDSS1蛋白进行共同孵育并进行western blotting和ThT检测。结果显示,用Aβ蛋白抗体可以检测到sDSS1蛋白与Aβ 1-42寡聚物形式的反应,复合物含量随着反应 体系中sDSS1蛋白浓度增加而增加(图2A)。用sDSS1蛋白抗体同样可以检测sDSS1蛋白与Aβ蛋白形成的复合物(图2B)。在ThT荧光实验中,可以看到sDSS1蛋白可以与Aβ 1-40或Aβ 1-42的淀粉样沉淀发生反应,sDSS1蛋白与Aβ 1-40淀粉样沉淀反应后,荧光强度增加(图2C);sDSS1蛋白与Aβ 1-42淀粉样沉淀反应后,荧光强度显著降低(图2D)。以上结果说明,sDSS1蛋白也能与Aβ蛋白寡聚物形式发生结合形成复合物,这些相互作用可能抑制Aβ蛋白的细胞毒性。The oligomer of Aβ 1-40 or Aβ 1-42 is the major form of Aβ protein producing cytotoxicity. In order to detect the reaction of sDSS1 protein with Aβ protein oligomer, the Aβ protein was first incubated to form an oligomer, and then incubated with sDSS1 protein for western blotting and ThT detection. The results showed that the reaction of sDSS1 protein with Aβ 1-42 oligomer form was detected by Aβ protein antibody, and the complex content increased with the increase of sDSS1 protein concentration in the reaction system (Fig. 2A). A complex formed by the sDSS1 protein and the Aβ protein can also be detected using the sDSS1 protein antibody (Fig. 2B). In the ThT fluorescence experiment, it can be seen that the sDSS1 protein can react with the amyloid precipitation of Aβ 1-40 or Aβ 1-42, and the fluorescence intensity increases after the sDSS1 protein reacts with the Aβ 1-40 amyloid precipitate (Fig. 2C); After the reaction of the sDSS1 protein with the Aβ 1-42 amyloid precipitate, the fluorescence intensity was significantly reduced (Fig. 2D). The above results indicate that the sDSS1 protein can also form a complex with the Aβ protein oligomer form, and these interactions may inhibit the cytotoxicity of the Aβ protein.
实施例3、sDSS1蛋白降低Aβ蛋白与受体蛋白的结合效率。Example 3, sDSS1 protein reduces the binding efficiency of Aβ protein to receptor protein.
实验方法experimental method
1.分子互作实验 利用生物大分子相互作用仪(GE,Biacore3000)检测RAGE蛋白与sDSS1蛋白的结合效率。首先用PBS冲洗芯片20分钟直到基线稳定,继续用1μg/mL RAGE蛋白上的His标签把蛋白耦联到传感片NTA芯片(GE),耦联量大约50RU。把sDSS1蛋白稀释到10μg/mL,上样量20μL,上样时间200秒。完成后,用PBS溶液冲洗,检测洗脱曲线。1. Molecular interaction experiments The biomacromolecular interaction instrument (GE, Biacore 3000) was used to detect the binding efficiency of RAGE protein to sDSS1 protein. The chip was first rinsed with PBS for 20 minutes until the baseline was stable, and the protein was coupled to the sensor chip NTA chip (GE) with a His tag on 1 μg/mL RAGE protein with a coupling amount of approximately 50 RU. The sDSS1 protein was diluted to 10 μg/mL, the loading amount was 20 μL, and the loading time was 200 seconds. After completion, rinse with PBS solution and measure the elution curve.
2.配体受体结合实验 本实验采用ELISA方法检测Aβ蛋白单体与糖基化蛋白受体(receptor of advanced glycation end product,RAGE)结合效率的变化。RAGE蛋白购自北京义翘神州科技有限公司(11629-H02H)。空白酶标板用PBS清洗一遍,每孔加入100μL用PBS稀释的0.5μg/mL RAGE蛋白4℃孵育过夜,完成后用PBS清洗一遍。加入PBS稀释的2%BSA in PBST(0.05%Tween 20)在37℃封闭1个小时,完成后用PBS清洗一遍。加入100μL Aβ1-42蛋白或Aβ1-42蛋白与sDSS1蛋白混合物,设置Aβ1-42蛋白浓度分别为0.625μg/mL和0.3125μg/mL以及sDSS1蛋白浓度分别为0.2μg/mL和1μg/mL,室温孵育2小时。用PBST(0.05%Tween 20)清洗一遍,加入100μL Rabbit-anti-Aβ(1:10000稀释在PBST溶液,包 含0.5%BSA),室温孵育1小时,完成后用PBST清洗三遍。继续加入100μL辣根过氧化物酶(HRP)耦联的山羊来源抗兔抗体(1:10000稀释在PBST溶液,包含0.5%BSA),室温孵育1小时,完成后用PBST清洗6遍。加入200μL TMB显色液,室温孵育30分钟,完成后加入50μL终止液。96孔板在酶标仪上读取450nm吸光度值,反映配基配体结合量。2. Ligand receptor binding assay In this experiment, the ELISA method was used to detect the change of binding efficiency of Aβ protein monomer and receptor glycoprotein (receptor of advanced glycation end product (RAGE). RAGE protein was purchased from Beijing Yiqiao Shenzhou Technology Co., Ltd. (11629-H02H). The blank plate was washed once with PBS, and 100 μL of 0.5 μg/mL RAGE protein diluted with PBS was added to each well overnight at 4 ° C, and washed once with PBS. The PBS diluted 2% BSA in PBST (0.05% Tween 20) was added and blocked at 37 ° C for 1 hour, and then washed once with PBS. Add 100 μL of Aβ1-42 protein or Aβ1-42 protein and sDSS1 protein mixture, set Aβ1-42 protein concentration to 0.625μg/mL and 0.3125μg/mL, respectively, and sDSS1 protein concentration of 0.2μg/mL and 1μg/mL, respectively, incubate at room temperature. 2 hours. After washing once with PBST (0.05% Tween 20), 100 μL of Rabbit-anti-Aβ (1:10000 diluted in PBST solution containing 0.5% BSA) was added, and the mixture was incubated at room temperature for 1 hour, and then washed three times with PBST. Continue to add 100 μL of horseradish peroxidase (HRP)-coupled goat-derived anti-rabbit antibody (1:10000 diluted in PBST solution containing 0.5% BSA), incubate for 1 hour at room temperature, and wash 6 times with PBST after completion. Add 200 μL of TMB color solution and incubate for 30 minutes at room temperature. After completion, add 50 μL of stop solution. The 96-well plate reads the 450 nm absorbance value on the microplate reader, reflecting the ligand binding amount of the ligand.
实验结果Experimental result
利用BIAcore设备,检测发现sDSS1蛋白与RAGE蛋白不能发生结合,在BIAcore上显示没有明显的结合峰(图3A)。ELISA实验显示,Aβ1-42蛋白作为配体可以与RAGE蛋白结合,显示较高的结合量。加入0.2μg/mL和1μg/mL sDSS1蛋白可以降低Aβ1-42与RAGE的结合量,并且该效应呈现明显的浓度依赖效应(图3B)。这些结果说明,sDSS1蛋白与Aβ1-42蛋白反应后可以降低Aβ蛋白结合与受体的结合效率。Using the BIAcore device, it was found that the sDSS1 protein could not bind to the RAGE protein and showed no significant binding peak on BIAcore (Fig. 3A). ELISA experiments showed that Aβ1-42 protein as a ligand can bind to RAGE protein, showing a higher binding amount. The addition of 0.2 μg/mL and 1 μg/mL sDSS1 protein reduced the binding amount of Aβ 1-42 to RAGE, and this effect exhibited a significant concentration-dependent effect ( FIG. 3B ). These results indicate that the sDSS1 protein reacts with the Aβ1-42 protein to reduce the binding efficiency of Aβ protein binding to the receptor.
实施例4、sDSS1蛋白屏蔽Aβ寡聚物引起的细胞毒性。Example 4, sDSS1 protein shields cytotoxicity caused by Aβ oligomers.
实验方法experimental method
1.细胞培养 小鼠来源神经瘤母细胞(N2a)购自中国科学院典型培养物保藏委员会细胞库(细胞目录号:TCM29)。N2a细胞培养在含10%胎牛血清(FBS,Gibco,10091148)的Dulbecco's Modified Eagle Medium(DMEM,ThermoFisher Scientific,11995065)完全培养液中,每两天传代一次。1. Cell Culture Mouse-derived neuroblastoma cells (N2a) were purchased from the Cell Bank of the Type Culture Collection Committee of the Chinese Academy of Sciences (Cell No.: TCM29). N2a cells were cultured in Dulbecco's Modified Eagle Medium (DMEM, Thermo Fisher Scientific, 11995065) complete medium containing 10% fetal calf serum (FBS, Gibco, 10091148) and subcultured every two days.
2.原代神经元培养 怀孕16天的小鼠用乙醚深度麻醉后颈椎脱臼处死,解剖出E16期胎鼠。胎鼠用冰冷冻麻醉后,压迫颈椎处死,解剖出鼠脑,浸泡在预冷的DMEM溶液(含2%双抗)中,用眼科镊除去软脑膜并反复清洗去除血迹。解剖额叶皮层并用眼科剪剪碎组织,所有组织收集到离心管中,加入5mL 0.05%胰酶消化液在室温下消化15分钟。完成后,加入等体积DMEM完全培养基终止 消化,反复吹打组织并用40μm细胞滤器过滤,获得细胞悬液。细胞计数后按照3.0ⅹ10 4细胞每孔加入96孔板中,200μL DMEM完全培养基,培养板放在37℃培养。24小时后,换成200μL神经元培养基,包含98%Neurobasal(Gibco,21103049),2%B27 supplement(Gibco,17504044),1%GlutaMAX-1supplement(Gibco,35050061)。以后每两天换一次新鲜神经元培养基。培养的神经元8天之后成熟可以用于后续实验。 2. Primary neuron culture The mice that were pregnant for 16 days were deeply anesthetized with ether and then sacrificed by cervical dislocation. The E16 stage fetus was dissected. After the fetal rats were anesthetized with ice, the cervical vertebrae were sacrificed, the rat brain was dissected, and immersed in a pre-cooled DMEM solution (containing 2% double-antibody). The pia mater was removed by ophthalmology and repeatedly washed to remove blood. The frontal cortex was dissected and the tissue was cut with an ophthalmic scissors. All tissues were collected into a centrifuge tube and digested with 5 mL of 0.05% trypsin for 15 minutes at room temperature. After completion, the digestion was terminated by adding an equal volume of DMEM complete medium, and the tissue was repeatedly beaten and filtered with a 40 μm cell strainer to obtain a cell suspension. After the cells were counted, they were added to a 96-well plate at a rate of 3.0 x 10 4 cells per well, 200 μL of DMEM complete medium, and the plate was incubated at 37 °C. After 24 hours, switch to 200 μL of neuronal medium containing 98% Neurobasal (Gibco, 21103049), 2% B27 supplement (Gibco, 17504044), 1% GlutaMAX-1supplement (Gibco, 35050061). Fresh neuronal medium was changed every two days thereafter. Cultured neurons matured after 8 days and can be used in subsequent experiments.
