WO2014092227A1 - Composition for treatment of diseases related to removal of extracellular alpha-synuclein and method for screening therapeutic agent for said diseases - Google Patents

Composition for treatment of diseases related to removal of extracellular alpha-synuclein and method for screening therapeutic agent for said diseases Download PDF

Info

Publication number
WO2014092227A1
WO2014092227A1 PCT/KR2012/011024 KR2012011024W WO2014092227A1 WO 2014092227 A1 WO2014092227 A1 WO 2014092227A1 KR 2012011024 W KR2012011024 W KR 2012011024W WO 2014092227 A1 WO2014092227 A1 WO 2014092227A1
Authority
WO
WIPO (PCT)
Prior art keywords
synuclein
antibodies
antibody
cells
microglia
Prior art date
Application number
PCT/KR2012/011024
Other languages
French (fr)
Korean (ko)
Inventor
이승재
배은진
이혜진
Original Assignee
건국대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 건국대학교 산학협력단 filed Critical 건국대학교 산학협력단
Publication of WO2014092227A1 publication Critical patent/WO2014092227A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to compositions for treating diseases associated with extracellular ⁇ -synuclein removal and methods for screening the same.
  • Parkinson's disease is a chronic progressive motor nervous system disorder. About 50,000 Americans are diagnosed with PD every year. The main symptoms of these neurodegenerative disorders are tremor, stiffness, motor slowness and balance disorder. Many PD patients also suffer from a variety of other symptoms, including emotional changes, memory loss, speech disorders, or difficulty sleeping.
  • PD is caused by the specificity and progressive neuronal loss of midbrain dopamine (DA) neurons.
  • DA midbrain dopamine
  • these neurons produce dopamine, a chemical transporter responsible for signal transduction between melanocytes and striatum, resulting in smooth and intentional muscle activity.
  • dopamine excites the nerve cells of the striatum so that they can be impaired, impairing their ability to direct and control their movements.
  • Korean Patent Publication No. 1020080106928 is administered by administering 7-chloro-4-aminoquinoline compounds such as amodiaquine or glafenine.
  • Methods and kits for treating Parkinson's disease or inhibiting Parkinson's disease are characterized.
  • ⁇ -synuclein is a protein present in the protoplasts located at presynaptic sites (Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, Kittel A, Saitoh T (1995). Neuron 14: 467-475).
  • the present invention has been made in view of the above necessity, and an object of the present invention is to provide a novel Parkinson's disease (PD) and Lewy body dementia (DLB) therapeutic composition.
  • PD Parkinson's disease
  • DLB Lewy body dementia
  • Another object of the present invention is to screen candidate candidates for Parkinson's Disease (PD) and Lewy Body Dementia (DLB).
  • PD Parkinson's Disease
  • DLB Lewy Body Dementia
  • the present invention provides an antibody that specifically binds to the ⁇ -synuclein peptide set forth in SEQ ID NO: 1.
  • the present invention provides a composition for preventing or treating Parkinson's disease, comprising the antibody of the present invention as an active ingredient.
  • the present invention provides a composition for preventing or treating Lewy Body Dementia (DLB) comprising the antibody of the present invention as an active ingredient.
  • DLB Lewy Body Dementia
  • the present invention corresponds to a case where the candidate substance and the extracellular ⁇ -synuclein protein are contacted with the microglial cell under an environment suitable for the two components to interact, and the uptake and degradation rate of the ⁇ -synuclein protein are increased.
  • a method for identifying a candidate for Parkinson's disease or Lewy body dementia (DLB) treatment wherein the candidate is determined to be a candidate for Parkinson's disease or Lewy Body Dementia (DLB).
  • the candidate is preferably an antibody antisense oligonucleotide or a compound, but is not limited thereto.
  • epitope refers to a protein determinant capable of specifically binding to an antibody.
  • Epitopes usually consist of a group of chemically active surface molecules, such as amino acids or sugar side chains, and generally have specific three dimensional structural characteristics as well as specific charge characteristics. Three-dimensional epitopes and non-stereo epitopes are distinguished in that the binding to the former is lost but not to the latter in the presence of a denatured solvent.
  • the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more antibodies of the invention in combination with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and / or adjuvant do.
  • the acceptable formulation material is nontoxic at the dosages and concentrations used for the recipient.
  • a pharmaceutical composition comprising a therapeutically effective amount of an antibody of the invention.
  • preferably acceptable formulation materials are nontoxic to recipients at the dosages and concentrations employed.
  • the pharmaceutical composition modifies, maintains or preserves, for example, the pH, osmolarity, viscosity, purity, color, isotonicity, flavor, sterilization, stability, dissolution or release rate, adsorption or permeation of the composition.
  • suitable formulation materials include, but are not limited to: amino acids (eg, glycine, glutamine, asparagine, arginine or lysine); Antimicrobial agents; Antioxidants (eg, ascorbic acid, sodium sulfate or sodium hydrogen sulfide); Buffer solutions (eg, borate, bicarbonate, tris-HCl, citrate, phosphate or other organic acids); Volume control agents (eg mannitol or glycine); Chelating agents (eg ethylenediamine tetraacetic acid (EDTA); complexing agents (eg caffeine, polyvinylpyrrolidone, ⁇ -cyclodextrin or hydroxypropyl- ⁇ -cyclodextrin); fillers; monosaccharides; Disaccharides; and other carbohydrates (e.g.
  • amino acids eg, glycine, glutamine, asparagine, arginine or lysine
  • Antimicrobial agents eg, ascorbic acid, sodium
  • glucose, mannose, or dextrins proteins
  • proteins e.g. serum albumin, gelatin or immunoglobulins
  • pigments e.g. anti-microbial and diluents
  • emulsifiers hydrophilic polymers (e.g. poly Vinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (e.g. sodium); preservatives (e.g. benzalkonium chloride, benzoic acid, salicylic acid, chimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine Sorbic acid or hydrogen peroxide); solvents (e.g.
  • glycerin propylene glycol or polyethylene glycol
  • sugar alcohols e.g. mannitol or sorbitol
  • suspending agents e.g., surfactants or Wetting agents (e.g., polysorbates such as pluronic, PEG, sorbitan esters, polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol and tiloxapal); stability enhancers ( For example sucrose or sorbitol); enteric enhancers (e.g. alkali metal halide compounds, preferably sodium chloride, potassium chloride, mannitol sorbitol); transport excipients; diluents; prostheses; and / or pharmaceutical adjuvants. See REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, (AR Gennaro, ed.), 1990, obtained from Publishing Company.
  • the optimal pharmaceutical composition will be determined by one of ordinary skill in the art, for example, according to the intended route of administration, mode of transport and dosage required. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES above.
  • the composition affects the rate of in vivo clearance, rate of in vivo release, stability, and physical state of the antibodies of the invention.
  • the primary excipient or carrier in the pharmaceutical composition is aqueous or non-aqueous in nature.
  • a suitable excipient or carrier may be an injectable, physiological saline or artificial cerebrospinal fluid that is generally capable of supplementing other substances in the composition for parenteral administration.
  • Neutral buffered saline or physiological saline mixed with serum albumin is further typical excipients.
  • the pharmaceutical composition of the present invention comprises tris buffer of about pH 7.0-8.5 or acetate buffer of about pH 4.0-5.5, which further comprises sorbitol, sucrose, Tween-20 and / or Suitable substituents thereof.
  • the antibody compositions of the present invention may be formulated with selected compositions having the desired purity and optimal formulation formulations.
  • compositions of the invention can be selected for parenteral administration.
  • the composition may be selected for inhalation or transport through the digestive tract, such as oral. Preparation of such pharmaceutically acceptable compositions is within the skill of the art.
  • the formulation component is preferably present at an acceptable concentration at the site of administration.
  • buffers are used to maintain the composition at physiological or slightly lower pH, generally in the range of about pH 5 to about pH 8.
  • the formulations can also be administered orally.
  • Antibodies of the invention administered in this manner may be formulated with or without a carrier generally used in the preparation of solid pharmaceutical formulations such as tablets and capsules.
  • the capsule may be designed to release the effective portion of the formulation at a gastrointestinal tract location where the bioavailability is maximized and pre-systemic degradation is minimized.
  • Additional agents may be included to facilitate uptake of the antibodies of the invention. Diluents, flavors, low melting waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents and binders may also be used.
  • compositions of the invention are preferably provided comprising an effective amount of one or multiple antibodies of the invention in a mixture with non-toxic excipients suitable for the manufacture of tablets.
  • excipients include, but are not limited to: inert diluents such as calcium carbonate, sodium carbonate or sodium bicarbonate, lactose or calcium phosphate; Or binders such as starch, gelatin or acacia; Or lubricants such as magnesium stearate, stearic acid or talc.
  • compositions used for administration in vivo are generally provided as sterile preparations. It can be sterilized by filtration through sterile filtration membranes. When the composition is lyophilized, it can be sterilized using the method before or after lyophilization and reconstitution.
  • the composition for parenteral administration can be stored in lyophilized form or as a solvent.
  • Parenteral compositions are generally stored in containers with sterile access ports, such as vials or intravenous solution bags with stoppers that can be inserted into subcutaneous needles.
  • the pharmaceutical composition Once the pharmaceutical composition has been formulated, it is stored in sterile vials in the form of solvents, suspensions, gels, emulsions, solids or dehydrogenated or lyophilized powders. Such formulations are stored in an easy-to-use form or in a form that requires reconstitution (eg, lyophilized) prior to administration.
  • an effective amount of a pharmaceutical composition containing an antibody of the invention to be used therapeutically depends, for example, on the therapeutic situation and the subject.
  • dosage levels suitable for treatment may be partially determined by the transport molecule, signs of the use of the antibody of the present invention, route and size of administration (weight, body surface area or organ size) and / or symptoms of the patient (age and overall health). Will vary depending on the condition.
  • the clinician may titrate the dosage and alter the route of administration to obtain the optimal therapeutic effect.
  • Typical dosages range from about 0.1 ⁇ g / kg up to about 30 mg / kg or more, depending on the factors mentioned above.
  • the dosage range is from 0.1 ⁇ g / kg to about 30 mg / kg; More preferably from 1 ⁇ g / kg to about 30 mg / kg; Even more preferably from 5 ⁇ g / kg to about 30 mg / kg.
  • compositions were administered by a single administration or by two or more administrations (with or without the same amount of the desired molecule) or by continuous infusion via an insertion device or catheter over time.
  • refining at a suitable dosage is an area of common work typically made by a person skilled in the art and done by a person skilled in the art. Appropriate doses were identified using appropriate dose-response data.
  • antibodies of the invention can be administered to a patient for an extended period of time. Routine administration of the antibodies of the present invention minimized immune adverse reactions or allergic reactions generally associated with antibodies to human antigens, such as non-complete human antibodies produced in non-human species, in non-human animals.
  • the route of administration of the pharmaceutical composition is according to known methods, including, for example: oral; Injection by intravenous, intraperitoneal, intracranial (brain parenchymal), intraventricular, intramuscular, intraocular, intraarterial, intraportal, intralesional routes; Sustained release system; Or injection using an insertion device.
  • the composition may be administered by injection or infusion or infusion device continuously.
  • composition can be administered topically through the insertion of a membrane, sponge or other suitable material in which the required molecules are absorbed or encapsulated.
  • a membrane, sponge or other suitable material in which the required molecules are absorbed or encapsulated.
  • the device when using an insertion device, the device is inserted into an appropriate tissue or organ and the required molecules are transported by diffusion, sustained-release concentrates or continuous administration.
  • fibrils and oligomers contain both SDS-resistant aggregates (multiple high molecular weights) and SDS soluble aggregates, showing 16 kDa molecular weight as monomers (FIG. 1H, I). ).
  • the inventors cultured rat primary microglia to determine the rate at which these microglia uptake the ⁇ -synuclein protein secreted from neuronal cells.
  • conditioned media obtained from differentiated SH-SY5Y neuroblastoma cells that overexpress human ⁇ -synuclein.
  • the conditioned medium contains both monomeric and SDS stable aggregates (FIG. 1I).
  • uptake of a-synuclein secreted from neuronal cells reached faster and higher levels in 274 antibody treated primary microglia (FIG. 3E, F), which antibody Suggests that it helps microglia in the removal of neuronal secreted extracellular a-synuclein.
  • Antibody-assisted clearance of ⁇ -synuclein aggregates is mediated by Fc ⁇ receptors resulting in rapid delivery to lysosomes
  • Fc ⁇ receptors were present with oligomer ⁇ -synuclein on the surface of BV-2 cells only in the presence of 274 antibodies, not in the presence of control IgG or Fab fragments of 274 antibodies (FIG. 5B), which was responsible for the uptake process of Fc ⁇ receptors. Prove your involvement. Fc ⁇ receptors were markedly expressed in BV-2 microglia and rat primary microglia but were not detected in astrocytic or differentiated neuroblastoma cells (FIG. 5C).
  • Type II and Type III (CD32 / CD16) Fc ⁇ receptors are low affinity receptors for IgG and require a multivalent antigen-antibody complex for high avidity (Ravetch JV, Bolland S (2001) Annu Rev Immunol 19: 275-290). It has a high affinity and cannot bind to these Fc ⁇ receptors.
  • control IgG or monoclonal antibody 274 was precultured with various amounts of ⁇ -synuclein fibrils and added to BV-2 cells. The level of absorbed antibody was directly proportional to the amount of foreign ⁇ -synuclein aggregates (FIG. 5D).
  • LDL low density lipoproteins
  • CTB cholera toxin B subunit
  • microglia To confirm the involvement of microglia in the blockade of cell delivery in the cells, we used the dual immunofluorescence staining to investigate the position of human ⁇ -synuclein and 274 antibodies in microglia.
  • mice injected with 274 antibodies The number of microglia containing human ⁇ -synuclein showed a significant increase ipsilaterally in mice injected with 274 antibodies (FIGS. 8A, B).
  • the injected antibody was also detected in microglia in ipsilaterally in tg mice injected with 274 antibodies (FIG. 8C, D), but not in mice injected with control IgG (FIG. 8D).
  • the co-location of ⁇ -synuclein and injected antibodies was consistently observed in tg mice injected with 274 antibodies (FIG. 8E), but not in mice injected with control IgG.
  • ⁇ -synuclein immunostaining was restricted to neuropil in non-tg mice with no differences observed between the control IgG and 274 antibody groups (FIG. 9E-G). ).
  • IgG-treated ⁇ -synuclein tg mice of astrocytes (FIG. 9F) and neuronal cells (FIG. 9G) in ⁇ -synuclein (FIG. 9E) in neocortex (FIG. 9H) and hippocampus (FIG. 9I). There was extensive accumulation.
  • ⁇ -synuclein tg mice treated with 274 antibodies FIG.
  • FIG. 9E were found to have astrocytes (FIG. 9F) and neurons ⁇ -synuclein (FIG. 9G) in neocortex (FIG. 9H) and hippocampus (FIG. 9I). There was a significant decrease in the accumulation of.
  • mice We also analyzed the striatum of passively immunized mice and found that the levels of ⁇ -synuclein were reduced to a lower extent compared to the cortex and hippocampus.
  • ⁇ -synuclein-containing microglia were observed mainly in neural networks and rarely around the blood vessels.
  • ⁇ -synuclein tg mice treated with IgG alone showed increased levels of pro-immune cytokines, TNF ⁇ (FIG. 10 I, J), and IL-6 (FIG. 10K, L), whereas 274 antibodies Low immunization reduced that level to baseline. This result is consistent with the result that 274 antibodies promote the removal of extracellular ⁇ -synuclein.
  • the antibody against ⁇ -synuclein specifically promotes the removal of extracellular ⁇ -synuclein aggregates by microglia and forms ⁇ -synuclein when complexed with specific antibodies.
  • Synuclein aggregates are absorbed through Fc ⁇ receptors on the surface of microglial cells and after absorption their immune complexes are transported along the transport pathway and cause more efficient delivery to lysosomes.