2.Aβ1-42蛋白溶解及寡聚物制作 1mg Aβ1-42用无血清的DMEM-/-培养基稀释到100μM,稀释液放在4℃孵育24小时,获得Aβ1-42寡聚物蛋白。Aβ寡聚物蛋白按照孵育前Aβ单体蛋白浓度来标注。2. Aβ1-42 protein solubilization and oligomer preparation 1 mg of Aβ1-42 was diluted to 100 μM with serum-free DMEM-/- medium, and the dilution was incubated at 4 ° C for 24 hours to obtain Aβ1-42 oligomer protein. The Aβ oligomer protein is labeled according to the concentration of Aβ monomer protein before incubation.
3.细胞毒性实验 N2a细胞按照2ⅹ10 4细胞每孔接种到96孔板中。细胞贴壁12小时后,换成无血清DMEM培养基,细胞饥饿处理24小时。Aβ 1-42寡聚物用无血清DMEM稀释成20μM溶液。阴性对照组用无血清DMEM,阳性对照组加入20μM Aβ蛋白溶液,实验组除了20μM Aβ蛋白,分别包含10μM、20μM sDSS1蛋白。对于神经元细胞,细胞成熟后,用无血清DMEM按照N2a细胞实验过程稀释Aβ并设置相同的sDSS1蛋白梯度。神经元处理48小时。收集96孔板中的细胞的上清液,100g离心10分钟取上清液用于细胞毒性水平检测;细胞用于细胞活力检测。 3. Cytotoxicity assay N2a cells were seeded into 96-well plates at 2x10 4 cells per well. After the cells were attached for 12 hours, they were replaced with serum-free DMEM medium, and the cells were starved for 24 hours. The Aβ 1-42 oligomer was diluted to a 20 μM solution in serum-free DMEM. The negative control group was treated with serum-free DMEM, and the positive control group was added with 20 μM Aβ protein solution. The experimental group contained 10 μM and 20 μM sDSS1 protein in addition to 20 μM Aβ protein. For neuronal cells, after cell maturation, Aβ was diluted with serum-free DMEM according to the N2a cell experiment and the same sDSS1 protein gradient was set. Neurons were processed for 48 hours. The supernatant of the cells in the 96-well plate was collected, centrifuged at 100 g for 10 minutes, and the supernatant was used for cytotoxicity detection; the cells were used for cell viability assay.
4.细胞毒性水平检测 乳酸脱氢酶细胞毒性检测试剂盒购自碧云天生物科技有限公司(C0016)。检测酶活性时,首先根据试剂盒说明书配置好足量检测工作液。在96孔板中每孔加入120μL细胞毒性实验中收集的上清液,然后加入60μL检测工作液,稍混匀,在37℃孵育30分钟。在酶标仪上检测490nm吸光度值,吸光度高低与细胞毒性水平成正比。4. Detection of cytotoxicity The lactate dehydrogenase cytotoxicity test kit was purchased from Biyuntian Biotechnology Co., Ltd. (C0016). When detecting the enzyme activity, first configure a sufficient amount of the working fluid according to the kit instructions. The supernatant collected in a 120 μL cytotoxicity experiment was added to each well of a 96-well plate, then 60 μL of the test working solution was added, mixed slightly, and incubated at 37 ° C for 30 minutes. The absorbance at 490 nm was measured on a microplate reader, and the absorbance was directly proportional to the cytotoxicity level.
5.细胞活力检测 细胞增殖/细胞毒性检测试剂盒(CCK-8)购自东仁化学科技(上海)有限公司(CK04)。用无血清DMEM按照 1:20(体积比,v/v)稀释CCK-8溶液制成工作液。96孔中的细胞处理完成后,弃去旧培养基,每孔加入100μl CCK-8工作液。培养板放在培养箱中孵育1-2小时,然后在多功能酶标仪上检测450nm吸光度值,反映细胞活力。5. Cell viability assay The cell proliferation/cytotoxicity assay kit (CCK-8) was purchased from Dongren Chemical Technology (Shanghai) Co., Ltd. (CK04). The working solution was prepared by diluting the CCK-8 solution with 1:20 (volume ratio, v/v) in serum-free DMEM. After completion of the cell treatment in the 96 wells, the old medium was discarded, and 100 μl of CCK-8 working solution was added to each well. The plate was incubated in an incubator for 1-2 hours, and then the 450 nm absorbance value was measured on a multi-function microplate reader to reflect the cell viability.
实验结果Experimental result
为了检测sDSS1蛋白对Aβ1-42蛋白寡聚物细胞毒性的屏蔽作用,分别检测sDSS1蛋白对小鼠神经瘤母细胞和原代培养小鼠神经元的保护效应。实验结果显示,Aβ 1-42寡聚物蛋白能引起N2a细胞明显的细胞毒性,寡聚物处理48小时导致细胞变圆及异常聚集,加入sDSS1蛋白可以减少细胞毒性反应(图4A)。用CCK-8试剂盒检测细胞活力发现,sDSS1蛋白可以显著屏蔽Aβ 1-42寡聚物引起的细胞活力下降,20μM sDSS1蛋白可以保护细胞几乎不受Aβ 1-42寡聚物的影响(图4B)。LDH试剂盒检测细胞毒性反应水平,结果显示sDSS1蛋白组的细胞毒性反应水平明显下降(图4C)。在小鼠原代神经元细胞上,sDSS1蛋白同样显示对Aβ寡聚物毒性的屏蔽效应,神经元的细胞活力受到sDSS1蛋白保护(图5A),细胞毒性反应水平下降(图5B)。这些结果显示,sDSS1蛋白可以显著屏蔽Aβ寡聚物蛋白引起的细胞毒性。In order to detect the shielding effect of sDSS1 protein on the cytotoxicity of Aβ1-42 protein oligomer, the protective effect of sDSS1 protein on mouse neuroblastoma cells and primary cultured mouse neurons was examined. The results showed that Aβ 1-42 oligomer protein can cause significant cytotoxicity in N2a cells, and oligomer treatment for 48 hours leads to rounding and abnormal aggregation of cells, and addition of sDSS1 protein can reduce cytotoxicity (Fig. 4A). Using CCK-8 kit to detect cell viability, it was found that sDSS1 protein can significantly block the decrease of cell viability caused by Aβ 1-42 oligomer, and 20 μM sDSS1 protein can protect cells from Aβ 1-42 oligomer (Fig. 4B). ). The cytotoxicity level was measured by the LDH kit, and the results showed that the cytotoxicity level of the sDSS1 protein group was significantly decreased (Fig. 4C). On mouse primary neuronal cells, the sDSS1 protein also showed a shielding effect on the toxicity of Aβ oligomers, the cell viability of neurons was protected by sDSS1 protein (Fig. 5A), and the level of cytotoxicity was decreased (Fig. 5B). These results show that the sDSS1 protein can significantly block the cytotoxicity caused by Aβ oligomer proteins.
实施例5、sDSS1蛋白促进小胶质细胞清除Aβ蛋白并减少小胶质细胞激活。Example 5. The sDSS1 protein promotes microglia clearance of A[beta] protein and reduces microglial activation.
实验方法experimental method
1.细胞培养 小鼠小胶质细胞(BV2)购自国家实验细胞资源共享平台(3111C0001CCC000063)。BV2细胞培养在含10%FBS的DMEM培养液中,每两天传代一次。1. Cell Culture Mouse microglia (BV2) was purchased from the National Experimental Cell Resource Sharing Platform (3111C0001CCC000063). BV2 cells were cultured in DMEM medium containing 10% FBS and subcultured every two days.
2.Aβ清除实验 BV2细胞按照3ⅹ10 5每孔接种到6孔板中,细胞贴壁12小时。Aβ蛋白预先用DMEM完全培养液稀释成1μM溶液。阴性对照组不加蛋白,阳性对照组加入1μM Aβ蛋白溶液,实验 组除了1μM Aβ蛋白,分别包含0.5μM、2μM sDSS1蛋白。处理24小时后,收集上清液500μL,100g离心10分钟,取上清液用于ELISA实验检测Aβ1-42水平。离心收集所有细胞,裂解液后12000g离心收取裂解上清液用于ELISA检测Aβ1-42水平。细胞裂解液用BCA蛋白定量试剂盒(ThermoFisher,23252)进行蛋白浓度定量。 2. Aβ clearance assay BV2 cells were seeded into 6-well plates at 3x10 5 per well and cells were adhered for 12 hours. The Aβ protein was previously diluted with DMEM complete medium to a 1 μM solution. The negative control group was not added with protein, and the positive control group was added with 1 μM Aβ protein solution. The experimental group contained 0.5 μM and 2 μM sDSS1 protein except 1 μM Aβ protein. After 24 hours of treatment, 500 μL of the supernatant was collected, centrifuged at 100 g for 10 minutes, and the supernatant was taken for ELISA assay to detect Aβ 1-42 levels. All cells were collected by centrifugation, and the lysate was centrifuged at 12000 g to collect the lysate supernatant for ELISA to detect A? 1-42 levels. The cell lysate was quantified by protein concentration using a BCA protein quantification kit (ThermoFisher, 23252).