  • Removal of ⁇ -synuclein mediated by antibodies also inhibited the transfer of ⁇ -synuclein from cell to cell in the tg mouse model.
  • ⁇ -synuclein fibrils and oligomers show the properties of ⁇ -synuclein fibrils and oligomers.
  • AF EM image of fibrils ( A ), sonicated fibrils ( B ), and oligomers ( CF ).
  • Scale bar AC , 0.5 ⁇ m; DF , 30 nm.
  • G size exclusion chromatography of oligomers.
  • Asterisks (7 ml fractions) indicate oligomeric fractions. Monomers are present in 13-15 ml fractions.
  • Silver stained image of H SEC fractions.
  • I Western blot analysis of several forms of ⁇ -synuclein.
  • M monomers
  • F fibrils
  • O oligomers
  • CM conditioned medium of differentiated SH-SY5Y cells expressing ⁇ -synuclein.
  • Figure 2 shows the effect of monoclonal antibodies on the uptake of extracellular ⁇ -synuclein aggregates.
  • A uptake of ⁇ -synuclein fibrils in the presence of described monoclonal antibodies against ⁇ -synuclein or control IgG (-) in BV-2 microglia. 5 ⁇ g / ml ⁇ -synuclein fibrils (0.2 ⁇ M) Antibodies against control IgG or ⁇ -synuclein were preincubated for 5 minutes at room temperature and treated with BV-2 cells at 37 ° C. for 5 minutes. The amount of ⁇ -synuclein absorbed was analyzed by Western blot.
  • B the amount of ⁇ -synuclein absorbed was quantified and normalized to the level of beta-actin.
  • the number of y-axes indicates the amount of ⁇ -synuclein relative to the control.
  • n 4, * p ⁇ 0.05.
  • FIG. 3 shows the effect of 274 antibodies on uptake and degradation of extracellular ⁇ -synuclein in microglia.
  • Adsorbed ⁇ -synuclein aggregates were analyzed by western blotting at the time stated.
  • Graphs in A and C show the amount of ⁇ -synuclein compared to the maximum absorbed level with control IgG.
  • Graphs in B and D show the amount of ⁇ -synuclein remaining compared to that at time O. All values were normalized to beta-actin.
  • E , F show the effect of 274 antibodies on uptake of neuron secreted ⁇ -synuclein in primary microglia.
  • E primary rat microglia were treated with conditioned medium containing ⁇ -synuclein secreted from differentiated SH-SY5Y cells in the presence of control IgG or 274 antibodies.
  • a , C , E uptake rate.
  • the graph shows the amount of ⁇ -synuclein relative to the level of maximum absorption with control IgG. B , D , F , decomposition rate.
  • the graph shows the amount of ⁇ -synuclein remaining compared to that at time O. Pre-incubation time with ⁇ -synuclein fibrils was 12 hours ( B , D , F ). All ⁇ -synuclein data was normalized to beta-actin data. The curve of the graph is not significantly different.
  • FIG. 5 shows the role of Fc ⁇ receptors in the uptake of ⁇ -synuclein-antibody immune complexes.
  • FIG. 1 Shows indicate the co-existence of ⁇ -synuclein and Fc ⁇ receptors on the surface of the cells. Scale bar: 20 ⁇ m.
  • C Expression of Fc ⁇ receptors in several cell types. For BV-2, rat microglia, and rat astrocytes, anti-CD16 / CD32 antibodies specific to rodents were used, whereas for SH-SY5Y, anti-CD32 antibodies specific to human proteins were used. Expression of CD16 has not been reported in brain neurons in previous studies. Red, Fc ⁇ receptors; Blue, nuclear. Scale bar: 20 ⁇ m.
  • D uptake of antibody into BV-2 cells in the presence of several different amounts of ⁇ -synuclein fibrils or ⁇ -synuclein (0 -330 nM). Absorbed antibody was analyzed with anti-mouse IgG antibody. The bottom panel is beta-actin.
  • E Immunotype and microscopic results of ⁇ -synuclein fibrils and antibodies taken up in BV-2 cells. Scale bar: 20 ⁇ m. Quantification of fluorescence from antibodies absorbed at F , E. 150 cells were analyzed in three independent experiments (50 cells in each experiment). * p ⁇ 0.0001.
  • FIG. 6 shows altered intracellular transport and increased transport of lysosomes of uptake of ⁇ -synuclein aggregates.
  • A Altered intracellular transport of ⁇ -synuclein fibrils uptake in BV-2 cells. Fluorescently labeled tracers were treated with BV-2 microglia cells for 5 min at 37 ° C. with control IgG- ⁇ -synuclein mixture or ⁇ -synuclein-274 immune complex. Arrows indicate coexistence between ⁇ -synuclein and CTB in the presence of 274 antibody. Scale bar: 5 ⁇ m.
  • BE localization of ⁇ -synuclein aggregates absorbed in the endocytic compartment.
  • FIG. 7 shows astrocytic delivery in reduced neurons of ⁇ -synuclein in vivo following alone injection of 274 antibody.
  • a , B low-pan (20X) view of the cortex and hippocampus of PDGF- ⁇ -synuclein tg mice (line M) comparing the patterns of ⁇ -synuclein immunoreactivity. Scale bar: 200 ⁇ m.
  • FIG. 8 shows increased localization of ⁇ -synuclein and IgG in microglia cells in vivo after one injection of antibody.
  • PDGF- ⁇ -synuclein tg mice line M
  • images in the AE are obtained from the injection, ipsilateral region and dentate gyrus. Scale bar, 10 ⁇ m.
  • A double labeling with antibodies to ⁇ -synuclein (red) and microglia marker (Iba1, green).
  • B Image analysis for double label for measurement of proportion of Iba-1-positive cells co-located with ⁇ -synuclein.
  • N 8 per group, 9 months of age.
  • G percent of Iba-1-positive cells with ⁇ -synuclein after 1, 7, 14, and 28 days after injection.
  • I Image analysis for double labeling for proportional measurement of positive cells co-existing with ⁇ -synuclein.
  • N 8 per group, 9 months of age.
  • J double label with antibody against ⁇ -synuclein (red) and GFAP (green). Arrowheads indicate ⁇ -synuclein-positive neurons that do not coexist with GFAP; arrows indicate ⁇ -synuclein-positive cells coexist with GFAP.
  • FIG. 9 shows improved behavior and reduced ⁇ -synuclein accumulation in ⁇ -synuclein tg mice after passive immunization with 274 antibody.
  • A the total time traveled down the pole test.
  • B open field total spontaneous activity (number of crosses of photocell).
  • C Open field Total distance traveled 5 minutes after voluntary active testing.
  • D Open field Total number of roundings after 5 minutes of spontaneous active testing.
  • P ⁇ 0.05 by unilateral ANOVA with post hoc Dunnet's compared to AD , * non-tg and ⁇ -synuclein tg; # p ⁇ 0.05 by unilateral ANOVA with post hoc compared to IgG and 274 antibodies in ⁇ -synuclein tg.
  • E low magnification view (x20) of ⁇ -synuclein immunoreactivity in cortex and hippocampus. Squares show magnified sites in F and G. Scale bar, 250 ⁇ m.
  • F , G high magnification (x630) views ( F , astrocytes; G , neurons). Scale bar, 20 ⁇ m.
  • H- sh respectively neocortex, hippocampus, image analysis of glial and neuronal cells can be estimated (dissection method) representing the ⁇ - synuclein in the striatum. * Student's t by test p ⁇ 0.001.
  • K double labeled with antibody against ⁇ -synuclein (red) and neuronal marker (NeuN, green) in hippocampus. Arrow heads indicate coexistence.
  • L Image analysis for double labeling to assess the proportion of NeuN-positive cells co-existing with ⁇ -synuclein.
  • M double labeled with antibodies against ⁇ -synuclein (red) and GFAP (green) in the hippocampus. Arrowheads indicate ⁇ -synuclein-positive cells coexisting with astrocytes.
  • N Image analysis for double labeling for evaluation of the proportion of GFAP positive cells co-existing with antibodies to ⁇ -synuclein.
  • O double labeled with antibodies against ⁇ -synuclein (red) and Iba-1 (green) in hippocampus. Arrowheads indicate ⁇ -synuclein-positive cells coexist with microglia.
  • P Image analysis for double labeling for evaluation of the proportion of Iba-1-positive cells co-existing with antibodies to ⁇ -synuclein.
  • Q double labeled with antibodies against ⁇ -synuclein (red) and cathepsin D (green) in the hippocampus. Arrow heads indicate coexistence.
  • FIG. 10 shows reduced neurodegenerative and pre-inflammatory cytokines after passive immunization with 274 antibodies.
  • ⁇ -synuclein tg mice treated with IgG show neuronal loss in the hippocampus (arrow). Scale bar, 250 ⁇ m.
  • B Stereoscopic Analysis of NeuN in Hippocampus (Anatomy Method).
  • C high magnification views (x630), scale bars, 20 ⁇ m of sections immunostained with antibodies to synaptophysin in ⁇ -synuclein tg and non-tg mice treated with IgG or 274 antibodies, respectively.
  • D stereoscopic analysis of synaptophysin in the hippocampus (anatomist method).
  • ⁇ -synuclein tg mice treated with IgG show increased astrocytes in the neocortex and hippocampus. Scale bar, 250 ⁇ m.
  • F Image analysis of GFAP immune response expressed at absorbance. The measurement is performed on the entire hippocampus.
  • G high magnification view (x630), scale bar, 20 ⁇ m of hippocampal sections immunostained with antibodies against Iba-1 in ⁇ -synuclein tg and non-tg mice treated with IgG or 274 antibodies, respectively.
  • H Image analysis of the estimated number of microglia in the hippocampus. * p ⁇ 0.05 by unilateral ANOVA with post hoc Dunnet s compared to non-tg and ⁇ -synuclein tg; p ⁇ 0.05 by unilateral ANOVA with post hoc Fisher compared to IgG and 274 antibodies at # ⁇ -synuclein tg.
  • Antibodies used in the present invention include: ⁇ -synuclein monoclonal antibody (BD Biosciences, # 610787), ⁇ -synuclein polyclonal antibody (Cell Signaling Technology, # 2642), myc polyclonal antibody (Abcam, # ab9106), CD32 Polyclonal antibodies (United States Biological, # c2384-0B), and CD16 / CD32 monoclonal antibodies (Abcam, # ab25235), GM130 monoclonal antibodies (BD Biosciences, # G65120), cathepsin D monoclonal antibodies (Abcam, # ab6313), and caveolin-1 monoclonal antibody (BD Biosciences, # C13620).
  • ⁇ -synuclein monoclonal antibody BD Biosciences, # 610787
  • ⁇ -synuclein polyclonal antibody Cell Signaling Technology, # 2642
  • myc polyclonal antibody Abcam, # ab9106
  • CD32 Polyclonal antibodies
  • FITC-labeled cholera toxin B subunit was purchased from Sigma.
  • Bodipy-labeled GM1, Bodipy-FL LDL, and Alexa Fluor 568-attached Dextran were purchased from Invitrogen.
  • the antibodies used in the present invention do not distinguish between different forms.
  • 274 antibodies show immunoreactivity to both the extracellular and intraplasma forms of ⁇ -synuclein and to the monomer and aggregate forms.
  • Mixtures of control IgG, mouse IgG isotypes were prepared from normal mouse serum collected using protein A / G columns.
  • Example 2 Purification of ⁇ -synuclein and production of fibrils and oligomers
  • Wild-type human ⁇ -synuclein was purified as described by Lee HJ, Bae EJ, Jang A, Ho DH, Cho ED, Suk JE, Yun YM, Lee SJ (2011b) J Neurosci Methods 199: 249-257.
  • ⁇ -synuclein 3 mg / ml in PBS
  • ⁇ -synuclein was incubated at 37 ° C. for 2 weeks with continued stirring at 250 rpm.
  • ⁇ -synuclein was incubated for another week. After incubation, the protein was centrifuged at 100,000 ⁇ g for 1 hour and the pellet was resuspended in PBS.
  • ⁇ -synuclein oligomers were prepared by the methods described in Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, Kretzschmar H, Hengerer B, Kostka M (2007) J Neurosci 27: 9220-9232. Lyophilized ⁇ -synuclein was dissolved in 50 mM sodium phosphate, pH 7.0 with 20% ethanol to a final concentration of 0.1 mg / ml and stirred at 250 rpm for 4 hours at room temperature.
  • ⁇ -synuclein was lyophilized again and resuspended in 50 mM sodium phosphate, pH 7.0 with 10% ethanol of 1/2 starting volume. The protein was incubated at room temperature for 24 hours with the lid open for evaporation of residual ethanol.
  • SH-SY5Y cells were maintained and differentiated as described in Lee HJ, Khoshaghideh F, Patel S, Lee SJ (2004) J Neurosci 24: 1888-1896.
  • BV-2 microglia cell lines were maintained in DMEM with 5% fetal bovine serum (FBS) and penicillin and streptomycin.
  • Hybridoma cells were cultured in DMEM with 10% FBS and 100 ⁇ M hypoxanthine supplement (Invitrogen). Rat primary glial cultures were prepared as described in Lee HJ, Suk JE, Bae EJ, Lee SJ (2008) Biochem Biophys Res Commun 372: 423-428.
  • Differentiated SH-SY5Y cells were injected with adeno / ⁇ -syn. Two days after injection, cells were washed three times with DMEM and incubated in serum-free DMEM. After 18 hours of incubation at 37 ° C, the conditioned medium was collected and 4 ° C, 250x g in After centrifugation for 10 minutes, the supernatant was removed at 10,000 x 4 ° C to remove cellular debris. g in Centrifuge for 10 minutes. The supernatant was concentrated using an Amicon 10K MWCO filter (Millipore).
  • BV-2 microglia and astrocytes were divided into petri dishes the day before the experiment.
  • 0.2 ⁇ M ⁇ -synuclein fibrils or oligomers previously incubated with 5 ⁇ g / ml normal mouse IgG or ⁇ -synuclein antibodies at room temperature were added to the culture medium and cells. Monomer was added at 1 ⁇ M.
  • Conditioned medium (5 ⁇ concentrated) was preincubated with antibody for 5 minutes. Cells were then incubated at 37 ° C. and collected at the time indicated. To determine the rate of degradation, aggregates were taken up as above and cells were washed twice with cold PBS. Fresh culture medium was added and culture of cells was performed at 37 ° C. for the time stated.
  • BV-2 cells treated with 5 ⁇ g / ml of 274 antibody or control IgG were preincubated with various amounts of ⁇ -synuclein fibrils (0, 2.66, 13.3, 66.5, 133, and 330 nM).
  • ⁇ -synuclein fibrils (0, 2.66, 13.3, 66.5, 133, and 330 nM).
  • 5 ⁇ g / ml of 274 antibody preincubated with various amounts of ⁇ -synuclein monomer (0, 66.5, and 330 nM) was added to the cells.
  • Levels of absorbed antibodies were detected with anti-mouse IgG antibodies.
  • cells cultured on poly-L-lysine-coated coverslips were immobilized in 4% paraformaldehyde in PBS and taken up in 0.1% Triton X-100.
  • Cells were incubated in blocking solution (5% bovine serum albumin / 3% goat serum in PBS) prior to addition of the primary antibody diluted in blocking solution. After washing, the cells were incubated with fluorescent dye attached secondary antibody.
  • Nuclei were stained with TOPRO-3 iodide (Invitrogen) and coverslips mounted on slide glass using antifade reagent (Invitrogen). Olympus FV1000 confocal laser scanning microscope was used for the observation of the cells.
  • mice (line M) overexpressing ⁇ -synuclein derived from the platelet-derived growth factor-beta (PDGF-beta) promoter was used (Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M Takeda A, Sagara Y, Sisk A, Mucke L (2000) Science 287: 1265-1269).
  • This model was chosen because mice derived from these lines generate ⁇ -synuclein aggregates in both neuronal and glial cells distributed throughout the renal cortex, hippocampus, and striatum, similar to those described in Lewy body disease.
  • mice In these mice, ⁇ -synuclein accumulates in neural networks and neuronal cell bodies in the deep layers of the neocortex and hippocampus. ⁇ -synuclein begins to accumulate in neurons at 3 months of age and has maximum accumulation at 12 months of age. Thus, in this tg mouse model, ⁇ -synuclein pathology proceeds in a very constant and spatiotemporal order. All animal testing procedures were approved by the Experimental Animal Ethics Committee. All experiments were performed in female mice. Eight mice per group were used for each experiment. A total of 32 mice were used for this experiment; 16 mice were ⁇ -synuclein tg mice and the other 16 non-tg animal controls (9 months of age).
  • mice placed on Koft stereotactic devices and coordinates hippocampus: AP: 2.0 mm, outer 1.5 mm, depth 1.3 mm
  • mice placed in Franklin and Paxinos (Franklin KBJ, Paxinos G, (2008) The mouse brain in stereotaxic coordinates , Ed 3. Burlington, MA: Elsevier).
  • Hamilton syringes connected to a hydraulic system were used for antibody delivery for injection of the solution.
  • the needles were held for 5 minutes after completion of injection.
  • mice were unilaterally injected (right) for comparison with the contralateral side. Mice were survived 4 weeks after antibody injection. An additional group of twelve ⁇ -synuclein tg mice (3 months old) were unilaterally injected into the hippocampus with antibodies against ⁇ -synuclein (clone 274; 1 mg / ml) and at 1, 7, 14, and 28 days after injection Sacrificed for analysis. Mice were anesthetized with chloral hydrate and perfused to the heart with 0.9% saline according to the National Institutes of Health guidelines for human treatment of animals. The brain was removed and fixed for 48 hours at 4 °C in phosphate-buffer 4% paraformaldehyde, pH 7.4 for neuropathology analysis.
  • mice A total of 32 mice were used in this experiment, of which 16 were ⁇ -synuclein tg mice (line M, 10 months old) and the remaining 16 were non-tg animal controls (10 months old).
  • mice were survived for 1 month after the first injection and as described in Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L (2000) Science 287: 1265-1269.
  • pole test a test for motor function and a spontaneous activity in the open field were performed. Behavioral tests were performed to perform mouse neuropathology analysis.
  • ⁇ -synuclein accumulation was performed on a series of sections, pre-floating, derived from non-tg and ⁇ -synuclein tg mice treated with antibodies to IgG or ⁇ -synuclein (clone 274). Sections were overnight incubated with anti- ⁇ -synuclein antibody (1: 500, affinity purified rabbit polyclonal; Millipore Bioscience Research Reagents) at 4 ° C., followed by biotinylated goat anti-earth IgG (1: 100; Vector Laboratories) After treatment with Avidin D-horseradish peroxidase (1: 200, ABC Elite; Vector Laboratories), it was detected with diaminobenzidine (DAB).
  • anti- ⁇ -synuclein antibody 1: 500, affinity purified rabbit polyclonal; Millipore Bioscience Research Reagents
  • biotinylated goat anti-earth IgG 1: 100; Vector Laboratories
  • Sections were imaged with an Olympus brightfield digital microscope. For each case, three sections were analyzed by dissection analysis using the Stereo-Investigator System (MBF Bioscience) and the results were averaged and expressed in number / mm 3 . To analyze the effect of antibodies on neurodegenerative and neuroinflammatory responses, sections were analyzed for NeuN (Millipore), glial fibrillary acidic protein (GFAP) (Millipore), Iba1 (Wako), IL6 (Novus Biologicals), and tumor necrosis factor Mouse monoclonal antibody against Abcam) and detected with DAB.
  • NeuN Millipore
  • GFAP glial fibrillary acidic protein
  • Iba1 Wako
  • IL6 Novus Biologicals
  • tumor necrosis factor Mouse monoclonal antibody against Abcam tumor necrosis factor Mouse monoclonal antibody against Abcam
  • Sections immunostained with antibody GFAP and Iba-1 were imaged with an Olympus brightfield digital microscope and absorbance and cell counts were analyzed with an Image Pro-Plus system. Sections immunostained with antibodies to NeuN were analyzed by dissection analysis using Stereo-Investigator System (MBF Bioscience) and the results were averaged and expressed in number / mm 3 .
  • ⁇ -synuclein (Millipore Bioscience Research Reagents, affinity purified polyclonal, 1: 500), mouse IgG (to detect 274 antibody distribution), and Double labeling with a combination of antibodies to Iba-1 (microglia marker).
  • ⁇ -synuclein immunoreactive structures were detected by the Tyramide Signal Amplification-Direct (Red) system (1: 100; NEN Life Sciences) while mouse IgG or Iba-1 were fluorescein isothiocyanate (FITC) -attached horse anti-mouse antibodies (1:75; Vector Laboratories).
  • FITC fluorescein isothiocyanate
  • ⁇ -synuclein was detected by the Tyramide Signal Amplification-Direct (Red) system (1: 100; NEN Life Sciences), while NeuN or GFAP was detected by FITC.

Abstract

The present invention relates to a composition for treatment of diseases related to the removal of extracellular alpha-synuclein and to a method for screening a therapeutic agent for said diseases.

Description

세포외 α-시누클레인 제거와 관련된 질환 치료용 조성물 및 그 치료제 스크리닝 방법Compositions for the treatment of diseases associated with extracellular α-synuclein removal and methods for screening the same
본 발명은 세포외 α-시누클레인 제거와 관련된 질환 치료용 조성물 및 그 치료제 스크리닝 방법에 관한 것이다.The present invention relates to compositions for treating diseases associated with extracellular α-synuclein removal and methods for screening the same.
파킨슨병(PD)은 만성 진행성 운동 신경계 이상이다. 약 5만 정도의 미국인들이 매년 PD로 진단된다. 이러한 퇴행성신경 질환의 주증세들은 떨림, 경직, 운동 완서 및 균형 장애이다. 또한, 많은 PD 환자들은 감정변화, 기억상실, 언어 장애 또는 수면 곤란을 포함하는 다양한 다른 증세들을 겪는다.Parkinson's disease (PD) is a chronic progressive motor nervous system disorder. About 50,000 Americans are diagnosed with PD every year. The main symptoms of these neurodegenerative disorders are tremor, stiffness, motor slowness and balance disorder. Many PD patients also suffer from a variety of other symptoms, including emotional changes, memory loss, speech disorders, or difficulty sleeping.
PD는 중뇌 도파민(DA) 신경세포들의 특이성 및 진행성 신경세포 손실에 의하여 유발된다. 주로, 이러한 신경세포들은 흑색질 및 선조체 사이의 신호 전달을 담당하는 화학 전달물질인 도파민을 생성하여 매끄럽고 의도적인 근육 활성을 발생시킨다. 그러나, 도파민의 손실은 선조체의 신경세포들이 제어할 수 없을 만큼 흥분하게 하여 환자들이 자신의 운동을 지시하고 제어하는 능력에 손상을 입게 한다.PD is caused by the specificity and progressive neuronal loss of midbrain dopamine (DA) neurons. Primarily, these neurons produce dopamine, a chemical transporter responsible for signal transduction between melanocytes and striatum, resulting in smooth and intentional muscle activity. However, the loss of dopamine excites the nerve cells of the striatum so that they can be impaired, impairing their ability to direct and control their movements.
PD에 대하여 현재 이용되는 요법은 도파민 전구체인 L-DOPA(L-디히드록시페닐-알라닌(L-dihydroxyphenylalanine))을 환자에게 경구 투여하여 도파민을 보충하는데 크게 의존한다. 이러한 요법은 치료가 계속됨에 따라 투여량의 증가를 필요로 하고, 결국 심각한 부작용들을 도출하게 된다. PD에 대한 다른 요법들에 대한 요구가 있다.Currently used therapies for PD rely heavily on supplementing dopamine by oral administration of L-DOPA (L-dihydroxyphenylalanine), a dopamine precursor, to a patient. Such therapy requires an increase in dosage as treatment continues, resulting in serious side effects. There is a need for other therapies for PD.
관련 선행특허로 대한민국 특허공개번호 제1020080106928호는 아모다이아퀸(amodiaquine) 또는 글라페닌(glafenine)과 같은 7-클로로-4-아미노퀴놀린 화합물들(7-chloro-4-aminoquinoline compounds)을 투여하여 파킨슨병을 치료하거나 파킨슨병 발달을 억제시키는 방법들 및 키트들을 특징으로 한다.As a related prior patent, Korean Patent Publication No. 1020080106928 is administered by administering 7-chloro-4-aminoquinoline compounds such as amodiaquine or glafenine. Methods and kits for treating Parkinson's disease or inhibiting Parkinson's disease are characterized.
한편 파킨슨병(PD) 및 루이소체 치매(DLB)와 같은 α-시누클레인(synuclein)의 축적을 가지는 질환은 운동 장애 및 노인 인구에서 치매의 일반적인 원인이다. 생리적인조건에서 α-시누클레인은 시냅스전(presynaptic) 부위에 위치한 원형질에 존재하는 단백질이다(Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, Kittel A,Saitoh T (1995) Neuron 14:467-475).Meanwhile, diseases with accumulation of α-synuclein, such as Parkinson's disease (PD) and Lewy body dementia (DLB), are a common cause of dementia in motor disorders and the elderly population. Under physiological conditions, α-synuclein is a protein present in the protoplasts located at presynaptic sites (Iwai A, Masliah E, Yoshimoto M, Ge N, Flanagan L, de Silva HA, Kittel A, Saitoh T (1995). Neuron 14: 467-475).
본 발명은 상기의 필요성에 의하여 안출된 것으로서 본 발명의 목적은 새로운 파킨슨병(PD) 및 루이소체 치매(DLB) 치료제 조성물을 제공하는 것이다.The present invention has been made in view of the above necessity, and an object of the present invention is to provide a novel Parkinson's disease (PD) and Lewy body dementia (DLB) therapeutic composition.
본 발명의 다른 목적은 파킨슨병(PD) 및 루이소체 치매(DLB) 치료제 후보물질을 스크리닝하는 것이다.Another object of the present invention is to screen candidate candidates for Parkinson's Disease (PD) and Lewy Body Dementia (DLB).
상기의 목적을 달성하기 위하여 본 발명은 서열번호 1에 기재된 α-시누클레인 펩타이드에 특이적으로 결합하는 항체를 제공한다.In order to achieve the above object, the present invention provides an antibody that specifically binds to the α-synuclein peptide set forth in SEQ ID NO: 1.
또 본 발명은 상기 본 발명의 항체를 유효성분으로 포함하는 파킨슨 질환 예방 또는 치료용 조성물을 제공한다.In another aspect, the present invention provides a composition for preventing or treating Parkinson's disease, comprising the antibody of the present invention as an active ingredient.
또 본 발명은 상기 본 발명의 항체를 유효성분으로 포함하는 루이소체 치매(DLB) 예방 또는 치료용 조성물을 제공한다.In another aspect, the present invention provides a composition for preventing or treating Lewy Body Dementia (DLB) comprising the antibody of the present invention as an active ingredient.
또 본 발명은 후보 물질과 세포외 α-시누클레인 단백질을 이 두 성분이 상호작용하기에 적합한 환경 하에서 미세아교세포주에 접촉시키고, 상기 α-시누클레인 단백질의 업테이크 및 분해 속도가 증가한 경우에 해당 후보물질을 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질로 판정하는 것을 특징으로 하는 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질 동정 방법을 제공한다.In addition, the present invention corresponds to a case where the candidate substance and the extracellular α-synuclein protein are contacted with the microglial cell under an environment suitable for the two components to interact, and the uptake and degradation rate of the α-synuclein protein are increased. Provided is a method for identifying a candidate for Parkinson's disease or Lewy body dementia (DLB) treatment, wherein the candidate is determined to be a candidate for Parkinson's disease or Lewy Body Dementia (DLB).
본 발명의 일 구현예에 있어서, 상기 후보물질은 항체 안티센스 올리고뉴클레오티드인 또는 화합물인 것이 바람직하나 이에 한정되지 아니한다.In one embodiment of the present invention, the candidate is preferably an antibody antisense oligonucleotide or a compound, but is not limited thereto.
본 발명에서 "에피토프"라는 용어는 항체에 특이적으로 결합할 수 있는 단백질 결정부위 (determinant)를 의미한다. 에피토프는 통상화학적으로 활성인 표면 분자군, 예를 들어 아미노산 또는 당 측쇄로 구성되며, 일반적으로 특정한 3차원의 구조적 특징뿐만 아니라 특정한 전하 특성을 갖는다. 입체적 에피토프 및 비입체적 에피토프는 변성 용매의 존재하에서 전자에 대한 결합은 소실되지만 후자에 대해서는 소실되지 않는다는 점에서 구별된다.As used herein, the term "epitope" refers to a protein determinant capable of specifically binding to an antibody. Epitopes usually consist of a group of chemically active surface molecules, such as amino acids or sugar side chains, and generally have specific three dimensional structural characteristics as well as specific charge characteristics. Three-dimensional epitopes and non-stereo epitopes are distinguished in that the binding to the former is lost but not to the latter in the presence of a denatured solvent.
바람직한 실시형태에서, 본 발명은 약학적으로 용인가능한 희석제, 담체, 가용화제, 유화제, 방부제 및/또는 아쥬반트와 함께 본 발명의 하나 또는 그 이상의 항체의 치료학적 유효량을 포함하는 약학적 조성물을 제공한다. 바람직하게, 용인가능한 제형화 물질은 수용자에게 사용된 투여량 및 농도에서 비독성이다. 바람직한 실시형태에서, 본 발명의 항체의 치료학적 유효량을 포함하는 약학적 조성물을 제공한다.In a preferred embodiment, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more antibodies of the invention in combination with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and / or adjuvant do. Preferably, the acceptable formulation material is nontoxic at the dosages and concentrations used for the recipient. In a preferred embodiment, there is provided a pharmaceutical composition comprising a therapeutically effective amount of an antibody of the invention.
특정 실시형태에서, 바람직하게 용인가능한 제형 물질은 사용된 투여량 및 농도에서 수용자에게 비독성이다.In certain embodiments, preferably acceptable formulation materials are nontoxic to recipients at the dosages and concentrations employed.