3.BV2细胞激活实验 BV2细胞按照1.5ⅹ10 5每孔接种到6孔板中,12小时后换成无血清的DMEM培养基,细胞饥饿处理24小时。Aβ 1-42寡聚物用无血清DMEM稀释成20μM溶液。阴性对照组不加蛋白,阳性对照组加入20μM Aβ蛋白溶液,实验组除了20μM Aβ蛋白,分别包含10μM、20μM sDSS1蛋白。细胞处理24小时后,收集上清液,100g离心10分钟取上清液用于ELISA实验检测溶液TNFα水平。收集所有细胞用于ROS水平检测。 3.BV2 cell activation experiments BV2 cells were seeded per well in accordance 1.5ⅹ10 5 to 6 well plates, DMEM medium was changed to serum-free 12 hours, cells were starved for 24 hours. The Aβ 1-42 oligomer was diluted to a 20 μM solution in serum-free DMEM. The negative control group was not added with protein, and the positive control group was added with 20 μM Aβ protein solution. The experimental group contained 10 μM and 20 μM sDSS1 protein except 20 μM Aβ protein. After the cells were treated for 24 hours, the supernatant was collected, centrifuged at 100 g for 10 minutes, and the supernatant was taken for ELISA assay to detect the TNFα level of the solution. All cells were collected for ROS level detection.
4.ELISA检测 将上述Aβ清除实验中获得培养液用于ELISA实验中,培养液样品用试剂盒提供的标准品蛋白稀释液(Standard diluent buffer)稀释50倍用于后续检测。按ELISA试剂盒(Invitrogen,Human Aβ42 ELISA kit,KHB3441)说明书要求,依次完成标准品蛋白和待测线虫蛋白孵育和清洗,加入显色底物孵育20分钟,加入终止液,在酶标仪上检测450nm吸光度值。根据标准曲线计算出蛋白样品中的Aβ蛋白浓度。4. ELISA assay The culture solution obtained in the above Aβ scavenging experiment was used in an ELISA experiment, and the culture solution sample was diluted 50 times with a standard diluent buffer provided by the kit for subsequent detection. According to the instructions of ELISA kit (Invitrogen, Human Aβ42 ELISA kit, KHB3441), the standard protein and the nematode protein to be tested were sequentially incubated and washed, and the chromogenic substrate was added for 20 minutes. The stop solution was added and detected on the microplate reader. 450 nm absorbance value. The Aβ protein concentration in the protein sample was calculated from the standard curve.
5.ROS水平检测活性氧检测试剂盒购自碧云天生物科技有限公司(S0033)。阳性对照细胞预先在37℃用1μM Rosup处理20分钟。取300μL细胞悬液,100g离心5分钟,加PBS清洗细胞两次。加入300μL无血清DMEM稀释的荧光探针(DCFDA)(1:1000),37℃孵育20分钟。100g离心5分钟,用PBS清洗细胞三次,用流式细胞仪(Millipore,guava Easycyte)检测。绿色荧光(488nm)强度反映了细胞ROS水平。5. ROS level detection Active oxygen detection kit was purchased from Biyuntian Biotechnology Co., Ltd. (S0033). Positive control cells were pretreated with 1 μM Rosup for 20 minutes at 37 °C. 300 μL of the cell suspension was taken, centrifuged at 100 g for 5 minutes, and the cells were washed twice with PBS. 300 μL of serum-free DMEM-diluted fluorescent probe (DCFDA) (1:1000) was added and incubated at 37 ° C for 20 minutes. After centrifugation at 100 g for 5 minutes, the cells were washed three times with PBS and detected by flow cytometry (Millipore, guava Easycyte). Green fluorescence (488 nm) intensity reflects cellular ROS levels.
6.TNFα水平检测 BV2细胞激活实验中收集的上清液用试剂盒提供的Standard Dilute buffer稀释10倍用于ELISA检测。按 ELISA试剂盒(Invitrogen,TNF alpha Mouse ELISA Kit,KMC3011C)说明书要求,依次完成标准品蛋白和待测线虫蛋白孵育和清洗,加入显色底物孵育20分钟,加入终止液,在酶标仪上检测450nm吸光度值。根据标准曲线计算出上清液中的TNFα浓度。6. Detection of TNFα level The supernatant collected in the BV2 cell activation experiment was diluted 10-fold with Standard Dilute buffer provided in the kit for ELISA detection. According to the instructions of ELISA kit (Invitrogen, TNF alpha Mouse ELISA Kit, KMC3011C), the standard protein and the nematode protein to be tested were sequentially incubated and washed, incubated with chromogenic substrate for 20 minutes, and the stop solution was added to the microplate reader. The 450 nm absorbance value was measured. The concentration of TNFα in the supernatant was calculated from the standard curve.
实验结果Experimental result
小胶质细胞是神经组织中清除Aβ蛋白等毒性蛋白的主要细胞。为了在检测sDSS1蛋白能否帮助小胶质细胞清除Aβ蛋白,在培养的小鼠小胶质细胞中加入1μM Aβ1-42蛋白和不同浓度的sDSS1蛋白。结果显示,小胶质细胞胞浆中检测到Aβ蛋白,随着sDSS1蛋白加入,细胞胞浆Aβ蛋白浓度显著提高(图6A)。相应的,培养液中Aβ蛋白残留减少,随着溶液中sDSS1蛋白浓度增加,Aβ蛋白浓度越来越低(图6B)。这些结果说明sDSS1蛋白帮助小胶质细胞清除Aβ蛋白。在培养液中加入高浓度Aβ蛋白寡聚物并加入不同浓度的sDSS1蛋白,可以看到培养液中的sDSS1蛋白能显著降低激活的小胶质细胞数量,细胞ROS水平显著降低(图6C),炎症因子TNF-α释放随着sDSS1蛋白浓度增加逐渐降低(图6D)。这些结果说明,sDSS1蛋白能刺激小胶质细胞清除Aβ蛋白并屏蔽Aβ寡聚物对小胶质细胞的影响。Microglia are the main cells in nerve tissue that remove toxic proteins such as Aβ protein. In order to detect whether sDSS1 protein can help microglia clear Aβ protein, 1 μM Aβ1-42 protein and different concentrations of sDSS1 protein were added to cultured mouse microglia. The results showed that Aβ protein was detected in the cytoplasm of microglia, and the concentration of cytosolic Aβ protein was significantly increased with the addition of sDSS1 protein (Fig. 6A). Correspondingly, the Aβ protein residue in the culture solution decreased, and the Aβ protein concentration became lower and lower as the concentration of sDSS1 protein in the solution increased (Fig. 6B). These results indicate that the sDSS1 protein helps microglia clear Aβ protein. Adding high concentration of Aβ protein oligomers to the culture medium and adding different concentrations of sDSS1 protein, it can be seen that the sDSS1 protein in the culture medium can significantly reduce the number of activated microglia, and the cell ROS level is significantly reduced (Fig. 6C). The release of the inflammatory factor TNF-α gradually decreased as the concentration of sDSS1 protein increased (Fig. 6D). These results indicate that sDSS1 protein can stimulate microglia to clear Aβ protein and block the effect of Aβ oligomer on microglia.
实施例6、sDSS1蛋白降低AD模型线虫的疾病症状。Example 6. The sDSS1 protein reduces disease symptoms in the AD model nematode.
实验方法experimental method
1.AD模型线虫瘫痪检测 线虫实验平台由上海南方模式生物科技股份有限公司提供,AD线虫模型由线虫实验平台提供。首先将成年CL4176线虫挑入固体培养基中,放入16℃培养箱,培养2小时后,挑出全部成虫,将虫卵放入16℃培养箱。48小时后,培养皿转入25℃培养箱中继续培养并分别加入不同浓度的sDSS1蛋白药物处理,对照组0μg/ml,低剂量组500μg/ml,高剂量组1000μg/ml。药物连续处理18小时后,开始统计CL4167线虫瘫痪个数,每2 小时统计一次,统计完毕后作图分析。线虫瘫痪标准是金属丝轻触线虫时,线虫身体僵直不能活动。1. AD model nematode detection The nematode experimental platform was provided by Shanghai Southern Model Biotechnology Co., Ltd., and the AD nematode model was provided by the nematode experimental platform. First, the adult CL4176 nematode was picked into a solid medium and placed in a 16 ° C incubator. After 2 hours of culture, all the adults were picked and the eggs were placed in a 16 ° C incubator. After 48 hours, the culture dishes were transferred to a 25 ° C incubator for further culture and treated with different concentrations of sDSS1 protein, 0 μg/ml in the control group, 500 μg/ml in the low dose group, and 1000 μg/ml in the high dose group. After 18 hours of continuous drug treatment, the number of CL4167 nematodes was counted and counted every 2 hours. After the statistics were completed, the analysis was performed. The standard for nematodes is that when the wire is lightly nematode, the nematode is stiff and incapable of moving.
2.线虫蛋白样品收集 CL4176线虫在不同浓度药物中处理20小时,完成后收集线虫,每组400条线虫。线虫用PBS清洗三遍并盛装在1.5mL离心管中,每管加入150μL线虫裂解液(线虫裂解液含62mM Tris-HCl,2%SDS,10%甘油,4%β-巯基乙醇,cocktail蛋白酶抑制剂),液氮冻融2次,60Hz超声5分钟,12000rpm离心10分钟,取上清液即为线虫总蛋白。线虫蛋白加入上样缓冲液(Loading buffer)并在100℃加热处理10分钟,制成可用于Wetern blotting分析的蛋白样品。2. Collection of nematode protein samples CL4176 nematodes were treated for 20 hours in different concentrations of drugs, and nematodes were collected after completion, with 400 nematodes per group. The nematodes were washed three times with PBS and placed in a 1.5 mL centrifuge tube. 150 μL of nematode lysate was added to each tube (C. elegans lysate containing 62 mM Tris-HCl, 2% SDS, 10% glycerol, 4% β-mercaptoethanol, cocktail protease inhibition) Agent), liquid nitrogen freeze-thaw 2 times, 60Hz ultrasound for 5 minutes, centrifugation at 12000rpm for 10 minutes, take the supernatant is the total protein of nematodes. The nematode protein was added to a loading buffer and heat treated at 100 ° C for 10 minutes to prepare a protein sample for Wetern blotting analysis.