특정 실시형태에서, 약학적 조성물은 예를 들어, 조성물의 pH, 삼투 몰농도, 점도, 정화도, 색, 등장, 향, 살균, 안정성, 용해나 방출 속도, 흡착 또는 투과를 변형, 유지 또는 보존하기 위한 제형화 물질을 포함한다. 이러한 실시형태에서, 적합한 제형화 물질로는 다음을 포함하나 이에 한정되지는 않는다 : 아미노산(예를 들어, 글리신, 글루타민, 아스파라긴, 아르기닌또는 리신) ; 항미생물제 ; 항산화제(예를 들어, 아스코르브산, 황산나트륨 또는 황화수소나트륨) ; 완충용액(예를 들어, 붕산염, 중탄산염, 트리스-HCl, 시트르산염, 인산염 또는 다른 유기산) ; 부피 조절제(예를 들어, 만니톨 또는 글리신) ; 킬레이트제(예를 들어, 에틸렌디아민 테트라아세트산(EDTA) ; 착화제(예를 들어, 카페인, 폴리비닐피롤리돈, β-시클로덱스틴 또는 히드록시프로필-β-시클로덱스트린) ; 충전제 ; 단당류 ; 이당류 ; 및 그외 탄수화물(예를 들어, 글루코스, 만노스 또는 덱스트린) ; 단백질(예를 들어, 혈청 알부민, 겔라틴 또는 면역글로불린) ; 색소, 항미제 및 희석제 ; 유화제 ; 친수성 중합체(예를 들어, 폴리비닐피롤리돈) ; 저 분자량 폴리펩티드 ; 염-형성 대이온(예를 들어, 나트륨) ; 방부제(예를 들어, 염화벤잘코늄, 벤조산, 살리실산, 치메로살, 페네틸알콜, 메틸파라벤, 프로필파라벤, 클로르헥시딘, 소르브산 또는 과산화수소) ; 용매(예를 들어, 글리세린, 프로필렌글리콜 또는 폴리에틸렌글리콜) ; 당알콜(예를 들어, 만니톨 또는 소르비톨) ; 현탁화제 ;계면활성제 또는 습윤제(예를 들어, 플루로닉, PEG, 소르비탄에스텔류, 폴리소르베이트 20, 폴리소르베이트 80 과 같은 폴리소르베이트, 트리톤, 트로메타민, 레시틴, 콜레스테롤 및 틸옥사팔) ; 안정성 증강제(예를 들어, 수크로스 또는 소르비톨) ; 장도 증강제(예를 들어, 알칼리 금속 할로겐 화합물, 바람직하게, 염화나트륨, 염화칼륨, 만니톨 소르비톨) ; 수송 부형제 ; 희석제 ; 보형약 ; 및/또는 약학적 아쥬반트. Mack Publishing Company 로부터 입수한 REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, (A.R. Gennaro, ed.), 1990 을 참조.In certain embodiments, the pharmaceutical composition modifies, maintains or preserves, for example, the pH, osmolarity, viscosity, purity, color, isotonicity, flavor, sterilization, stability, dissolution or release rate, adsorption or permeation of the composition. Formulation materials for In such embodiments, suitable formulation materials include, but are not limited to: amino acids (eg, glycine, glutamine, asparagine, arginine or lysine); Antimicrobial agents; Antioxidants (eg, ascorbic acid, sodium sulfate or sodium hydrogen sulfide); Buffer solutions (eg, borate, bicarbonate, tris-HCl, citrate, phosphate or other organic acids); Volume control agents (eg mannitol or glycine); Chelating agents (eg ethylenediamine tetraacetic acid (EDTA); complexing agents (eg caffeine, polyvinylpyrrolidone, β-cyclodextrin or hydroxypropyl-β-cyclodextrin); fillers; monosaccharides; Disaccharides; and other carbohydrates (e.g. glucose, mannose, or dextrins); proteins (e.g. serum albumin, gelatin or immunoglobulins); pigments, anti-microbial and diluents; emulsifiers; hydrophilic polymers (e.g. poly Vinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (e.g. sodium); preservatives (e.g. benzalkonium chloride, benzoic acid, salicylic acid, chimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine Sorbic acid or hydrogen peroxide); solvents (e.g. glycerin, propylene glycol or polyethylene glycol); sugar alcohols (e.g. mannitol or sorbitol); suspending agents; surfactants or Wetting agents (e.g., polysorbates such as pluronic, PEG, sorbitan esters, polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol and tiloxapal); stability enhancers ( For example sucrose or sorbitol); enteric enhancers (e.g. alkali metal halide compounds, preferably sodium chloride, potassium chloride, mannitol sorbitol); transport excipients; diluents; prostheses; and / or pharmaceutical adjuvants. See REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, (AR Gennaro, ed.), 1990, obtained from Publishing Company.
특정 실시형태에서, 최적의 약학적 조성물은 예를 들어, 의도된 투여 경로, 수송 형식 및 요구되는 투여량에 따라 본 분야의 숙련자들에 의해 결정될 것이다. 예를 들어, 상기의 REMINGTON'S PHARMACEUTICAL SCIENCES 을 참조하라. 특정 실시형태에서, 상기 조성물은 본 발명의 항체의 생체내 제거 속도, 생체내 방출 속도, 안정성 및 물리적 상태에 영향을 미친다.In certain embodiments, the optimal pharmaceutical composition will be determined by one of ordinary skill in the art, for example, according to the intended route of administration, mode of transport and dosage required. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES above. In certain embodiments, the composition affects the rate of in vivo clearance, rate of in vivo release, stability, and physical state of the antibodies of the invention.
특정 실시형태에서, 약학적 조성물 내의 원발성 부형제 또는 담체는 자연 상태에서 수성이거나 비-수성이다. 예를 들어,적합한 부형제 또는 담체는 비경구적으로 투여하기 위한 조성물에 일반적으로 다른 물질을 보충하는 것이 가능한 주사용물, 생리식염수 또는 인공 뇌척수액일 수 있다. 중성인 완충생리식염수 또는 혈청 알부민과 혼합된 생리식염수는 추가로 전형적인 부형제이다. 바람직한 실시형태에서, 본 발명의 약학적 조성물은 약 pH 7.0-8.5 의 트리스 완충용액 또는 약 pH 4.0-5.5 의 아세트산염 완충용액을 포함하며, 이는 추가로 소르비톨, 수크로스, Tween-20 및/또는 적합한 이의 치환체를 포함한다. 본 발명의 특정 실시형태에서, 본 발명의 항체 조성물을 바람직한 순도를 갖는 선택 조성물과 최적의 제형화 제제In certain embodiments, the primary excipient or carrier in the pharmaceutical composition is aqueous or non-aqueous in nature. For example, a suitable excipient or carrier may be an injectable, physiological saline or artificial cerebrospinal fluid that is generally capable of supplementing other substances in the composition for parenteral administration. Neutral buffered saline or physiological saline mixed with serum albumin is further typical excipients. In a preferred embodiment, the pharmaceutical composition of the present invention comprises tris buffer of about pH 7.0-8.5 or acetate buffer of about pH 4.0-5.5, which further comprises sorbitol, sucrose, Tween-20 and / or Suitable substituents thereof. In certain embodiments of the present invention, the antibody compositions of the present invention may be formulated with selected compositions having the desired purity and optimal formulation formulations.
를 혼합하여(상기 참고문헌으로 인용한 REMINGTON'S PHARMACEUTICAL SCIENCES 을 참조) 동결건조된 케이크 또는 수용액 형태로 저장하기 위해 제조하였다. Were prepared for storage in the form of lyophilized cake or aqueous solution by mixing (see REMINGTON'S PHARMACEUTICAL SCIENCES cited above).
본 발명의 약학적 조성물을 비경구적 투여를 위해 선택할 수 있다. 선택적으로, 조성물을 경구와 같은 소화관을 통한 수송 이나 또는 흡입을 위해 선택할 수 있다. 이러한 약학적으로 용인가능한 조성물의 제조는 본 분야의 기술에 속한다.The pharmaceutical compositions of the invention can be selected for parenteral administration. Optionally, the composition may be selected for inhalation or transport through the digestive tract, such as oral. Preparation of such pharmaceutically acceptable compositions is within the skill of the art.
제형화 성분은 바람직하게 투여 부위에 용인가능한 농도로 존재한다. 특정 실시형태에서, 생리학적 pH 또는 약간 낮은 pH, 일반적으로 약 pH 5 내지 약 pH 8 의 범위로 조성물을 유지하기 위해서 완충용액을 사용하였다.The formulation component is preferably present at an acceptable concentration at the site of administration. In certain embodiments, buffers are used to maintain the composition at physiological or slightly lower pH, generally in the range of about pH 5 to about pH 8.
또한 제형을 또한 경구적으로 투여할 수 있다. 이러한 방식으로 투여되는 본 발명의 항체를 정제 및 캡슐과 같은 고체 약제학적 제형의 조제시에 일반적으로 사용되는 담체와 함께 또는 담체 없이 제형화할 수 있다. 특정 실시형태에서, 캡슐은 생체내이용효율이 최대화되고 예비-전신성의(pre-systemic) 분해가 최소화되는 위장관 위치에서 제형의 유효 부분이 방출되도록 설계될 수 있다. 부가적인 제제를 본 발명의 항체의 흡수를 용이하게 하기 위해 포함할 수 있다. 희석제, 향미료, 저융점 왁스, 식물성 오일, 윤활유, 현탁화제, 정제 붕해제 및 결합제를 또한 사용할 수 있다.The formulations can also be administered orally. Antibodies of the invention administered in this manner may be formulated with or without a carrier generally used in the preparation of solid pharmaceutical formulations such as tablets and capsules. In certain embodiments, the capsule may be designed to release the effective portion of the formulation at a gastrointestinal tract location where the bioavailability is maximized and pre-systemic degradation is minimized. Additional agents may be included to facilitate uptake of the antibodies of the invention. Diluents, flavors, low melting waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents and binders may also be used.
본 발명의 약학적 조성물은 정제의 제조에 적절한 비-독성 부형제와 함께 혼합물에서 하나 또는 다수의 본 발명의 항체의 유효량을 포함하여 제공되는 것이 바람직하다. 정제를 멸균수 또는 다른 적절한 부형제에 용해시킴으로써, 용액을 단위 약제학적 제형으로 제조할 수 있다. 적절한 부형제는 다음을 포함하지만 이로 제한하지 않는다 : 탄산칼슘, 탄산나트륨 또는 중탄산나트륨, 락토스 또는 인산칼슘과 같은 비활성 희석제 ; 또는 녹말, 겔라틴 또는 아카시아와 같은 결합제 ; 또는 스테아르산 마그네슘, 스테아르산 또는 활석과 같은 윤활제.Pharmaceutical compositions of the invention are preferably provided comprising an effective amount of one or multiple antibodies of the invention in a mixture with non-toxic excipients suitable for the manufacture of tablets. By dissolving the tablets in sterile water or other suitable excipients, solutions may be prepared in unit pharmaceutical formulations. Suitable excipients include, but are not limited to: inert diluents such as calcium carbonate, sodium carbonate or sodium bicarbonate, lactose or calcium phosphate; Or binders such as starch, gelatin or acacia; Or lubricants such as magnesium stearate, stearic acid or talc.
일반적으로 생체내로 투여하기 위해 사용되는 약학적 조성물은 일반적으로 멸균 제제로서 제공된다. 멸균 여과막을 통하여 여과시킴으로써 멸균할 수 있다. 조성물을 동결건조시킬 경우, 동결건조 및 재구성 전에 또는 후에 상기 방법을 이용하여 멸균 처리할 수 있다. 비경구 투여용 조성물을 동결건조된 형태나 용제로 저장할 수 있다. 일반적으로 비경구용 조성물을 멸균 접근 구멍(sterile access port)을 구비한 용기, 예를 들어 피하 주사바늘로 찔러넣을 수 있는 스토퍼(stopper)를 구비한 바이알 또는 정맥내 용액백 내에 보관하였다.Pharmaceutical compositions used for administration in vivo are generally provided as sterile preparations. It can be sterilized by filtration through sterile filtration membranes. When the composition is lyophilized, it can be sterilized using the method before or after lyophilization and reconstitution. The composition for parenteral administration can be stored in lyophilized form or as a solvent. Parenteral compositions are generally stored in containers with sterile access ports, such as vials or intravenous solution bags with stoppers that can be inserted into subcutaneous needles.
일단 약학적 조성물이 제형화되면, 용제, 현탁액, 겔, 에멀젼, 고형제 또는 탈수소화되었거나 동결건조된 분말의 형태로 멸균 바이알에 저장하였다. 이러한 제형을 사용하기 쉬운 형태로 저장하거나 또는 투여하기에 전에 재구성이 요구되는 형태(예를 들어, 동결건조된 형태)로 저장하였다.Once the pharmaceutical composition has been formulated, it is stored in sterile vials in the form of solvents, suspensions, gels, emulsions, solids or dehydrogenated or lyophilized powders. Such formulations are stored in an easy-to-use form or in a form that requires reconstitution (eg, lyophilized) prior to administration.
치료학적으로 이용되는 본 발명의 항체를 함유한 약학적 조성물의 유효량은 예를 들어, 치료 상황 및 대상에 좌우된다. 본 분야에 숙련자들은 치료에 적합한 투여량 수준이 부분적으로 수송 분자, 본 발명의 항체가 사용되는 징후, 투여 경로와 크기(체중, 체표면적 또는 기관 크기) 및/또는 환자의 증상(연령 및 총체적인 건강 상태)에 따라 다양하다는 것을 인지할 것이다. 특정 실시형태에서, 임상의는 최적의 치료 효과를 얻기 위해 투여량을 적정하고 투여 경로를 변경할 수 있다. 일반적인 투여량의 범위는 상기 언급한 인자에 따라 약 0.1 ㎍/㎏ 내지 최대 약 30 ㎎/㎏ 또는 그 이상이다. 바람직한 실시형태에 있어서, 투여량 범위는 0.1 ㎍/㎏ 내지 약 30 ㎎/㎏ 까지 ; 보다 바람직하게 1 ㎍/㎏ 내지 약 30 ㎎/㎏ 까지 ; 보다 더 바람직하게 5 ㎍/㎏ 내지 약 30 ㎎/㎏ 까지이다.An effective amount of a pharmaceutical composition containing an antibody of the invention to be used therapeutically depends, for example, on the therapeutic situation and the subject. Those skilled in the art will appreciate that dosage levels suitable for treatment may be partially determined by the transport molecule, signs of the use of the antibody of the present invention, route and size of administration (weight, body surface area or organ size) and / or symptoms of the patient (age and overall health). Will vary depending on the condition. In certain embodiments, the clinician may titrate the dosage and alter the route of administration to obtain the optimal therapeutic effect. Typical dosages range from about 0.1 μg / kg up to about 30 mg / kg or more, depending on the factors mentioned above. In a preferred embodiment, the dosage range is from 0.1 μg / kg to about 30 mg / kg; More preferably from 1 μg / kg to about 30 mg / kg; Even more preferably from 5 μg / kg to about 30 mg / kg.
투여 횟수는 사용된 제형의 특정 본 발명의 항체의 약물동력학 파라미터에 좌우될 것이다. 일반적으로, 임상의는 투여량이 바람직한 효과를 달성할 때까지 조성물을 투여하였다. 그러므로, 조성물을 단일 투여 또는 시간에 따라 두 번 또는 그 이상의 투여(원하는 분자의 동일양을 포함하거나 포함하지 않는) 또는 삽입 장치 또는 카테터를 통해 지속적으로 주입함으로써 투여하였다. 더욱이, 적합한 투여량의 정련은 본 분야의 숙련자에 의해 통상적으로 만들어지며 숙련자에 의해 행해지는 통상적인 일의 영역이다. 적합한 투여량을 적절한 투여-반응 데이터를 이용하여 확인하였다. 특정 실시형태에서, 본 발명의 항체를 연장된 시간 기간 동안 환자에게 투여할 수 있다. 본 발명의 항체의 상습적인 투여는 비-인체 동물에서 인체 항원에 대한 항체, 예를 들어 비-인체 종에서 생산된 비-완전 인간 항체와 일반적으로 관련된 면역 역반응 또는 알레르기성 반응을 최소화하였다.The number of administrations will depend on the pharmacokinetic parameters of the antibodies of the invention of the formulation used. In general, the clinician has administered the composition until the dosage achieves the desired effect. Therefore, the compositions were administered by a single administration or by two or more administrations (with or without the same amount of the desired molecule) or by continuous infusion via an insertion device or catheter over time. Moreover, refining at a suitable dosage is an area of common work typically made by a person skilled in the art and done by a person skilled in the art. Appropriate doses were identified using appropriate dose-response data. In certain embodiments, antibodies of the invention can be administered to a patient for an extended period of time. Routine administration of the antibodies of the present invention minimized immune adverse reactions or allergic reactions generally associated with antibodies to human antigens, such as non-complete human antibodies produced in non-human species, in non-human animals.