3.Western Blotting 线虫蛋白样品用SDS-PAGE电泳进行分离,电泳条件为:10%变性预制胶胶(C#NP0321BOX,购自Life technology公司),MES电泳液,200V,30分钟。电泳完毕后,进行转膜,转膜条件为:100V恒压湿转60分钟,PVDF膜。完成的膜首先用5%脱脂奶粉(溶解在PBST中)室温封闭1小时,然后依次用兔来源Aβ抗体稀释液4℃孵育过夜(1:3000稀释在封闭液中),PBST清洗三次,HRP耦联驴来源抗兔抗体稀释液室温孵育1小时(1:5000稀释到PBST中)。ECL显色后压片并拍照保存。内参采用tubulin蛋白(抗体购自碧云天生物技术有限公司,AF0001)。3. Western Blotting The nematode protein samples were separated by SDS-PAGE electrophoresis. The electrophoresis conditions were: 10% denatured pre-formed gel (C#NP0321BOX, purchased from Life Technology), MES electrophoresis solution, 200V, 30 minutes. After the electrophoresis was completed, the film was transferred, and the film transfer conditions were: 100 V constant pressure wet rotation for 60 minutes, PVDF film. The completed membrane was first blocked with 5% skim milk powder (dissolved in PBST) for 1 hour at room temperature, then sequentially incubated with rabbit-derived Aβ antibody dilution at 4 ° C overnight (1:3000 dilution in blocking solution), washed three times with PBST, HRP coupling The conjugated anti-rabbit antibody dilution was incubated for 1 hour at room temperature (1:5000 dilution into PBST). After the ECL color is developed, the tablet is compressed and photographed and saved. The internal reference used tubulin protein (antibody was purchased from Biyuntian Biotechnology Co., Ltd., AF0001).
4.ELISA检测 将上述Western blotting实验中提取的线虫总蛋白用于ELISA实验中,蛋白样品用试剂盒提供的标准品蛋白稀释液(Standard diluent buffer)稀释三倍用于后续检测,蛋白稀释液加入体积根据Western blotting实验中内参的数据进行调整,保证加入的总蛋白量一致。按ELISA试剂盒说明书要求,依次完成标准品蛋白和待测线虫蛋白孵育和清洗,加入显色底物孵育20分钟,加入终止液,在酶标仪上检测450nm吸光度值。根据标准曲线计算出蛋白样品中的Aβ蛋白浓度。4. ELISA detection The total nematode protein extracted from the above Western blotting experiment was used in the ELISA experiment, and the protein sample was diluted three times with the standard diluent buffer provided by the kit for subsequent detection, and the protein dilution was added. The volume was adjusted according to the data of the internal reference in the Western blotting experiment to ensure that the total amount of protein added was consistent. According to the instructions of the ELISA kit, the standard protein and the nematode protein to be tested were sequentially incubated and washed, incubated with a chromogenic substrate for 20 minutes, and the stop solution was added to measure the absorbance at 450 nm on a microplate reader. The Aβ protein concentration in the protein sample was calculated from the standard curve.
5.线虫免疫组织化学染色 CL4176线虫在不同浓度药物中处理20小时,完成后收集线虫,每组50-100条线虫。线虫用PBS清洗一遍并盛装在1.5mL离心管中,弃去上清后加入4%多聚甲醛(溶解在PBS中,pH7.2)4℃条件下固定过夜。固定好的线虫首先用清洗缓冲液(包含10mm Tris/HCl,1%Triton-X100,1mM EDTA,pH7.4)冲洗一遍。线虫依次用继续用含1%β巯基乙醇的清洗液室温孵育1小时,硼酸缓冲液(包含25mM H 3BO 3,12.5mM NaOH,10mM DTT,pH9.2)室温孵育1慢慢5分钟,含1%H 2O 2的清洗缓冲液室温孵育15分钟。完成后,线虫用清洗缓冲液清洗一遍,加入封闭液(包含1%BSA、0.5%Triton-X100,1mM EDTA,PBS)室温封闭两个小时。处理好的线虫依次经过一抗稀释液(Aβ抗体,1:100稀释到封闭液中)4℃孵育过夜,二抗稀释液(按照1:1000比例添加Hoechst)(Donkey-anti-Rabbit-Alex Fluor 488(购自Life Technology公司,A-11008)按照1:5000稀释在PBST中)室温孵育1小时。PBST清洗三遍,完成后封片并用Confocol激光共聚焦显微镜(Nikon,Nikon C1Plus)观察,拍照分析。 5. C. elegans immunohistochemical staining CL4176 nematodes were treated in different concentrations of drugs for 20 hours, and after completion, nematodes were collected, with 50-100 nematodes in each group. The nematodes were washed once with PBS and placed in a 1.5 mL centrifuge tube, and the supernatant was discarded, and 4% paraformaldehyde (dissolved in PBS, pH 7.2) was added at 4 ° C overnight. The fixed nematode was first rinsed once with wash buffer (containing 10 mm Tris/HCl, 1% Triton-X100, 1 mM EDTA, pH 7.4). The nematodes were sequentially incubated with a washing solution containing 1% β-mercaptoethanol for 1 hour at room temperature, boric acid buffer (containing 25 mM H 3 BO 3 , 12.5 mM NaOH, 10 mM DTT, pH 9.2) was incubated for 1 minute at room temperature for 5 minutes. 1% H 2 O 2 wash buffer was incubated for 15 minutes at room temperature. After completion, the nematodes were washed once with wash buffer and blocked by adding blocking solution (containing 1% BSA, 0.5% Triton-X100, 1 mM EDTA, PBS) for two hours at room temperature. The treated nematodes were sequentially incubated overnight at 4 °C with a primary antibody dilution (Aβ antibody, diluted 1:100 in blocking solution), and the secondary antibody dilution (added Hoechst at a ratio of 1:1000) (Donkey-anti-Rabbit-Alex Fluor) 488 (purchased from Life Technology, Inc., A-11008) was incubated for 1 hour at room temperature with a dilution of 1:5000 in PBST. The PBST was washed three times, and after completion, it was mounted and observed with a Confocol laser confocal microscope (Nikon, Nikon C1Plus), and photographed.
实验结果Experimental result
CL4176线虫作为AD动物模型用于检测急性Aβ 1-42蛋白蓄积引起的毒性,被用于药物评价。在线虫瘫痪实验中,加入不同浓度的sDSS1蛋白可以显著降低线虫瘫痪水平,且效应呈现出显著的浓度依赖(图7A)。在43小时后检测,对照组未瘫痪线虫的比例低于1%,而给药组仍旧有18.42%(500μg/mL sDSS1蛋白)和48.57%(1000μg/mL sDSS1蛋白)的线虫表现正常。总体分析,半数线虫发生瘫痪的中位时间分别是对照组22小时、低浓度给药组24小时、高浓度给药组42小时。这些结果说明,sDSS1蛋白能有效降低AD模型线虫的疾病症状。利用免疫组织化学检测线虫体内Aβ沉积,可以在对照线虫体内看到明显的颗粒状聚集体,而sDSS1蛋白给药组没有检测到聚集体或者检测到很少的颗粒,说明sDSS1蛋白减少 了Aβ聚集物形成(图7B)。利用Western blotting实验检测线虫研磨液中Aβ蛋白含量,结果同样表明,sDSS1蛋白减少了Aβ蛋白多聚体的含量(图7C)。用ELISA实验检测组织裂解液中Aβ 1-42蛋白含量,可以看到sDSS1蛋白处理的线虫中Aβ蛋白显降低,1000μg/mL sDSS1蛋白处理组Aβ蛋白量仅是对照组的59.09%(图7D)。通过以上实验,我们在AD线虫模型上验证sDSS1蛋白可以降低Aβ 1-42聚集体形成,减少组织Aβ蓄积,缓解AD模型线虫的疾病症状。CL4176 nematode was used as an animal model for AD to detect toxicity caused by accumulation of acute Aβ 1-42 protein and was used for drug evaluation. In the linear worm experiment, the addition of different concentrations of sDSS1 protein significantly reduced the nematode sputum level, and the effect showed a significant concentration dependence (Fig. 7A). After 43 hours, the proportion of the non-nematode in the control group was less than 1%, while the nematodes with 18.42% (500 μg/mL sDSS1 protein) and 48.57% (1000 μg/mL sDSS1 protein) in the administration group were normal. In the overall analysis, the median time of occurrence of sputum in half of the nematodes was 22 hours in the control group, 24 hours in the low concentration group, and 42 hours in the high concentration group. These results indicate that the sDSS1 protein is effective in reducing the disease symptoms of the AD model nematode. Immunohistochemistry was used to detect Aβ deposition in nematodes, and obvious granular aggregates were observed in the control nematodes. However, no aggregates were detected or few particles were detected in the sDSS1 protein administration group, indicating that sDSS1 protein reduced Aβ aggregation. Object formation (Fig. 7B). Western blotting assay was used to detect the Aβ protein content in nematode grinding solution. The results also showed that sDSS1 protein reduced the content of Aβ protein multimer (Fig. 7C). The Aβ 1-42 protein content in the tissue lysate was detected by ELISA. It can be seen that the Aβ protein in the sDSS1 protein-treated nematode was decreased, and the Aβ protein content in the 1000 μg/mL sDSS1 protein treatment group was only 59.09% of the control group (Fig. 7D). . Through the above experiments, we verified that sDSS1 protein can reduce the formation of Aβ 1-42 aggregates, reduce the accumulation of tissue Aβ, and alleviate the disease symptoms of AD model nematodes in the AD nematode model.
实施例7、sDSS1蛋白加速小鼠体内Aβ蛋白的清除。Example 7. The sDSS1 protein accelerates clearance of Aβ protein in mice.
实验方法experimental method
1.APP/PS1小鼠血液Aβ清除实验 APP/PS1小鼠(2月龄,雄性)购自南京大学模式动物中心。APP/PS1小鼠从8月龄开始,按照25mg/kg体重连续尾静脉注射sDSS1蛋白8周,完成后处死小鼠,取血并离心取血浆用于ELISA检测血液中Aβ 1-42水平。1. APP/PS1 mouse blood Aβ clearance test APP/PS1 mice (2 months old, male) were purchased from the Model Animal Center of Nanjing University. The APP/PS1 mice were injected with sDSS1 protein at a dose of 25 mg/kg body weight for 8 weeks from the age of 8 months. After completion, the mice were sacrificed, blood was taken and centrifuged to obtain plasma for ELISA to detect Aβ 1-42 levels in the blood.
2.Aβ蛋白急性清除实验 ICR小鼠(6-8周龄,雄性)购自上海斯莱克实验动物有限责任公司。小鼠根据体重分成四组,包括阴性对照组(注射生理盐水)、阳性对照组(Aβ蛋白单独给药)、低剂量给药组(Aβ给药和5mg/kg体重注射sDSS1蛋白)、高剂量给药组(Aβ给药和30mg/kg体重注射sDSS1蛋白)。实验开始时,小鼠首先根据分组尾静脉注射sDSS1蛋白,阴性对照组和阳性对照组注射等量PBS,1小时后,阳性对照组和给药组按照10mg/kg体重尾静脉注射Aβ蛋白,阴性对照组注射等量生理盐水。注射完成后,小鼠正常饲养,2小时后处死小鼠,心脏取血,3500g离心20分钟取血浆用于ELISA检测Aβ1-42水平。2. Aβ protein acute clearance test ICR mice (6-8 weeks old, male) were purchased from Shanghai Slack Laboratory Animals Co., Ltd. The mice were divided into four groups according to their body weight, including a negative control group (injected with normal saline), a positive control group (administered with Aβ protein alone), a low-dose group (Aβ-administered and 5 mg/kg body weight injected with sDSS1 protein), and high dose. The administration group (Aβ administration and 30 mg/kg body weight injection of sDSS1 protein). At the beginning of the experiment, the mice were first injected with sDSS1 protein according to the tail vein of the group. The negative control group and the positive control group were injected with the same amount of PBS. After 1 hour, the positive control group and the drug-administered group were injected with Aβ protein at the tail vein of 10 mg/kg body weight. The control group was injected with the same amount of normal saline. After the injection was completed, the mice were normally reared, and the mice were sacrificed 2 hours later. Blood was taken from the heart, and the cells were centrifuged at 3,500 g for 20 minutes to obtain plasma for ELISA to detect A? 1-42 levels.
3.ELISA检测 将上述实验中获得的血浆样品(必要时稀释1-3倍)用于ELISA实验。按ELISA试剂盒说明书要求,依次完成标准品蛋白和待测血浆样品的孵育和清洗,加入显色底物孵育20分钟, 加入终止液,在酶标仪上检测450nm吸光度值。根据标准曲线计算出血浆中的Aβ蛋白浓度。3. ELISA test Plasma samples obtained in the above experiment (diluted 1-3 times if necessary) were used for the ELISA experiment. According to the instructions of the ELISA kit, the standard protein and the plasma sample to be tested are sequentially incubated and washed, and the chromogenic substrate is added for 20 minutes, the stop solution is added, and the 450 nm absorbance value is detected on the microplate reader. The Aβ protein concentration in plasma was calculated from the standard curve.
实验结果Experimental result
为了检测sDSS1蛋白能否帮助清除Aβ蛋白,我们分别采用急性Aβ水平增加模型和慢性Aβ水平增加模型。在急性模型中,通过尾静脉注射高浓度Aβ1-42蛋白,同时注射sDSS1蛋白。实验结果显示,不同浓度的sDSS1蛋白给药都能显著降低血液Aβ蛋白水平,并且该效应呈现出一定的剂量依赖性(图8A)。在慢性模型中,给AD模型APP/PS1小鼠长时间尾静脉注射sDSS1蛋白,结果发现sDSS1蛋白同样能减少血液Aβ蛋白的水平(图8B)。这两个结果说明sDSS1蛋白能帮助清除小鼠血液中Aβ蛋白。To test whether sDSS1 protein can help clear Aβ protein, we used an acute Aβ level increase model and a chronic Aβ level increase model, respectively. In the acute model, high concentrations of Aβ 1-42 protein were injected through the tail vein while the sDSS1 protein was injected. The results of the experiment showed that different concentrations of sDSS1 protein administration significantly reduced blood Aβ protein levels, and the effect was dose-dependent (Fig. 8A). In the chronic model, AD model APP/PS1 mice were injected with sDSS1 protein for a long time in the tail vein, and it was found that sDSS1 protein also reduced blood Aβ protein levels (Fig. 8B). These two results indicate that sDSS1 protein can help clear Aβ protein in the blood of mice.
实施例8、sDSS1蛋白降低AD模型小鼠脑组织斑块沉积。Example 8. sDSS1 protein reduces plaque deposition in brain tissue of AD model mice.
实验方法experimental method
1.AD模型小鼠给药 APP/PS1小鼠(2月龄,雄性)购自南京大学模式动物中心。APP/PS1小鼠从9月龄开始,按照50mg/kg体重连续尾静脉注射sDSS1蛋白8周,完成后小鼠用于行为学实验检测。行为学实验完成后,小鼠处死,取血浆用于血液指标检测,取脑组织并在4%多聚甲醛固定用于免疫组织化学染色分析斑块沉积水平。1. AD model mice administration APP/PS1 mice (2 months old, male) were purchased from the Model Animal Center of Nanjing University. The APP/PS1 mice were injected with sDSS1 protein at a dose of 50 mg/kg body weight for 8 weeks from the age of 9 months. After completion, the mice were used for behavioral experiments. After the behavioral experiment was completed, the mice were sacrificed, and plasma was taken for blood index detection. Brain tissue was taken and fixed in 4% paraformaldehyde for immunohistochemical staining to analyze plaque deposition levels.
2.免疫组织化学实验 处理完成的小鼠依次用50m生理盐水和50m预冷的4%多聚甲醛溶液进行心脏灌注,完成后取脑组织浸泡在4%多聚甲醛中固定2天,完成后用30%蔗糖溶液脱水并用包埋剂包埋制成组织块。脑组织用冰冻切片机切成10um厚的脑片用于后续免疫组织化学分析。脑片首先用PBS清洗一遍除去包埋剂,然后用FSB工作液(Dojindo,F308)(用50%乙醇溶液稀释成0.05%工作液)孵育30分钟,完成后切片浸泡到饱和碳酸锂溶液中,最后用50%乙醇溶液清洗一遍。切片用封片剂封片并用荧光显微镜观察,拍照分析淀粉样沉淀斑块数量和大小。2. Immunohistochemistry experiment The treated mice were perfused with 50m normal saline and 50m pre-cooled 4% paraformaldehyde solution. After completion, the brain tissue was immersed in 4% paraformaldehyde for 2 days. The tissue block was prepared by dehydration with a 30% sucrose solution and embedding with an embedding agent. Brain tissue was cut into 10 um thick brain slices using a cryostat for subsequent immunohistochemical analysis. The brain piece was firstly washed with PBS to remove the embedding agent, and then incubated with FSB working solution (Dojindo, F308) (diluted to 0.05% working solution with 50% ethanol solution) for 30 minutes, and then the slice was immersed in a saturated lithium carbonate solution. Finally, it was washed once with 50% ethanol solution. The sections were mounted with a sealing tablet and observed with a fluorescence microscope, and the number and size of amyloid precipitated patches were photographed.
实验结果Experimental result
AD模型小鼠给药8周后,脑片经染色观察组织中Aβ斑块沉积水平。结果显示,在对照组11月龄APP/PS1sDSS1小鼠的脑组织中检测到大小不一的斑块,但是sDSS1蛋白给药8周之后,组织中斑块沉积显著降低(图9A)。利用photoshop CS软件统计斑块面积,结果给药组斑块面积仅是对照组三分之一(图9B)。根据斑块大小统计斑块数量,结果看到给药组小鼠脑组织中型斑块数量显著降低,几乎找不到大型斑块沉积(图9C)。这些结果说明,sDSS1蛋白给药能显著降低AD模型小鼠脑组织Aβ蛋白沉积。Eight weeks after administration of AD model mice, brain slices were stained to observe the level of Aβ plaque deposition in the tissues. The results showed that plaques of different sizes were detected in the brain tissue of the 11-month-old APP/PS1sDSS1 mice in the control group, but plaque deposition was significantly reduced in the tissues after 8 weeks of administration of the sDSS1 protein (Fig. 9A). The plaque area was counted using photoshop CS software, and the plaque area of the drug-administered group was only one-third of the control group (Fig. 9B). The number of plaques was counted based on the plaque size, and it was found that the number of plaques in the brain tissue of the mice in the administration group was significantly reduced, and large plaque deposits were hardly found (Fig. 9C). These results indicate that sDSS1 protein administration can significantly reduce Aβ protein deposition in brain tissue of AD model mice.