약학적 조성물의 투여 경로는 예를 들어 다음을 포함하는 공지된 방법에 따른 것이다 : 경구 ; 정맥내, 복강내, 뇌내(뇌실질), 뇌실내, 근육내, 안내, 동맥내, 문맥내, 병변내 경로으로 주사 ; 서방성 시스템 ; 또는 삽입 장치를 이용한 주사. 특정 실시형태에서, 조성물을 농축괴 주사하거나 연속적으로 주입 또는 삽입 장치로 투여할 수 있다.The route of administration of the pharmaceutical composition is according to known methods, including, for example: oral; Injection by intravenous, intraperitoneal, intracranial (brain parenchymal), intraventricular, intramuscular, intraocular, intraarterial, intraportal, intralesional routes; Sustained release system; Or injection using an insertion device. In certain embodiments, the composition may be administered by injection or infusion or infusion device continuously.
조성물을 요구되는 분자가 흡수되거나 또는 캡슐화된 멤브레인, 스폰지 또는 다른 적절한 물질의 삽입을 통하여 국소적으로 투여할 수 있다. 특정 실시형태에서, 삽입 장치를 사용할 경우, 장치를 적절한 조직 또는 기관 내로 삽입하였으며, 요구되는 분자를 확산, 지효성 농축괴 또는 연속 투여함으로써 수송하였다.The composition can be administered topically through the insertion of a membrane, sponge or other suitable material in which the required molecules are absorbed or encapsulated. In certain embodiments, when using an insertion device, the device is inserted into an appropriate tissue or organ and the required molecules are transported by diffusion, sustained-release concentrates or continuous administration.
이하 본 발명을 설명한다.Hereinafter, the present invention will be described.
본 발명자들은 α-시누클레인에 대한 항체가 세포외 α-시누클레인 단백질을 특이적으로 타겟하고 미세아교세포(microglia)에 의한 제거에 도움을 주어 인접한 세포에 대한 그들의 작용을 억제한다는 것을 보여준다. 항체 지원된 제거는 신경세포나 성상교세포에서가 아니라 Fcγ 수용체를 통하여 미세아교세포에서 주로 일어난다. α-시누클레인 tg 마우스의 뇌에 항체의 정위방법적(Stereotaxic) 투여는 α-시누클레인의 뉴런에서 성상교세포로의 전달을 억제하여 미세아교세포에서 α-시누클레인 및 그 항체의 증가를 야기한다. 또한α-시누클레인 항체를 이용한 수동면역은 α-시누클레인의 신경 및 신경교 축적을 감소시키고 α-시누클레인 과발현과 관련된 행동 장애 및 신경퇴행을 완화시킨다. 이 결과들은 PD/DLB에 대한 면역치료를 위한 기전을 제공하고 치료타겟으로 α-시누클레인의 세포외 형태를 제안한다.We show that antibodies against α-synuclein specifically target extracellular α-synuclein proteins and aid in their removal by microglia, thus inhibiting their action on adjacent cells. Antibody-assisted clearance occurs mainly in microglia via Fcγ receptors, but not in neurons or astrocytes. Stereotaxic administration of antibodies to the brain of α-synuclein tg mice inhibits the transfer of α-synuclein to astrocytes in neurons resulting in an increase of α-synuclein and its antibodies in microglial cells . Passive immunization with α-synuclein antibodies also reduces neuronal and glial accumulation of α-synuclein and mitigates behavioral disorders and neurodegeneration associated with α-synuclein overexpression. These results provide a mechanism for immunotherapy against PD / DLB and suggest an extracellular form of α-synuclein as a therapeutic target.
이하 본 발명을 상세하게 설명한다.Hereinafter, the present invention will be described in detail.
미세아교세포에 의한 세포외 α-시누클레인 응집체의 항체 매개된 제거Antibody-Mediated Removal of Extracellular α-synuclein Aggregates by Microglia
α-시누클레인에 대한 항체의 치료효과를 조사하기 위하여, 본 발명자들은 단클론 인간 α-시누클레인에 대한 항체를 제조하고 미세아교세포에서 세포외 α-시누클레인 응집체의 제거에 대한 그들의 효과를 테스트하였다.To investigate the therapeutic effect of antibodies against α-synuclein, we prepared antibodies against monoclonal human α-synuclein and tested their effects on the removal of extracellular α-synuclein aggregates from microglia. .
본 발명자들은 재조합 인간 α-시누클레인으로부터 올리고머 및 피브릴을 생성하고; 전자현미경(EM), 크기 배제 크로마토그래피(SEC), 및 젤 전기영동으로 그들의 물리적인 특성을 확인하였다.α-시누클레인 올리고머는 EM 상에서 구형, 커비(curvy) 및 짧은 신장형(elongated) 구조의 이종 혼합체로 나타난다(도 1C-F). SEC 데이터에 기반하여, 올리고머에서 약 절반의 단백질은 높은 분자량 올리고머이고 다른 절반은 모노머이었다(도1G,H). 실버 염색 및 웨스턴 블럿팅에 의하여 본 발명자들은 피브릴과 올리고머는 SDS-저항성 응집체(다중 고 분자량) 및 SDS 가용성 응집체, 모노머로 16 kDa 분자량을 나타냄, 모두를 포함하는 것을 발견하였다(도 1H, I ).We produce oligomers and fibrils from recombinant human α-synuclein; Electron microscopy (EM), size exclusion chromatography (SEC), and gel electrophoresis confirmed their physical properties. [Alpha] -Synuclein oligomers were spherical, curvy, and short elongated structures on EM. It appears as a heterogeneous mixture (Fig. 1C-F). Based on SEC data, about half of the proteins in the oligomers were high molecular weight oligomers and the other half were monomers (FIG. 1G, H). By silver staining and western blotting we found that fibrils and oligomers contain both SDS-resistant aggregates (multiple high molecular weights) and SDS soluble aggregates, showing 16 kDa molecular weight as monomers (FIG. 1H, I). ).
먼저 본 발명자들은 BV-2 미세아교세포주에서 α-시누클레인 피브릴의 업테이크에 대한 네 클론의 단클론 항체의 효과를 평가하였다. 이들 중, 169 및 274 두 클론은 업테이크를 촉진한 반면, 다른 두 클론(62 및 171)은 효과가 없었다(도 2A,B). 이 항체들의 효과는 용량 의존적이고 포화 지점에 도달한다(도 2C). 이 예비적인 특성화에 기초하여 274 항체를 본 발명에서 선택하여 사용하였다. 274 항체는 인간 형의 α-시누클레인에 특이적이고, 단백질의 C 말단에 에피토프가 존재한다. 본 발명자들은 274 항체 또는 대조군 IgG의 존재 하에서 BV-2 미세아교세포주에서 α-시누클레인 응집체의 업테이크 및 분해 속도를 비교하였다. 업테이크 실험을 위하여, 5μg/ml의 274 항체 또는 대조군 IgG로 미리 배양된 0.2 μM α-시누클레인 응집체를 세포에 첨가하였다. 세포들을 37℃에서 배양 후 기재된 시간에 수집하였다. 분해 실험을 위하여, 그 응집체를 피브릴 및 올리고머 각각에 대해서 10분 및 5 분 동안 미리배양하였고 그 세포들을 세척하고 기재된 시간 동안 신선한 배양 배지에서 37℃에서 배양하였다. 274 항체의 존재에서, 흡수된(internalized) 피브릴 α-시누클레인의 최대 수준에 도달하는데 필요한 시간은 30에서 10 분으로 감소하였고(도 3A), 들어온(imported) 피브릴 α-시누클레인의 반감기는 77에서 11 분으로 감소하였다(도 3B). 올리고머 α-시누클레인 응집체의 제거의 경우, 최대 수준에 도달하기 위한 흡수된 α-시누클레인 올리고머에 필요한 시간은 274 항체의 존재에서 11에서 4분으로 되었고 흡수된 올리고머의 반감기는 18에서 6 분으로 감소하였다(도3C,D). 한편 62 항체(도 2)의 첨가는 BV-2 세포에서 α-시누클레인 피브릴 또는 올리고머의 업테이크 및 분해 속도에 영향이 없었고(데이터 도시 안함), 이것은 이러한 효과가 항체 특이적이라는 것을 시사한다. 피브릴 및 올리고머와 대조적으로, 모노머에 대한 업테이크 속도는 미세아교세포에서 274 항체의 첨가에 의하여 변화가 없었다(도시 안함). 이들 결과는 274 항체의 존재에서, 모노머가 아닌 세포외α-시누클레인 응집체의 업테이크 및 분해는 미세아교세포에서 촉진된다는 것을 나타낸다.We first evaluated the effect of monoclonal monoclonal antibodies on the uptake of α-synuclein fibrils in BV-2 microglia cell line. Of these, two clones 169 and 274 promoted uptake, while the other two clones 62 and 171 were ineffective (FIGS. 2A, B). The effect of these antibodies is dose dependent and reaches the point of saturation (FIG. 2C). Based on this preliminary characterization, 274 antibodies were selected and used in the present invention. The 274 antibody is specific for α-synuclein of the human type, with an epitope at the C terminus of the protein. We compared the uptake and degradation rates of α-synuclein aggregates in BV-2 microglia in the presence of 274 antibody or control IgG. For the uptake experiments, 0.2 μM α-synuclein aggregates previously incubated with 5 μg / ml of 274 antibody or control IgG were added to the cells. Cells were collected at the indicated time after incubation at 37 ° C. For digestion experiments, the aggregates were preincubated for 10 and 5 minutes for fibrils and oligomers respectively, and the cells were washed and incubated at 37 ° C. in fresh culture medium for the stated time. In the presence of 274 antibody, the time required to reach the maximum level of internalized fibril α-synuclein was reduced from 30 to 10 minutes (FIG. 3A) and the half-life of imported fibrils α-synuclein Decreased from 77 to 11 minutes (FIG. 3B). For removal of oligomer α-synuclein aggregates, the time required for absorbed α-synuclein oligomers to reach maximum levels was 11 to 4 minutes in the presence of 274 antibodies and the half-life of the absorbed oligomer was 18 to 6 minutes. Decreased (Figure 3C, D). The addition of 62 antibodies (FIG. 2) on the other hand had no effect on the uptake and degradation rates of α-synuclein fibrils or oligomers in BV-2 cells (data not shown), suggesting that this effect is antibody specific. . In contrast to the fibrils and oligomers, the uptake rate for the monomers was unchanged by the addition of 274 antibody in the microglia (not shown). These results indicate that in the presence of 274 antibody, uptake and degradation of extracellular α-synuclein aggregates, but not monomers, are promoted in microglia.
뇌 세포 환경을 더욱 재생하기 위하여, 본 발명자들은 쥐 1차 미세아교세포 배양을 하여 이들 미세아교세포가 뉴런 세포로부터 분비된 α-시누클레인 단백질의 업테이크하는 속도를 측정하였다. 이 실험을 위하여, 본 발명자들은 인간 α-시누클레인을 과발현하는 분화된 SH-SY5Y 신경모세포종(neuroblastoma) 세포로부터 얻은 조건화된 배지를 사용하였다. 그 조건화된 배지는 모노머 및 SDS 안정적인 응집체 모두를 포함한다(도 1I). 대조군 IgG로 처리된 세포와 비교하여, 뉴런 세포로부터 분비된 a-시누클레인의 업테이크는 274 항체 처리된 1차 미세아교세포에서 더 빠르고 높은 수준에 도달하였고(도 3E,F),그것은 그 항체가 뉴런 분비된 세포외 a-시누클레인의 제거에서 미세아교세포를 돕는다는 것을 시사한다.In order to further regenerate the brain cell environment, the inventors cultured rat primary microglia to determine the rate at which these microglia uptake the α-synuclein protein secreted from neuronal cells. For this experiment, we used conditioned media obtained from differentiated SH-SY5Y neuroblastoma cells that overexpress human α-synuclein. The conditioned medium contains both monomeric and SDS stable aggregates (FIG. 1I). In comparison to cells treated with control IgG, uptake of a-synuclein secreted from neuronal cells reached faster and higher levels in 274 antibody treated primary microglia (FIG. 3E, F), which antibody Suggests that it helps microglia in the removal of neuronal secreted extracellular a-synuclein.
본 발명자들은 다음으로 분화된 SH-SY5Y 인간 신경모세포종(neuroblastoma) 세포뿐만 아니라 쥐 1차 성상세포 및 피질(cortical) 뉴런에서 외래(exogenous) α-시누클레인 피브릴의 업테이크 및 분해 속도에 대한 274 항체의 효과를 조사하였다. 미세아교세포와 대조적으로, 세포외 α-시누클레인 피브릴의 업테이크 또는 분해 속도의 변화가 이들 세포에서는 관찰되지 않았다(도 4A-F). 유사하게, 모노머의 업테이크는 성상세포 또는 뉴런에서 그 항체에 의하여 영향이 없었다(도시 안함). 따라서 세포외 α-시누클레인의 항체 매개된 제거는 미세아교세포에 선택적인 과정이고 모노머가 아니라 응집체에만 적용된다는 것을 나타낸다.We next describe the rate of uptake and degradation of exogenous α-synuclein fibrils in differentiated SH-SY5Y human neuroblastoma cells as well as rat primary astrocytes and cortical neurons. The effect of the antibody was investigated. In contrast to microglia, no change in the uptake or degradation rate of extracellular α-synuclein fibrils was observed in these cells (FIGS. 4A-F). Similarly, uptake of monomer was not affected by the antibody in astrocytes or neurons (not shown). Thus, antibody mediated removal of extracellular α-synuclein is a selective process for microglia and only applies to aggregates, not monomers.
α-시누클레인 응집체의 항체 지원된 제거는 라이소좀으로 신속한 배달을 야기하는 Fcγ 수용체에 의하여 매개됨Antibody-assisted clearance of α-synuclein aggregates is mediated by Fcγ receptors resulting in rapid delivery to lysosomes
항체 매개된 α-시누클레인 제거에서 Fcγ 수용체의 역할을 평가하기 위하여, Fcγ 수용체를 Fcγ 수용체 II 및 III을 블럭하는 CD16/CD32에 대한 단클론 항체(Wijngaarden S, van Roon JA, van de Winkel JG, Bijlsma JW, Lafeber FP(2005) Rheumatology 44:729-734)로 배양하여 억제하였다. Fcγ 수용체의 봉쇄(Blockage)는 α-시누클레인 올리고머의 업테이크에 대한 274항체의 효과를 제거하였다 (도5A). Fcγ 수용체는 대조군 IgG 또는 274항체의 Fab 절편의 존재시가 아니라 274 항체의 존재시에만 BV-2 세포의 표면 상에서 올리고머 α-시누클레인와 함께 존재하였고(도 5B), 그것은 업테이크 과정에서 Fcγ 수용체의 관여를 증명한다. Fcγ 수용체는 BV-2 미세아교세포 및 쥐 1차 미세아교세포에서 현저하게 발현을 나타내었으나 성상세포 또는 분화된 신경모세포종 세포에서는 검출되지 않았다(도 5C).타입 II 및 타입 III (CD32/CD16) Fcγ 수용체는 IgG에 대한 낮은 친화도 수용체이고 높은 결합력을 위해서는 다가(multivalent) 항원-항체 복합체를 필요로 한다(Ravetch JV, Bolland S (2001) Annu Rev Immunol19:275-290).따라서 항체 단독으로는 높은 친화도를 가지고 이들 Fcγ 수용체에 결합할 수 없다. α-시누클레인-항체 면역 복합체의 업테이크를 확인하기 위하여, 대조군 IgG 또는 단클론 항체 274를 여러 양의 α-시누클레인 피브릴과 미리 배양하고 BV-2 세포에 첨가하였다. 흡수된 항체의 수준은 외래 α-시누클레인 응집체의 양에 직접적으로 비례하였다(도5D). α-시누클레인과 미리배양된 대조군 IgG는 세포에 의하여 흡수되지 아니하였고,외래 β-시누클레인과 미리배양된 274항체도 그러하였다(도 5D). 또, 면역형광 분석은 외래 α-시누클레인 응집체의 존재에서 항체 업테이크에서 큰 증가를 나타내었고, α-시누클레인와 항체의 공동존재는 흡수 후 원형질에서 증명되었다(도 5E,F). 전체적으로, 이들 결과는 미세아교세포에서 α-시누클레인-항체 복합체의 특이적인 흡수를 시사한다.To assess the role of the Fcγ receptor in antibody mediated α-synuclein removal, monoclonal antibodies against CD16 / CD32 that block Fcγ receptors II and III (Wijngaarden S, van Roon JA, van de Winkel JG, Bijlsma) JW, Lafeber FP (2005) Rheumatology 44: 729-734) and inhibited. Blockage of the Fcγ receptor eliminated the effect of 274 antibodies on uptake of the α-synuclein oligomer (FIG. 5A). Fcγ receptors were present with oligomer α-synuclein on the surface of BV-2 cells only in the presence of 274 antibodies, not in the presence of control IgG or Fab fragments of 274 antibodies (FIG. 5B), which was responsible for the uptake process of Fcγ receptors. Prove your involvement. Fcγ receptors were markedly expressed in BV-2 microglia and rat primary microglia but were not detected in astrocytic or differentiated neuroblastoma cells (FIG. 5C). Type II and Type III (CD32 / CD16) Fcγ receptors are low affinity receptors for IgG and require a multivalent antigen-antibody complex for high avidity (Ravetch JV, Bolland S (2001) Annu Rev Immunol 19: 275-290). It has a high affinity and cannot bind to these Fcγ receptors. To confirm the uptake of the α-synuclein-antibody immune complexes, control IgG or monoclonal antibody 274 was precultured with various amounts of α-synuclein fibrils and added to BV-2 cells. The level of absorbed antibody was directly proportional to the amount of foreign α-synuclein aggregates (FIG. 5D). α-synuclein and pre-cultured control IgG were not taken up by the cells, as did the foreign β-synuclein and 274 antibodies pre-cultured (FIG. 5D). In addition, immunofluorescence analysis showed a large increase in antibody uptake in the presence of foreign α-synuclein aggregates, and co-existence of α-synuclein and antibody was demonstrated in the plasma after absorption (FIGS. 5E, F). Overall, these results suggest specific uptake of α-synuclein-antibody complexes in microglia.