Claims (34)

  1. 一种蛋白在制备预防和治疗痴呆症的药物中的应用,其特征在于,所述的应用是把sDSS1蛋白用于制备痴呆症预防药物和治疗药物。Use of a protein for the preparation of a medicament for preventing and treating dementia, characterized in that the application is to use the sDSS1 protein for the preparation of a dementia prevention drug and a therapeutic drug.
  2. 根据权利要求1所述的应用,其特征在于,所述的痴呆症是指退行性痴呆症,包括早老性痴呆症、老年性痴呆症、路易氏体型痴呆症、额颞叶型痴呆症。The use according to claim 1, wherein the dementia refers to degenerative dementia, including Alzheimer's disease, Alzheimer's disease, Louis deer type dementia, and frontotemporal dementia.
  3. 根据权利要求1所述的应用,其特征在于,所述的痴呆症是血管性痴呆症。The use according to claim 1, wherein the dementia is vascular dementia.
  4. 根据权利要求1所述的应用,其特征在于,所述的痴呆症是痴呆前期痴呆症、轻度痴呆期痴呆症、中度痴呆期痴呆症、重度痴呆期痴呆症。The application according to claim 1, wherein the dementia is pre-dementia dementia, mild dementia dementia, moderate dementia dementia, and severe dementia dementia.
  5. 根据权利要求1所述的应用,其特征在于,所述的sDSS1蛋白包括人、黑猩猩、倭黑猩猩、大猩猩、红毛猩猩、白颊长臂猿、川金丝猴、恒河猴、滇金丝猴、东非狒狒、安哥拉疣猴、白顶白眉猴、鬼狒、豚尾猴中的任一sDSS1蛋白序列形成的基础蛋白,其中人sDSS1的氨基酸序列如SEQ ID NO:1,黑猩猩sDSS1的氨基酸序列如SEQ ID NO:2,倭黑猩猩sDSS1的氨基酸序列如SEQ ID NO:3,大猩猩sDSS1的氨基酸序列如SEQ ID NO:4,红毛猩猩sDSS1的氨基酸序列如SEQ ID NO:5,白颊长臂猿sDSS1的氨基酸序列如SEQ ID NO:6,川金丝猴sDSS1的氨基酸序列如SEQ ID NO:7,恒河猴sDSS1的氨基酸序列如SEQ ID NO:8,滇金丝猴sDSS1的氨基酸序列如SEQ ID NO:9,东非狒狒sDSS1的氨基酸序列如SEQ ID NO:10,安哥拉疣猴sDSS1的氨基酸序列如SEQ ID NO:11,白顶白眉猴sDSS1的氨基酸序列如SEQ ID NO:12,鬼狒sDSS1的氨基酸序列如SEQ ID NO:13,豚尾猴sDSS1的氨基酸序列如SEQ ID NO:14。The use according to claim 1, wherein the sDSS1 protein comprises a human, a chimpanzee, a bonobo, a gorilla, an orangutan, a white-cheeked gibbon, a Rhesus monkey, a rhesus monkey, a golden monkey, an East African ape, A basic protein formed by any of the sDSS1 protein sequences in Angora marmoset, white-tailed white-browed macaque, podophyllum, and porpoise monkey, wherein the amino acid sequence of human sDSS1 is SEQ ID NO: 1, and the amino acid sequence of chimpanzee sDSS1 is SEQ ID NO: 2. The amino acid sequence of the chimpanzee sDSS1 is SEQ ID NO: 3, the amino acid sequence of the gorilla sDSS1 is SEQ ID NO: 4, the amino acid sequence of the orangutan sDSS1 is SEQ ID NO: 5, and the amino acid sequence of the white-cheeked gibbon sDSS1 is as SEQ ID NO: 6, the amino acid sequence of Rhinopithecus sDSS1 is SEQ ID NO: 7, the amino acid sequence of rhesus sDSS1 is SEQ ID NO: 8, and the amino acid sequence of Rhinopithecus sDSS1 is SEQ ID NO: 9, East Africa 狒狒 sDSS1 The amino acid sequence of SEQ ID NO: 10, the amino acid sequence of Angora simian sDSS1 is SEQ ID NO: 11, and the amino acid sequence of leucocephalus sDSS1 is SEQ ID NO: 12, sneaky s The amino acid sequence of DSS1 is SEQ ID NO: 13, and the amino acid sequence of porpoise monkey sDSS1 is SEQ ID NO: 14.
  6. 根据权利要求5所述的应用,其特征在于,所述的sDSS1蛋白是与所述的基础蛋白相似度达到70%以上的第一种蛋白。The use according to claim 5, wherein said sDSS1 protein is the first protein having a similarity to said basic protein of 70% or more.
  7. 根据权利要求5所述的应用,其特征在于,所述的sDSS1蛋白是以所述的基础蛋白的氮端58个氨基酸为基础,在氮端或碳端融合其他多肽片段,用于融合的多肽片段的结构特征或氨基酸序列特征与所述的基础蛋白的碳端31个序列相同或相似的第二种蛋白。The use according to claim 5, wherein the sDSS1 protein is based on 58 amino acids of the nitrogen terminus of the basic protein, and the other polypeptide fragments are fused at the nitrogen terminal or the carbon terminal, and the polypeptide is used for fusion. A second protein having a structural feature or amino acid sequence characteristic of the fragment that is identical or similar to the carbon terminal 31 sequences of the basic protein.
  8. 根据权利要求5所述的应用,其特征在于,所述的sDSS1蛋白是以所述的基础蛋白的氮端58个氨基酸为基础,在氮端或碳端融合其他氨基酸片段,融合后的蛋白能实现跨膜转运功能的第三种蛋白。The use according to claim 5, wherein the sDSS1 protein is based on 58 amino acids of the nitrogen terminus of the basic protein, and the other amino acid fragments are fused at the nitrogen or carbon end, and the protein can be fused. A third protein that achieves transmembrane transport function.
  9. 根据权利要求5-8任一所述的应用,其特征在于,所述的sDSS1蛋白是利用所述的基础蛋白、第一种蛋白、第二种蛋白或第三种蛋白与该种蛋白自身、载体蛋白、抗体或其他任意长度氨基酸片段连接形成的融合蛋白。The use according to any one of claims 5-8, wherein the sDSS1 protein utilizes the basic protein, the first protein, the second protein or the third protein and the protein itself, A fusion protein formed by the joining of a carrier protein, antibody or other amino acid fragment of any length.
  10. 根据权利要求5-9任一所述的应用,其特征在于,所述的sDSS1蛋白是所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白进行的修饰产生的多肽/蛋白修饰物。The use according to any one of claims 5-9, wherein the sDSS1 protein is modified by the basic protein, the first protein, the second protein, the third protein or the fusion protein. Polypeptide/protein modification.
  11. 根据权利要求10所述的应用,其特征在于,所述多肽/蛋白修饰物是针对氨基酸侧链上的氨基、氨基酸侧链上的羰基、氮端末端氨基、碳端末端羰基、半胱氨酸、酪氨酸、丝氨酸、色氨酸进行的特异性或非特异性的1-20个位点的化学修饰。The use according to claim 10, wherein the polypeptide/protein modification is directed to an amino group on an amino acid side chain, a carbonyl group on an amino acid side chain, a nitrogen terminal amino group, a carbon terminal carbonyl group, or a cysteine. Specific or non-specific chemical modification of 1-20 sites by tyrosine, serine, tryptophan.
  12. 根据权利要求10所述的应用,其特征在于,所述多肽/蛋白修饰物的修饰方法包括糖基化修饰、脂肪酸修饰、酰基化修饰、Fc片段融合、白蛋白融合、聚乙二醇修饰、右旋糖苷修饰、肝素修饰、聚乙烯吡咯烷酮修饰、聚氨基酸修饰、多聚唾液酸修饰、壳聚糖及其衍生物修饰、凝集素修饰、海藻酸钠修饰、卡波姆修饰、聚乙烯吡咯烷酮修饰、羟丙基甲基纤维素修饰、羟丙基纤维素修饰、乙酰化修饰、 甲酰化修饰、磷酸化修饰、甲基化修饰、磺酸化修饰以及其他医药上可用的多肽/蛋白药物修饰方法的一种或一种以上。The method according to claim 10, wherein the modification method of the polypeptide/protein modification comprises glycosylation modification, fatty acid modification, acylation modification, Fc fragment fusion, albumin fusion, polyethylene glycol modification, Dextran modification, heparin modification, polyvinylpyrrolidone modification, polyamino acid modification, polysialic acid modification, chitosan and its derivative modification, lectin modification, sodium alginate modification, carbomer modification, polyvinylpyrrolidone modification , hydroxypropyl methylcellulose modification, hydroxypropyl cellulose modification, acetylation modification, formylation modification, phosphorylation modification, methylation modification, sulfonation modification, and other pharmaceutically useful polypeptide/protein drug modification methods One or more of them.
  13. 根据权利要求5-9所述的应用,其特征在于,所述的sDSS1蛋白是利用所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白的氨基酸序列为基础进行的20种基本氨基酸以外的氨基酸进行的1-31个任意氨基酸位点替换的非天然氨基酸替代蛋白。The use according to claims 5-9, characterized in that the sDSS1 protein is based on the amino acid sequence of the basic protein, the first protein, the second protein, the third protein or the fusion protein. A non-natural amino acid replacement protein substituted with 1-31 arbitrary amino acid positions of amino acids other than the 20 essential amino acids.
  14. 根据权利要求13所述的应用,其特征在于,所述的非天然氨基酸替代蛋白的氨基酸替换包括替换成羟脯氨酸、羟赖氨酸、硒代半胱氨酸、D-型氨基酸或者人工合成的非天然氨基酸及其衍生物。The use according to claim 13, wherein the amino acid substitution of the non-natural amino acid replacement protein comprises replacement with hydroxyproline, hydroxylysine, selenocysteine, D-type amino acid or artificial Synthetic unnatural amino acids and derivatives thereof.
  15. 根据权利要求5-14任一所述的应用,其特征在于,所述的sDSS1蛋白是把所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或非天然氨基酸替代蛋白与医药上可应用的药物载体形成的部分或全部复合体。The use according to any one of claims 5 to 14, wherein the sDSS1 protein is the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/ A partial or total complex of a protein modified or non-natural amino acid replacement protein with a pharmaceutically acceptable pharmaceutical carrier.
  16. 根据权利要求15所述的应用,其特征在于,所述复合体的药物载体包含肠溶衣制剂、胶囊、微球/囊、脂质体、微乳液、复乳液、纳米颗粒、磁颗粒、明胶和凝胶中的一种或一种以上。The use according to claim 15, wherein the pharmaceutical carrier of the complex comprises an enteric coating preparation, a capsule, a microsphere/capsule, a liposome, a microemulsion, a double emulsion, a nanoparticle, a magnetic particle, a gelatin And one or more of the gels.