다음으로 본 발명자들은 α-시누클레인 단독에 의해서 수행되는 경로와 비교하여 α-시누클레인-항체 면역 복합체의 세포내 수송(trafficking) 경로를 조사하였다. 이를 위하여, 본 발명자들은 clathrin-매개된 엔도사이토시스의 마커로 형광 표지된 저밀도 리포프로틴(LDL), 및 지질 raft 매개된 엔도사이토시스에 대한 마커로 콜레라 톡신 B 서브유닛(CTB)(Sharma DK, Brown JC, Choudhury A, Peterson TE, Holicky E, Marks DL,Simari R, Parton RG, Pagano RE (2004) Mol Biol Cell15:3114-3122)을 사용하였다. 대조군 항체의 존재에서, 흡수된 α-시누클레인 응집체 단독으로는 어느 한 마커와 광범위하게 공동으로 존재하지 않았고(도 6A), 그것은 새로운 수송 기작을 시사한다. 그러나 α-시누클레인 응집체를 274 항체와 함께 적용한 경우, 많은 부분의 흡수된 α-시누클레인 응집체가 CTB와 공동으로 존재하였다(도 6A).The inventors next examined the intracellular trafficking pathway of the α-synuclein-antibody immune complex as compared to the pathway performed by α-synuclein alone. To this end, we have described low density lipoproteins (LDL) fluorescently labeled with markers of clathrin-mediated endocytosis, and cholera toxin B subunit (CTB) (Sharma DK,) as markers for lipid raft mediated endocytosis. Brown JC, Choudhury A, Peterson TE, Holicky E, Marks DL, Simari R, Parton RG, Pagano RE (2004) Mol Biol Cell 15: 3114-3122). In the presence of the control antibody, the absorbed α-synuclein aggregates alone did not exist extensively in common with either marker (FIG. 6A), which suggests a new transport mechanism. However, when the α-synuclein aggregate was applied with the 274 antibody, a large portion of the absorbed α-synuclein aggregate coexisted with the CTB (FIG. 6A).
이 결과들을 확인하기 위하여, 본 발명자들은 면역위치화 실험을 수행하였다. 흡수된 α-시누클레인 응집체는 골지체 마커인 GM130과 공동으로 존재하지 않았고, 이것은 생합성 경로와 중복되는 결핍을 시사한다(도시 안함). 한편, 274 항체로 배양한 경우,약 38.6%의 흡수된 α-시누클레인 응집체가 caveolin-1에 대한 포지티브 구역에서 발견된 반면에, 대조군 IgG의 존재에서, 그들은 거의 caveolin-1와 함께 존재하지 않았다(도 6B,C).To confirm these results, we performed an immunolocalization experiment. Absorbed α-synuclein aggregates did not co-exist with the Golgi apparatus marker GM130, which suggests a deficiency overlapping with the biosynthetic pathway (not shown). On the other hand, when incubated with 274 antibodies, about 38.6% of the absorbed α-synuclein aggregates were found in the positive zone for caveolin-1, whereas in the presence of control IgG they were rarely present with caveolin-1. (Figures 6B, C).
이것은 274 항체가 CTB에 의하여 수행된 흡수된 α-시누클레인의 수송 경로를 변화시킨다는 결과와 일치한다(도 6A). 수송 경로에서 변화는 라이소좀에서 증가된 공동존재를 야기한다; 274 항체의 존재에서, 라이소좀 마커인 카텝신 D와 α-시누클레인의 공동존재는 28에서 93%로 증가되었다(도 6D,E). 종합적으로, 이들 결과는 항체와 복합체를 형성한 경우에 α-시누클레인 응집체는 Fcγ 수용체를 통하여 흡수되고 다른 세포내 수소 경로를 수행하고 자유 α-시누클레인 응집체에 비하여 더 효과적으로 라이소좀에 배달되고 거기서 더 빠르게 분해될 수 있다는 것을 시사한다.This is consistent with the result that 274 antibody alters the transport pathway of absorbed α-synuclein performed by CTB (FIG. 6A). Changes in the transport pathway lead to increased coexistence in lysosomes; In the presence of 274 antibody, the co-existence of the lysosomal marker cathepsin D with α-synuclein was increased from 28 to 93% (Figure 6D, E). Overall, these results indicate that, when complexed with antibodies, α-synuclein aggregates are absorbed through Fcγ receptors, perform different intracellular hydrogen pathways, and are delivered to lysosomes more effectively than free α-synuclein aggregates. Suggests that it can decompose faster.
274 항체는 tg 마우스 모델에서 신경퇴행 질환, 행동 장애 및 α-시누클레인의 세포에서 세포 전달을 감소 274 Antibodies Reduce Neurodegenerative Diseases, Behavioral Disorders and Cell Delivery in Cells of α-Synuclein in a tg Mouse Model
세포외 α-시누클레인 응집체의 항체 지원된 제거가 세포내 응집체 전달을 억제하는지를 결정하기 위하여 본 발명자들은 274 항체를 인간α-시누클레인을 발현하는 tg 마우스의 해마에 주사하고 α-시누클레인 단백질의 뉴런에서 성상세포로 전달의 양을 분석하였다.To determine whether antibody-assisted removal of extracellular α-synuclein aggregates inhibits intracellular aggregate delivery, we injected 274 antibodies into the hippocampus of tg mice expressing human α-synuclein and The amount of transfer from neurons to astrocytes was analyzed.
대조군 IgG의 주사는 뉴런에서 성상세포로 α-시누클레인 전달에 대한 효과를 나타내지 않았다; 해마의 동측(ipsilateral) 및 대측(contralateral) 면 모두는 동일한 정도의 성상아교세포(astroglial) α-시누클레인 축적을 나타내었다(도 7A,E,K). 반면에, 274 항체의 주사는 동측면에서 α-시누클레인의 성상아교세포 축적의 현저한 감소를 야기하였다(도 7B,F,L). 항체의 주사는 뉴런에서 α-시누클레인 발현에 영향은 없었고(도 7C,D), 성상세포의 수도 변화시키지 않았으며 (도 7G,H), 따라서 항체 처리가 α-시누클레인의 세포에서 세포 전달을 억제한다는 것을 시사한다. 대조군 IgG의 주사는 미세아교세포에 대한 효과를 나타내지 않았다(도 7I ); 그러나 274항체로 주사된 해마의 동측 및 대측 면 모두는 미세아교 활성화에서 약한 증가를 나타내었다(도 7J ).Injection of control IgG showed no effect on α-synuclein delivery from neurons to astrocytes; Both the ipsilateral and contralateral sides of the hippocampus showed the same degree of astroglial α-synuclein accumulation (FIGS. 7A, E, K). In contrast, injection of the 274 antibody resulted in a significant decrease in astrocyte glial accumulation of α-synuclein (FIGS. 7B, F, L). Injection of the antibody had no effect on α-synuclein expression in neurons (FIG. 7C, D) and did not change the number of astrocytic cells (FIG. 7G, H), thus antibody treatment resulted in cell transfer in cells of α-synuclein. Suggests that Injection of control IgG showed no effect on microglia (FIG. 7I); However, both ipsilateral and contralateral sides of the hippocampus injected with 274 antibodies showed a slight increase in microglial activation (FIG. 7J).
세포에서 세포 전달의 봉쇄에서 미세아교세포의 관여를 확인하기 위하여 본 발명자들은 이중 면역형광 염색을 사용하여 미세아교에서 인간 α-시누클레인 및 274 항체의 위치를 조사하였다.To confirm the involvement of microglia in the blockade of cell delivery in the cells, we used the dual immunofluorescence staining to investigate the position of human α-synuclein and 274 antibodies in microglia.
인간 α-시누클레인을 포함하는 미세아교세포의 수는 274 항체로 주사된 마우스에서 동측면에서 현저한 증가를 나타내었다(도 8A,B). 주사된 항체는 274 항체로 주사된 tg 마우스에서 동측면에서 미세아교세포에서도 검출되었지만(도 8C,D), 대조군 IgG로 주사된 마우스에서는 그러하지 않았다(도 8D). α-시누클레인 및 주사된 항체의 같은위치 존재는 274 항체로 주사된 tg 마우스에서 일치되게 관찰되었지만(도 8E), 대조군 IgG로 주사된 마우스에서는 그러하지 않았다. 이 결과들은 α-시누클레인 항체가 미세아교세포에 의한 뉴런 유래된 α-시누클레인의 제거를 촉진하여 α-시누클레인 응집체의 세포에서 세포 전달을 억제한다는 것을 시사한다.The number of microglia containing human α-synuclein showed a significant increase ipsilaterally in mice injected with 274 antibodies (FIGS. 8A, B). The injected antibody was also detected in microglia in ipsilaterally in tg mice injected with 274 antibodies (FIG. 8C, D), but not in mice injected with control IgG (FIG. 8D). The co-location of α-synuclein and injected antibodies was consistently observed in tg mice injected with 274 antibodies (FIG. 8E), but not in mice injected with control IgG. These results suggest that α-synuclein antibodies promote the removal of neuron-derived α-synuclein by microglia to inhibit cell delivery in cells of α-synuclein aggregates.
274 항체 주사에 수반하는 α-시누클레인의 미세아교세포 지원된 제거에 대한 카이네틱스를 평가하기 위하여, α-시누클레인 및 Iba-1에 대한 항체로 이중 표지를 여러 시간 지점에서 수행하였다(도 8F). 주사 1일 후에,약 20%의 미세아교세포가 α-시누클레인 축적을 나타낸 반면, 7일 및 14일에서 약 10%의 미세아교세포가 α-시누클레인을 가졌고, 28일에는 약 5%로 감소하였다(도 8G).To assess kinetics for microglia supported removal of α-synuclein following 274 antibody injections, double labeling with antibodies to α-synuclein and Iba-1 was performed at various time points (FIG. 8F). After 1 day of injection, about 20% of the microglia showed α-synuclein accumulation, while on day 7 and 14 about 10% of the microglia had α-synuclein and at about 5% on day 28 Decreased (FIG. 8G).
274 항체가 뉴런 및 성상아교세포로부터 α-시누클레인의 전달을 저해하는지를 확인하기 위하여, 이중 표지 연구를 NeuN 및 GFAP 항체로 수행하였다.To confirm whether the 274 antibody inhibited the transfer of α-synuclein from neurons and astroglia, a dual labeling study was performed with NeuN and GFAP antibodies.
공초점 지원된 이미지는 피라미드형 NeuN-포지티브 뉴런과 같은위치에 존재하는 α-시누클레인이 약 20%의 세포를 나타낸다는 것을 나타내었고(도 8H), IgG 및 274 항체 군 사이의 현저한 차이가 관찰되지 않았다(도 8I). 대조적으로 방추형을 가지는 α-시누클레인-포지티브 세포는 NeuN과 동시에 존재하지 아니하고, 차라리 GFAP와 존재한다(도 8J). 약 15%의 GFAP 세포는 대조군에서 α-시누클레인을 포함하였다. 그러나 274 항체로 처리는 주사 부위에서 GFAP/α-시누클레인-포지티브 세포의 감소를 야기하였다(도 8K).Confocal supported images showed that α-synuclein present in the same position as the pyramidal NeuN-positive neurons represented about 20% of the cells (FIG. 8H) and noticeable differences between the IgG and 274 antibody groups were observed. (FIG. 8I). In contrast, the α-synuclein-positive cells with fusiforms do not exist simultaneously with NeuN, but rather with GFAP (FIG. 8J). About 15% of GFAP cells contained α-synuclein in the control group. However, treatment with 274 antibody resulted in a decrease in GFAP / α-synuclein-positive cells at the injection site (FIG. 8K).
GFAP 및 Iba-1-포지티브 세포의 이미지 분석은 자국을 따르는 흔적(scar)의 비특이적인 효과를 회피하기 위하여 바늘 자국의 부위 밑에 해마에서 수행하였다. 주사자국을 따라 성상교세포증(astrogliosis)은 주사 후 4 주간 지속된 반면에 자국을 따라 미세아교세포 반응은 일시적이고 2주 후에 감소하기 시작하였다. 유사하게 해마(주사 자국 밑)에서 α-시누클레인을 함유한 활성화된 미세아교세포의 수는 시간에 따라 감소하였다(도 8F,G). Image analysis of GFAP and Iba-1-positive cells was performed in the hippocampus underneath the site of the needle tread to avoid the nonspecific effect of the scar along the tread. Along with the injection traces, astrogliosis lasted 4 weeks after injection, while the microglial response along the traces was transient and began to decrease after 2 weeks. Similarly, the number of activated microglia containing α-synuclein in the hippocampus (under injection marks) decreased with time (FIG. 8F, G).
기능적 및 신경병리학적 파라미터에 대한 세포외 α-시누클레인 응집체의 항체 지원된 제거의 효과를 더 평가하기 위하여 수동 면역 실험을 non-tg 및 tg 마우스로 대조군 IgG 또는 274 항체의 복강내 주사에 의하여 수행하였다. 폴(pole) 테스트에서 기능적 모터 연합(coordination)의 분석은 IgG 및 274-처리된 non-tg와 비교하여, IgG-처리된 tg 마우스는 그 패러다임을 완성하는데 더 오랜 시간이 소요되었다(도 9A). 대조적으로, 274 항체로 처리된 tg 마우스는 non-tg 군과 유사하게 수행되었다(도 9A). 오픈 필드에서, IgG-처리된 tg 마우스는 non-tg와 비교하여 일부 과민활성을 나타내었다(도 9B). α-시누클레인 tg 마우스에서 이 효과는 274 항체의 처리에 의하여 역전되었다(도 9B). 전체 거리 및 사육(rearing)을 포함한 오픈 필드에서 다른 활성들은 네 군 중에서 차이가 없었다(도 9C,D).To further assess the effect of antibody-assisted clearance of extracellular α-synuclein aggregates on functional and neuropathological parameters, passive immunoassays were performed by intraperitoneal injection of control IgG or 274 antibodies with non-tg and tg mice. It was. Analysis of functional motor coordination in the pole test compared to IgG and 274-treated non-tg, IgG-treated tg mice took longer to complete the paradigm (FIG. 9A). . In contrast, tg mice treated with 274 antibodies performed similarly to the non-tg group (FIG. 9A). In the open field, IgG-treated tg mice showed some hypersensitivity compared to non-tg (FIG. 9B). In α-synuclein tg mice this effect was reversed by treatment of 274 antibody (FIG. 9B). Other activities in the open field, including total distance and breeding, did not differ among the four groups (Figures 9C, D).
종간(cross-species) 다클론 항체로, α-시누클레인 면역염색은 대조군 IgG 및 274 항체 군 사이에서 관찰된 차이가 없는 것을 가지는 non-tg 마우스에서 신경망(neuropil)에 제한되었다(도 9E-G). IgG-처리된 α-시누클레인 tg 마우스에서,신피질(neocortex) (도 9H) 및 해마(도 9I)에서 성상아교세포 (도 9F) 및 뉴런 세포(도 9G) α-시누클레인 (도 9E)의 광범위한 축적이 있었다. 반면, 274 항체로 처리된 α-시누클레인 tg 마우스(도 9E)는 신피질(neocortex) (도 9H) 및 해마(도 9I)에서 성상아교세포 (도 9F) 및 뉴런 α-시누클레인(도 9G)의 축적에서 상당한 감소를 나타내었다.With cross-species polyclonal antibodies, α-synuclein immunostaining was restricted to neuropil in non-tg mice with no differences observed between the control IgG and 274 antibody groups (FIG. 9E-G). ). In IgG-treated α-synuclein tg mice, of astrocytes (FIG. 9F) and neuronal cells (FIG. 9G) in α-synuclein (FIG. 9E) in neocortex (FIG. 9H) and hippocampus (FIG. 9I). There was extensive accumulation. In contrast, α-synuclein tg mice treated with 274 antibodies (FIG. 9E) were found to have astrocytes (FIG. 9F) and neurons α-synuclein (FIG. 9G) in neocortex (FIG. 9H) and hippocampus (FIG. 9I). There was a significant decrease in the accumulation of.
본 발명자들은 또한 수동적으로 면역화된 마우스의 선조체(striatum)를 분석하였고 α-시누클레인의 레벨이 피질 및 해마와 비교할 때 더 낮은 정도로 감소하였다는 것을 발견하였다.We also analyzed the striatum of passively immunized mice and found that the levels of α-synuclein were reduced to a lower extent compared to the cortex and hippocampus.
피질 및 해마에서 274는 α-시누클레인의 레벨을 약 70-80%로 감소시키고(도 9H, I); 선조체에서는 그 레벨은 30-35%로 감소되었고(도 9J), 이것은 항체의 차별적인 부위별 수송을 시사한다.274 in the cortex and hippocampus reduced the level of α-synuclein to about 70-80% (FIG. 9H, I); In the striatum, the level was reduced to 30-35% (FIG. 9J), suggesting differential site-specific transport of antibodies.
본 발명자들은 대조군 및 항체 처리된 α-시누클레인 마우스의 CSF 및 뇌에서α-시누클레인에 대한 ELISA를 수행하였다. 비히클 처리된 α-시누클레인 tg 마우스의 뇌 균질체(homogenates)에서 α-시누클레인의 레벨은 13.57±1.610 ng/ml (n=8)인 반면, 274 항체로 처리된 α-시누클레인 tg는 5.031±1.011 ng/ml (n= 8)이었다. 그러나, CSF에서 α-시누클레인의 레벨은 본 발명의 ELISA 시스템의 검출 한계 하에서 매우 낮다.We performed ELISA for α-synuclein in CSF and brain of control and antibody treated α-synuclein mice. The level of α-synuclein in brain homogenates of vehicle treated α-synuclein tg mice was 13.57 ± 1.610 ng / ml (n = 8), whereas α-synuclein tg treated with 274 antibody was 5.031. ± 1.011 ng / ml (n = 8). However, the level of α-synuclein in CSF is very low under the detection limits of the ELISA system of the present invention.
α-시누클레인-함유 미세아교세포는 신경망에서 주로 관찰되었고 혈관 주위에서는 거의 관찰되지 않았다.α-synuclein-containing microglia were observed mainly in neural networks and rarely around the blood vessels.
수동 면역에서 α-시누클레인의 제거에 대한 274항체의 효과를 더 평가하기 위하여, NeuN, GFAP, 및 Iba-1에 대한 항체로 이중 표지 연구를 수행하였다. 공초점 이미지 분석은 α-시누클레인 면역반응성을 나타낸 피라미드형 NeuN-포지티브 세포의 비율은 274 항체로 주사한 군에서 감소하였다(도 9K,L)는 것을 나타낸다. 게다가, IgG로 처리한 α-시누클레인 tg 마우스와 비교하여, 274 항체로 처리는 α-시누클레인을 포함한 방추형 GFAP 포지티브 성상아교세포의 감소를 야기하였다(도 9M,N).To further assess the effect of 274 antibodies on the elimination of α-synuclein in passive immunization, a double labeling study was conducted with antibodies to NeuN, GFAP, and Iba-1. Confocal image analysis indicated that the proportion of pyramidal NeuN-positive cells showing α-synuclein immunoreactivity was decreased in the group injected with 274 antibodies (FIG. 9K, L). In addition, compared to α-synuclein tg mice treated with IgG, treatment with 274 antibody resulted in the reduction of fusiform GFAP positive astroglia including α-synuclein (FIG. 9M, N).
반면, IgG 단독으로 면역화된 α-시누클레인 tg 마우스와 비교하여, 274 항체로 처리는 α-시누클레인을 포함한 Iba-1-포지티브 미세아교세포의 증가를 야기하였다 (도 9 O,P). 본 발명자들은 또한 α-시누클레인 및 cathepsin D에 대한 이중 표지 분석을 수행하고 274로 면역화된 동물의 미세아교세포에서 두 마커 사이에 같은 위치에 존재하는 것을 발견하였다 (도 9Q,R).In contrast, treatment with 274 antibodies resulted in an increase in Iba-1-positive microglia with α-synuclein, as compared to α-synuclein tg mice immunized with IgG alone (FIG. 9 O, P). We also performed double labeling assays for α-synuclein and cathepsin D and found that they were present in the same position between the two markers in microglia of animals immunized with 274 (FIG. 9Q, R).
신경퇴행 및 신경염증 병리학에 대한 수동 면역의 효과를 연구하기 위하여 면역조직화학 및 이미지 분석을 수행하였다. NeuN에 대한 항체로, non-tg 대조군과 비교하여,IgG-처리된 α-시누클레인 tg 마우스는 해마에서 입체학적 신경세포 평가에서 감소를 나타내었다(도 10A,B). 뉴런의 손실은 해마의 CA3 부위에서 명확하였다(도 10A, 화살표). 반면, 274항체로 처리된 α-시누클레인 tg 마우스는 non-tg 대조군 상당하는 해마 NeuN 세포 카운트를 나타내었다(도 10A,B).Immunohistochemistry and image analysis were performed to study the effects of passive immunity on neurodegeneration and neuroinflammatory pathology. As antibodies to NeuN, IgG-treated α-synuclein tg mice showed a decrease in stereoscopic neuronal evaluation in the hippocampus compared to non-tg controls (FIGS. 10A, B). The loss of neurons was evident at the CA3 site of the hippocampus (FIG. 10A, arrow). In contrast, α-synuclein tg mice treated with 274 antibodies showed hippocampal NeuN cell counts corresponding to the non-tg control (FIGS. 10A, B).
시냅스 변화도 분석하였다. 본 발명자들은 IgG-처리된 tg에서 시냅토파이신(synaptophysin)에서 28%감소를 발견하였고, 이 시냅스 손상은 274 항체로 처리에 의하여 현저하게 완화되었다(도 10C,D). GFAP에 대한 항체로 면역조직화학은 non-tg 대조군과 비교하여, α-시누클레인 tg 마우스는 신피질(neocortex)의 깊은 층(도 10E, 삽도) 및 해마(도 10E,F)에서 증가된 성상교세포증(astrogliosis)을 나타내었다는 것을 보였다. 274 항체로 수동면역은 α-시누클레인 tg 마우스에서 신피질(도 10E,삽도) 및 해마(도 10E,F)에서 GFAP 면역반응성의 레벨을 감소시켰다. 최종적으로, 미세아교세포 마커, Iba-1에 대한 항체로 미세아교세포 카운트를 수행한 결과, 유사한 레벨의 미세아교세포가 IgG 대조군으로 처리된 a-시누클레인 tg 마우스 뿐만 아니라 IgG 및 274 항체로 처리된 non-tg 마우스 사이에서 검출되었다(도 10G). 그러나, 274항체로 처리된 α-시누클레인 tg 마우스는 Iba-1 면역염색의 레벨에서 적당한 증가를 나타내었다(도 10G,H). 이것과 일치하게 IgG 단독으로 처리된 α-시누클레인 tg 마우스는 전 면역성 사이토카인, TNFα (도 10 I, J ), 및 IL-6 (도 10K,L)의 증가된 레벨을 나타낸 반면, 274 항체로 면역화는 그 레벨을 기준선까지로 감소시켰다. 이 결과는 274 항체가 세포외 α-시누클레인의 제거를 촉진한다는 결과와 일치한다. Synaptic changes were also analyzed. We found a 28% reduction in synaptophysin in IgG-treated tg, and this synaptic damage was significantly alleviated by treatment with 274 antibody (FIG. 10C, D). Immunohistochemistry with antibodies to GFAP compared to non-tg controls, α-synuclein tg mice increased astrocytes in the deep layers of neocortex (Figure 10E, inset) and hippocampus (Figures 10E, F). It was shown that it showed astrogliosis. Passive immunization with 274 antibody reduced levels of GFAP immunoreactivity in the neocortex (FIG. 10E, inset) and hippocampus (FIG. 10E, F) in α-synuclein tg mice. Finally, microglial counts were performed with an antibody against the microglia marker, Iba-1, which resulted in similar levels of microglia treated with IgG and 274 antibodies as well as a-synuclein tg mice treated with IgG controls. Was detected between non-tg mice (FIG. 10G). However, α-synuclein tg mice treated with 274 antibodies showed a moderate increase in the level of Iba-1 immunostaining (FIG. 10G, H). Consistent with this, α-synuclein tg mice treated with IgG alone showed increased levels of pro-immune cytokines, TNFα (FIG. 10 I, J), and IL-6 (FIG. 10K, L), whereas 274 antibodies Low immunization reduced that level to baseline. This result is consistent with the result that 274 antibodies promote the removal of extracellular α-synuclein.
종합하면, 이들 결과들은 274 항체로 면역화는 α-시누클레인 tg 마우스에서 기능적인 손상을 감소시키고, 이것은 신경교 및 뉴런 세포에서 α-시누클레인의 축적에서 감소 및 신경퇴행의 완화에 동반된다.Taken together, these results show that immunization with 274 antibodies reduces functional damage in α-synuclein tg mice, which is accompanied by reduction in accumulation of α-synuclein in glial and neuronal cells and alleviation of neurodegeneration.
본 발명을 통하여 알 수 있는 바와 같이, α-시누클레인에 대한 항체는 미세아교세포에 의하여 특이적으로 세포외 α-시누클레인 응집체의 제거를 촉진하고, 특정항체와 복합체를 형성하는 경우, α-시누클레인 응집체는 미세아교세포의 표면 상 Fcγ 수용체를 통하여 흡수되고 흡수 후 그 면역 복합체는 수송경로를 따라서 운송되고 라이소좀으로 더 효과적인 전달을 야기한다. 또한 항체에 의해 매개된 α-시누클레인의 제거는 tg 마우스 모델에서 세포에서 세포로 α-시누클레인의 전달을 억제하였다. 이 결과들은 α-시누클레인 면역치료가 세포외 α-시누클레인의 미세아교세포 매개된 제거의 촉진을 통하여 치료 및 예방효과를 수행할 수 있다는 것을 시사한다.As can be seen through the present invention, the antibody against α-synuclein specifically promotes the removal of extracellular α-synuclein aggregates by microglia and forms α-synuclein when complexed with specific antibodies. Synuclein aggregates are absorbed through Fcγ receptors on the surface of microglial cells and after absorption their immune complexes are transported along the transport pathway and cause more efficient delivery to lysosomes. Removal of α-synuclein mediated by antibodies also inhibited the transfer of α-synuclein from cell to cell in the tg mouse model. These results suggest that α-synuclein immunotherapy can perform therapeutic and prophylactic effects through the promotion of microglia mediated removal of extracellular α-synuclein.
도 1은 α-시누클레인 피브릴 및 올리고머의 특성. A-F, 피브릴의 EM 이미지 (A), 소니케이트된 피브릴 (B), 및 올리고머(C-F). 스케일 바: A-C, 0.5μm; D-F, 30 nm. G, 올리고머의 크기 배제 크로마토그래피. 별표(7 ml 분획)는 올리고머 분획을 나타낸다. 모노머는 13-15 ml 분획에서 존재한다. H, SEC 분획의 실버 염색 이미지. I, 여러 형태의 α-시누클레인의 웨스턴 블럿 분석. M, 모노머; F, 피브릴; O, 올리고머; CM, α-시누클레인을 발현하는 분화된 SH-SY5Y 세포의 조건화된 배지.1 shows the properties of α-synuclein fibrils and oligomers. AF , EM image of fibrils ( A ), sonicated fibrils ( B ), and oligomers ( CF ). Scale bar: AC , 0.5 μm; DF , 30 nm. G , size exclusion chromatography of oligomers. Asterisks (7 ml fractions) indicate oligomeric fractions. Monomers are present in 13-15 ml fractions. Silver stained image of H , SEC fractions. I , Western blot analysis of several forms of α-synuclein. M, monomers; F, fibrils; O, oligomers; CM, conditioned medium of differentiated SH-SY5Y cells expressing α-synuclein.
도 2는 세포외 α-시누클레인 응집체의 업테이크에 대한 단클론 항체의 효과를 나타낸 그림.A, BV-2 미세아교세포에서 α-시누클레인 또는 대조군 IgG (-)에 대한 기재된 단클론 항체의 존재에서 α-시누클레인 피브릴의 흡수.α-시누클레인 피브릴(0.2μM)을 5μg/ml의 대조군 IgG 또는α-시누클레인에 대한 항체들로 상온에서 5분간 미리배양하고 BV-2 세포로 5 분 동안 37℃에서 처리하였다. 흡수된 α-시누클레인의 양을 웨스턴 블럿으로 분석하였다. B, 흡수된 α-시누클레인의 양을 정량화하고 베타-actin의 레벨로 표준화하였다. y-축의 수는 대조군에 대한 α-시누클레인의 양을 나타냄. n= 4, *p〈0.05. C, α-시누클레인 피브릴의 항체 용량 의존적인 업테이크(n=3, p〈0.05) *, 274; #, 169.Figure 2 shows the effect of monoclonal antibodies on the uptake of extracellular α-synuclein aggregates. A , uptake of α-synuclein fibrils in the presence of described monoclonal antibodies against α-synuclein or control IgG (-) in BV-2 microglia. 5 μg / ml α-synuclein fibrils (0.2 μM) Antibodies against control IgG or α-synuclein were preincubated for 5 minutes at room temperature and treated with BV-2 cells at 37 ° C. for 5 minutes. The amount of α-synuclein absorbed was analyzed by Western blot. B , the amount of α-synuclein absorbed was quantified and normalized to the level of beta-actin. The number of y-axes indicates the amount of α-synuclein relative to the control. n = 4, * p <0.05. C , antibody dose dependent uptake of α-synuclein fibrils (n = 3, p <0.05) *, 274; #, 169.