  17. 根据权利要求5-16任一所述的应用,其特征在于,所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或复合体发挥屏蔽毒性蛋白毒性的起始工作浓度为不小于0.2μg/mL。The use according to any one of claims 5-16, wherein the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the complex are exerted. The initial working concentration for shielding toxic protein toxicity is not less than 0.2 μg/mL.
  18. 根据权利要求5-16任一所述的应用,其特征在于,所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物或复合体发挥屏蔽毒性蛋白毒性的摩尔浓度比率为不小于0.010;所述摩尔浓度比率是指反应体系中蛋白、多肽/蛋白修饰物或复合体的摩尔浓度与毒性蛋白摩尔浓度的比率。The use according to any one of claims 5-16, wherein the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/protein modification or the complex are exerted. The molar concentration ratio of the toxic protein toxicity is not less than 0.010; the molar ratio refers to the ratio of the molar concentration of the protein, the polypeptide/protein modification or the complex in the reaction system to the molar concentration of the toxic protein.
  19. 根据权利要求1所述的应用,其特征在于,所述的sDSS1蛋白是以个体自身sDSS1蛋白为靶点,通过外源药物影响个体自身sDSS1蛋白的水平。The use according to claim 1, wherein the sDSS1 protein targets the individual's own sDSS1 protein and affects the level of the individual's own sDSS1 protein by the exogenous drug.
  20. 根据权利要求19所述的应用,其特征在于,所述的药物是以sDSS1蛋白、sDSS1蛋白的基因、sDSS1的基因的调控元件或sDSS1的基因的转录产物为药物作用靶点。The use according to claim 19, wherein the drug is a drug target of a sDSS1 protein, a gene of sDSS1 protein, a regulatory element of a gene of sDSS1 or a gene of sDSS1.
  21. 根据权利要求19所述的应用,其特征在于,所述的药物是通过影响血液或脑脊液中蛋白酶/肽酶活性从而调节sDSS1蛋白在血液或脑脊液中的含量。The use according to claim 19, wherein the drug modulates the content of the sDSS1 protein in blood or cerebrospinal fluid by affecting protease/peptidase activity in blood or cerebrospinal fluid.
  22. 根据权利要求19-21任一所述的应用,其特征在于,所述的药物是化学小分子药物、抗体、多肽/蛋白药物、核酸药物或纳米药物形成的第一种药物。The use according to any one of claims 19-21, wherein the drug is a first drug formed by a chemical small molecule drug, an antibody, a polypeptide/protein drug, a nucleic acid drug or a nano drug.
  23. 根据权利要求5-22任一所述的应用,其特征在于,所述的sDSS1蛋白是以所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物、复合体、第一种药物中的任一一种成分的两种或多种的组合形成的第二种药物。The use according to any one of claims 5 to 22, wherein the sDSS1 protein is the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/ A second drug formed by a combination of two or more of any one of a protein modification, a complex, and a first drug.
  24. 根据权利要求5-22任一所述的应用,其特征在于,所述的sDSS1蛋白是以所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白、融合蛋白、多肽/蛋白修饰物、非天然氨基酸替代蛋白、复合体、第一种药物中的任一一种成分的一种、两种或多种与医药上可用的赋形剂的组合形成的第三种药物。The use according to any one of claims 5 to 22, wherein the sDSS1 protein is the basic protein, the first protein, the second protein, the third protein, the fusion protein, the polypeptide/ A third drug formed by a combination of a protein modification, a non-natural amino acid replacement protein, a complex, one of the first drugs, a combination of two or more, and a pharmaceutically acceptable excipient.
  25. 根据权利要求5-9任一所述的应用,其特征在于,所述的sDSS1蛋白是通过表达体系把编码所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白的核苷酸序列导入体内并表达获得的第四种蛋白。The use according to any one of claims 5-9, wherein the sDSS1 protein encodes the basic protein, the first protein, the second protein, the third protein or a fusion by an expression system. The nucleotide sequence of the protein is introduced into the body and the resulting fourth protein is expressed.
  26. 根据权利要求25所述的应用,其特征在于,所述的表达体系是真核表达质粒载体、腺病毒、腺相关病毒、慢病毒、逆转录病毒、 杆状病毒、疱疹病毒、伪狂犬病毒、ZFN基因编辑技术、TALEN基因编辑技术、CRISPR/Cas基因编辑技术或其他医疗上可用的基因编辑技术或病毒载体。The use according to claim 25, wherein the expression system is a eukaryotic expression plasmid vector, an adenovirus, an adeno-associated virus, a lentivirus, a retrovirus, a baculovirus, a herpes virus, a pseudorabies virus, ZFN gene editing technology, TALEN gene editing technology, CRISPR/Cas gene editing technology or other medically useful gene editing techniques or viral vectors.
  27. 根据权利要求5-9任一所述的应用,其特征在于,所述的sDSS1蛋白是通过移植细胞在个体体内获得的所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白形成的第五种蛋白。The use according to any one of claims 5-9, wherein the sDSS1 protein is the basic protein, the first protein, the second protein, and the third obtained in an individual by transplanting cells. The fifth protein formed by a protein or fusion protein.
  28. 根据权利要求27所述的应用,其特征在于,所述的细胞是任意一种人的干细胞、前体细胞或成体细胞。The use according to claim 27, wherein said cell is a stem cell, a precursor cell or an adult cell of any one of humans.
  29. 根据权利要求28所述的应用,其特征在于,所述的干细胞是胚胎干细胞、诱导多能干细胞、转分化得到的细胞,或者来源于原代培养的干细胞、由母细胞分化得到的多能或单能干细胞。The use according to claim 28, wherein the stem cells are embryonic stem cells, induced pluripotent stem cells, transdifferentiated cells, or derived from primary cultured stem cells, pluripotently differentiated from mother cells or Single energy stem cells.
  30. 根据权利要求5-9任一所述的应用,其特征在于,所述的sDSS1蛋白是通过移植组织或器官在个体体内获得的所述的基础蛋白、第一种蛋白、第二种蛋白、第三种蛋白或融合蛋白形成的第六种蛋白。The use according to any one of claims 5-9, wherein the sDSS1 protein is the basic protein, the first protein, the second protein, the first obtained in the individual by transplanting tissues or organs. The sixth protein formed by three proteins or fusion proteins.
  31. 根据权利要求30所述的应用,其特征在于,所述的组织是脑、肝、肾、脾、胰岛的完整器官或部分组织块,或血液、脂肪、肌肉、骨髓、皮肤。The use according to claim 30, wherein the tissue is a whole organ or part of a tissue block of the brain, liver, kidney, spleen, islet, or blood, fat, muscle, bone marrow, skin.
  32. 根据权利要求1所述的应用,其特征在于,所述的sDSS1蛋白是通过血清、脑脊液、组织间液输注引入个体体内的第七种蛋白。The use according to claim 1, wherein the sDSS1 protein is a seventh protein introduced into the body by serum, cerebrospinal fluid, and interstitial fluid infusion.
  33. 根据权利要求1-32任一所述的应用,其特征在于,所述的预防药物是包含基础蛋白、第一种至第七种任一蛋白、融合蛋白、多肽/蛋白修饰物、非天然氨基酸替代蛋白、复合体、第一种药物、第二种药物、第三种药物、在表达体系、细胞、组织、器官、体液、组织液的蛋白药物、多肽药物、核酸药物、化学小分子药物、细胞产品、商业化移植组织、注射液、冻干粉、保健品、食品添加剂中的一种或多种。The use according to any one of claims 1 to 32, wherein the prophylactic agent comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, and an unnatural amino acid. Substitute protein, complex, first drug, second drug, third drug, protein drug, polypeptide drug, nucleic acid drug, chemical small molecule drug, cell in expression system, cell, tissue, organ, body fluid, tissue fluid One or more of products, commercial transplant tissues, injections, lyophilized powder, health care products, food additives.
  34. 根据权利要求1-32任一所述的应用,其特征在于,所述的治疗药物是包含基础蛋白、第一种至第七种任一蛋白、融合蛋白、多肽/蛋白修饰物、非天然氨基酸替代蛋白、复合体、第一种药物、第二种药物、第三种药物、在表达体系、细胞、组织、器官、体液、组织液的蛋白药物、多肽药物、核酸药物、化学小分子药物、细胞产品、商业化移植组织、注射液、冻干粉、保健品、食品添加剂中的一种或多种。The use according to any one of claims 1 to 32, wherein the therapeutic drug comprises a basic protein, any of the first to seventh proteins, a fusion protein, a polypeptide/protein modification, and an unnatural amino acid. Substitute protein, complex, first drug, second drug, third drug, protein drug, polypeptide drug, nucleic acid drug, chemical small molecule drug, cell in expression system, cell, tissue, organ, body fluid, tissue fluid One or more of products, commercial transplant tissues, injections, lyophilized powder, health care products, food additives.
PCT/CN2018/120172 2018-01-02 2018-12-11 Uses of protein in preparing drug for preventing and treating alzheimer's disease WO2019134481A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810002892.7 2018-01-02
CN201810002892.7A CN109985231A (en) 2018-01-02 2018-01-02 A kind of application of albumen in the drug that preparation prevents and treats dementia

Publications (1)

Publication Number Publication Date
WO2019134481A1 true WO2019134481A1 (en) 2019-07-11

Family

ID=67128294

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/120172 WO2019134481A1 (en) 2018-01-02 2018-12-11 Uses of protein in preparing drug for preventing and treating alzheimer's disease

Country Status (2)

Country Link
CN (1) CN109985231A (en)
WO (1) WO2019134481A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107573412A (en) * 2016-07-04 2018-01-12 上海清流生物医药科技有限公司 A kind of new type natural albumen and its application

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AT506819B1 (en) * 2008-06-12 2011-06-15 Affiris Forschungs Und Entwicklungs Gmbh VACCINE FOR THE TREATMENT OF ALZHEIMER DISEASE
CN102895659B (en) * 2011-07-29 2014-10-29 复旦大学 Composite vaccine for Alzheimer's disease prevention and treatment, and preparation method thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107573412A (en) * 2016-07-04 2018-01-12 上海清流生物医药科技有限公司 A kind of new type natural albumen and its application

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GONG, CHENGXIN: "DSSylation, a Novel Guide for Protein Degradation", PROTEIN & CELL, vol. 5, no. 2, 31 December 2014 (2014-12-31), pages 91 - 93, XP035734440, DOI: doi:10.1007/s13238-014-0020-9 *
ZHANG, YINGHAO ET AL.: "DSSylation, a Novel Protein Modification Targets Proteins Induced by Oxidative Stress, and Facilitates Their Degradation in Cells", PROTEIN & CELL, vol. 5, no. 2, 31 December 2014 (2014-12-31), pages 124 - 140, XP035734439, DOI: doi:10.1007/s13238-013-0018-8 *

Also Published As

Publication number Publication date
CN109985231A (en) 2019-07-09

Similar Documents

Publication Publication Date Title
US20180318383A1 (en) Peptide having neuronal loss prevention and regeneration effects, and composition containing same
AU2016271635B2 (en) Compositions and methods for degradation of misfolded proteins
Schlenzig et al. N‐Terminal pyroglutamate formation of Aβ38 and Aβ40 enforces oligomer formation and potency to disrupt hippocampal long‐term potentiation
JP6751097B2 (en) Peptide for preventing hearing damage and composition containing the same
JP6843625B2 (en) New use of JNK inhibitor molecules for the treatment of various diseases
Wang et al. Neuroinflammation in Alzheimer’s disease: microglia, molecular participants and therapeutic choices
JP2001501972A (en) Amyloid beta protein (globular assembly and its use)
CN113855804A (en) Methods and compositions for treating aging-related disorders
JP7384672B2 (en) C-terminal CDNF fragments and C-terminal MANF fragments, pharmaceutical compositions containing them, and uses thereof
JP2022514088A (en) Hybrid promoters and their use in therapy, especially for treating type II collagen disorders
CN110302362B (en) Application of protein in preparing medicine for preventing and treating diabetes complication
US20230365626A1 (en) Alloferon Peptide and Method Using the Same
WO2019134481A1 (en) Uses of protein in preparing drug for preventing and treating alzheimer&#39;s disease
EP2694977B1 (en) Amyloidosis target useful in methods of screening of compounds
JP6397122B2 (en) Use of peptides to treat angiogenesis-related diseases
WO2015077578A1 (en) Compositions and methods for delivery of bioencapsulated proteins across blood-brain and retinal barriers
JP2011513440A (en) Β-sheet destruction peptide for prevention and / or treatment of Alzheimer&#39;s disease
Lantz et al. The neuroprotective N-terminal amyloid-β core hexapeptide reverses reactive gliosis and gliotoxicity in Alzheimer’s disease pathology models
KR101131512B1 (en) Pharmaceutical Compositions for Preventing or Treating a Neurodegenerative Disorder
US20200230205A1 (en) Compositions and methods for treating myelin disorders
WO2023246847A1 (en) B2m-glun1 blocking peptide, and pharmaceutical composition and use thereof
WO2023143610A2 (en) C-TERMINAL FRAGMENT OF APOLIPOPROTEIN E AND APPLICATION THEREOF IN INHIBITING γ-SECRETASE ACTIVITY
JP2014144933A (en) REMOVAL OF AMYLOID β
CN117229384A (en) Novel somatostatin analogue peptide monomer, dimer or tetramer and preparation and application thereof
Sansa Zaragoza et al. Calpain Inhibition Increases SMN Protein in Spinal Cord Motoneurons and Ameliorates the Spinal Muscular Atrophy Phenotype in Mice

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: 18897999

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18897999

Country of ref document: EP

Kind code of ref document: A1