도 3은 미세아교에서 세포외α-시누클레인의 업테이크 및 분해에 대한 274항체의 효과를 나타냄.A-D, BV-2 세포에서 α-시누클레인 피브릴(A,B) 및 올리고머(C,D)의 업테이크 (A,C) 및 분해(B,D)의 속도. 흡수된 α-시누클레인 응집체를 기재된 시간에서 웨스턴 블럿팅에 의하여 분석하였다. A 및 C에서 그래프는 대조군 IgG로 최대 흡수된 레벨과 비교한 α-시누클레인 양의 나타냄. B 및 D에서 그래프는 시간 O에서의 것과 비교한 남아 있는 α-시누클레인의 양을 나타냄. 모든 값들은 베타-actin으로 표준화하였다. 각 그래프의 곡선은 현저하게 차이가 있다(n=4, p〈 0.05). E, F, 1차 미세아교에서 뉴런 분비된 α-시누클레인의 업테이크에 대한 274 항체의 효과를 나타냄. E, 1차 랫트 미세아교를 대조군 IgG 또는 274 항체의 존재 하에서 분화된 SH-SY5Y 세포로부터 분비된 α-시누클레인를 함유하는 조건화된 배지로 처리하였다. F, E에서 데이터의 정량분석. 곡선은 현저하게 다르다( p〈0.05, n=3). 대조군 블럿:베타-tubulin.3 shows the effect of 274 antibodies on uptake and degradation of extracellular α-synuclein in microglia. Rate of uptake ( A , C ) and degradation ( B , D ) of α-synuclein fibrils ( A , B ) and oligomers ( C , D ) in AD , BV-2 cells. Adsorbed α-synuclein aggregates were analyzed by western blotting at the time stated. Graphs in A and C show the amount of α-synuclein compared to the maximum absorbed level with control IgG. Graphs in B and D show the amount of α-synuclein remaining compared to that at time O. All values were normalized to beta-actin. The curves of each graph are significantly different (n = 4, p <0.05). E , F , show the effect of 274 antibodies on uptake of neuron secreted α-synuclein in primary microglia. E , primary rat microglia were treated with conditioned medium containing α-synuclein secreted from differentiated SH-SY5Y cells in the presence of control IgG or 274 antibodies. Quantitative analysis of data at F and E. The curves are significantly different (p <0.05, n = 3). Control blot: beta-tubulin.
도 4는 1차 성상세포 및 신경 세포에서 α-시누클레인 피브릴의 업테이크 및 분해에 대한 274 항체의 효과의 부족을 나타냄. A, B, 1차 성상세포 (n=4). C, D, 1차 피질 뉴런 (n=3). E, F, 분화된 SH-SY5Y 세포 (n=4). A, C, E, 업테이크 률. 그래프는 대조군 IgG로 최대 흡수된 레벨에 대한 α-시누클레인의 양을 나타냄. B, D, F, 분해 속도. 그래프는 시간 O에서의 것과 비교한 남아 있는 α-시누클레인의 양을 나타냄. α-시누클레인 피브릴로 미리배양 시간은 12시간 (B, D, F ). 모든 α-시누클레인 데이터는 베타-actin 데이터로 표준화함. 그래프의 곡선은 현저하게 다르지 않음.4 shows the lack of the effect of 274 antibodies on uptake and degradation of α-synuclein fibrils in primary astrocytes and neurons. A , B , primary astrocytic cells (n = 4). C , D , primary cortical neurons (n = 3). E , F , differentiated SH-SY5Y cells (n = 4). A , C , E , uptake rate. The graph shows the amount of α-synuclein relative to the level of maximum absorption with control IgG. B , D , F , decomposition rate. The graph shows the amount of α-synuclein remaining compared to that at time O. Pre-incubation time with α-synuclein fibrils was 12 hours ( B , D , F ). All α-synuclein data was normalized to beta-actin data. The curve of the graph is not significantly different.
도 5는 α-시누클레인-항체 면역 복합체의 업테이크에서 Fcγ 수용체의 역할을 나타냄. A, BV-2 세포에서 α-시누클레인 올리고머의 흡수에 대한 Fcγ 수용체 블럭킹 항체의 효과.흡수된 α-시누클레인 올리고머를 웨스턴 블럿으로 분석하였고 베타-actin으로 표준화하였다. 그래프의 y-축은 대조군 IgG로 최대 흡수된 레벨에 대한 α-시누클레인의 양을 나타냄. 적색 곡선은 다른 것과 현저하게 다르다 (n=7, p〈0.05).B, Fcγ 수용체 및 α-시누클레인의 면역형광 이미지. 화살표는 세포의 표면 상에 α-시누클레인 및 Fcγ 수용체의 동시존재를 나타냄. 스케일 바: 20μm. C, 여러 세포 타입에서 Fcγ 수용체의 발현. BV-2, 랫트 미세아교, 및 랫트 성상세포를 위해서,설치류에 특이적인 항-CD16/CD32 항체를 사용한 반면에 SH-SY5Y를 위해서, 인간 단백질에 특이적인 항-CD32 항체를 사용하였다. CD16의 발현은 기존 연구에서 뇌 뉴런에서 보고되지 않음. 적색, Fcγ 수용체; 청색, 핵. 스케일 바: 20μm. D, 여러 다른 양의 α-시누클레인 피브릴 또는 α-시누클레인 (0 -330 nM)의 존재에서 BV-2 세포로 항체의 흡수. 흡수된 항체를 항-마우스 IgG 항체로 분석함. 하단 패널은 베타-actin. E, BV-2 세포에서 흡수된 α-시누클레인 피브릴 및 항체의 면역형과 현미경 결과. 스케일 바: 20μm. F, E에서 흡수된 항체로부터 나온 형광을 정량화. 150개 세포를 세 번의 독립적인 실험으로 분석(각 실험 당 50 개의 세포). *p〈0.0001.5 shows the role of Fcγ receptors in the uptake of α-synuclein-antibody immune complexes. A , Effect of Fcγ receptor blocking antibodies on uptake of α-synuclein oligomers in BV-2 cells. The absorbed α-synuclein oligomers were analyzed by Western blot and normalized to beta-actin. The y-axis of the graph shows the amount of α-synuclein relative to the level of maximum absorption with control IgG. The red curve is significantly different from the others (n = 7, p <0.05). Immunofluorescence images of B , Fcγ receptors and α-synuclein. Arrows indicate the co-existence of α-synuclein and Fcγ receptors on the surface of the cells. Scale bar: 20 μm. C , Expression of Fcγ receptors in several cell types. For BV-2, rat microglia, and rat astrocytes, anti-CD16 / CD32 antibodies specific to rodents were used, whereas for SH-SY5Y, anti-CD32 antibodies specific to human proteins were used. Expression of CD16 has not been reported in brain neurons in previous studies. Red, Fcγ receptors; Blue, nuclear. Scale bar: 20 μm. D , uptake of antibody into BV-2 cells in the presence of several different amounts of α-synuclein fibrils or α-synuclein (0 -330 nM). Absorbed antibody was analyzed with anti-mouse IgG antibody. The bottom panel is beta-actin. E , Immunotype and microscopic results of α-synuclein fibrils and antibodies taken up in BV-2 cells. Scale bar: 20 μm. Quantification of fluorescence from antibodies absorbed at F , E. 150 cells were analyzed in three independent experiments (50 cells in each experiment). * p <0.0001.
도 6은 흡수된 α-시누클레인 응집체의 변경된 세포내 수송 및 라이소좀으로 증가된 운반을 나타냄. A,BV-2 세포에서 흡수된 α-시누클레인 피브릴의 변경된 세포내 수송. 형광 표지된 추적자를 대조군 IgG-α-시누클레인 혼합물 또는 α-시누클레인-274 면역 복합체로 BV-2 미세아교 세포에 5 분간 37℃에서 처리하였다. 화살표는 274 항체의 존재 하에서 α-시누클레인 및 CTB 사이에 동시존재를 나타냄. 스케일 바: 5μm. B-E, 식균 구획(endocytic compartment)에서 흡수된 α-시누클레인 응집체의 위치화. 면역 복합체를 BV-2 세포에 5분간 처리. C, E, α-시누클레인 점(puncta)의 수는 caveolin-1 (C)과 동위치에 존재하고 cathepsin D (E)를 정량화하였다. 3000 점을 세 독립적인 실험으로부터 분석하였다 (각 실험 당 1000 점). 스케일 바: 5 μm. *p〈0.005.6 shows altered intracellular transport and increased transport of lysosomes of uptake of α-synuclein aggregates. A , Altered intracellular transport of α-synuclein fibrils uptake in BV-2 cells. Fluorescently labeled tracers were treated with BV-2 microglia cells for 5 min at 37 ° C. with control IgG-α-synuclein mixture or α-synuclein-274 immune complex. Arrows indicate coexistence between α-synuclein and CTB in the presence of 274 antibody. Scale bar: 5 μm. BE , localization of α-synuclein aggregates absorbed in the endocytic compartment. Immune complexes were treated with BV-2 cells for 5 minutes. The number of C , E and α-synuclein points (puncta) is co-located with caveolin-1 ( C ) and quantified cathepsin D ( E ). 3000 points were analyzed from three independent experiments (1000 points for each experiment). Scale bar: 5 μιη. * p <0.005.
도 7은 274 항체의 단독 주사 후 인 비보에서 α-시누클레인의 감소된 뉴런에서 성상세포 전달을 나타냄. A, B, α-시누클레인 면역반응성의 패턴을 비교하는 PDGF-α-시누클레인 tg mice (line M)의 피질 및 해마의 낮은 파위 (20X)뷰. 스케일 바: 200 μm. C-J, 뉴런에서 α-시누클레인 축적을 보여주는 해마 CA1 부위(C, D) 및 신경교세포에서α-시누클레인 축적을 보여주는 치상회(dentate gyrus) 부위(E-J ) (E, F ), GFAP 성상교세포의 존재 (G, H), 및 Iba1 미세아교세포의 존재 (I, J )의 하이 파워 뷰(200X). 스케일 바: 25μm. K, L, α-시누클레인-포지티브 뉴런 및 신경교세포의 수의 이미지 분석. 그룹 당 n=8, 9 월령. * 양방 Student's t 테스트 p〈0.01,unpaired.7 shows astrocytic delivery in reduced neurons of α-synuclein in vivo following alone injection of 274 antibody. A , B , low-pan (20X) view of the cortex and hippocampus of PDGF-α-synuclein tg mice (line M) comparing the patterns of α-synuclein immunoreactivity. Scale bar: 200 μm. CJ , hippocampal CA1 site ( C , D ) showing α-synuclein accumulation in neurons and dentate gyrus site ( EJ ) ( E , F ) showing α-synuclein accumulation in glial cells, GFAP of astrocytes High Power View (200X) of presence ( G , H ), and presence of Iba1 microglia ( I , J ). Scale bar: 25 μm. Image analysis of the number of K , L , α-synuclein-positive neurons and glial cells. N = 8, 9 months per group. * Both Student's t test p <0.01, unpaired.
도 8은 항체의 일방 주사 후 인 비보에서 미세아교 세포에서 α-시누클레인 및 IgG의 증가된 위치화를 나타냄. PDGF-α-시누클레인 tg 마우스(line M)는 도 7과 같이 해마 내 주사 수여됨. A-E에서 이미지들은 주사와 동측 부위와 치상회로부터 얻음. 스케일 바, 10μm. A, α-시누클레인 (적색) 및 미세아교세포 마커(Iba1, 녹색)에 대한 항체로 이중 표지. B, α-시누클레인과 동시 위치하는 Iba-1-포지티브 세포의 비율의 측정용 이중 표지에 대한 이미지 분석. 그룹 당 n=8 , 9 월령. * 양방 Student's t 테스트 p〈0.01,unpaired. C, 마우스 IgG (적색) 및 Iba-1 (녹색)에 대한 항체로 이중 표지. D,마우스 IgG와 동시 위치하는 Iba-1-포지티브 세포의 비율의 측정용 이중 표지에 대한 이미지 분석. 그룹 당 n=8 , 9 월령. * 양방 Student's t 테스트 p〈0.01,unpaired. E,마우스 IgG (적색) 및 α-시누클레인(녹색)에 대한 항체로 이중 표지. 화살표는 각 패널에서 두 마커들의 동시존재를 나타냄. F-K에서 모든 이미지들은 주사와 동측 부위에서 해마로부터 얻음. F, α-시누클레인 (적색) 및 Iba-1 (녹색)에 대한 항체로 이중 표지된 미세아교세포의 예. G, 주사 후 1, 7, 14, 및 28 일 후 α-시누클레인을 가지는 Iba-1-포지티브 세포의 퍼센트. H, α-시누클레인 (적색) 및 뉴런 마커(NeuN, 녹색)에 대한 항체로 이중 표지. 화살표는 상부에서는 동시 존재를 나타낸 반면, 하부에서 화살표는 NeuN과 동시존재하지 않는 방추형 α-시누클레인 포지티브 세포를 나타냄. I, α-시누클레인과 동시존재하는 포지티브 세포의 비율 측정용 이중 표지를 위한 이미지 분석.그룹 당 n=8 , 9 월령. J, α-시누클레인 (적색) 및 GFAP(녹색)에 대한 항체로 이중 표지. 화살표 머리는 GFAP와 동시존재하지 않는 α-시누클레인-포지티브 뉴런을 나타냄;화살표는 GFAP와 동시존재하는 α-시누클레인-포지티브 세포를 나타냄. K, α-시누클레인에 대한 항체와 동시존재하는 GFAP-포지티브 세포의 비율 측정용 이중표지를 위한 이미지 분석 (그룹 당 n=8 , 9 월령.; * 일방 ANOVA,post hoc Tukey-Kramer에 의한 p〈0.05). 스케일 바: H, J, 25μm.FIG. 8 shows increased localization of α-synuclein and IgG in microglia cells in vivo after one injection of antibody. PDGF-α-synuclein tg mice (line M) were given for injection in the hippocampus as shown in FIG. 7. Images in the AE are obtained from the injection, ipsilateral region and dentate gyrus. Scale bar, 10 μm. A , double labeling with antibodies to α-synuclein (red) and microglia marker (Iba1, green). B , Image analysis for double label for measurement of proportion of Iba-1-positive cells co-located with α-synuclein. N = 8 per group, 9 months of age. * Both Student's t test p <0.01, unpaired. Double labeling with antibodies against C , mouse IgG (red) and Iba-1 (green). D , Image analysis for double label for measurement of the proportion of Iba-1-positive cells co-located with mouse IgG. N = 8 per group, 9 months of age. * Both Student's t test p <0.01, unpaired. E , double label with antibody to mouse IgG (red) and α-synuclein (green). Arrows indicate the coexistence of two markers in each panel. All images in FK are obtained from the hippocampus at the injection and ipsilateral site. Example of microglia double labeled with antibodies against F , α-synuclein (red) and Iba-1 (green). G , percent of Iba-1-positive cells with α-synuclein after 1, 7, 14, and 28 days after injection. Double label with antibody to H , α-synuclein (red) and neuronal marker (NeuN, green). Arrows indicate coexistence at the top, while arrows at the bottom indicate spindle-like α-synuclein positive cells that do not coexist with NeuN. I , Image analysis for double labeling for proportional measurement of positive cells co-existing with α-synuclein. N = 8 per group, 9 months of age. J , double label with antibody against α-synuclein (red) and GFAP (green). Arrowheads indicate α-synuclein-positive neurons that do not coexist with GFAP; arrows indicate α-synuclein-positive cells coexist with GFAP. K , Image analysis for double labeling for the ratio of GFAP-positive cells co-existing with antibodies to α-synuclein (n = 8, 9 months of age per group; * One-way ANOVA, post hoc Tukey-Kramer <0.05). Scale bar: H , J , 25 μm.
도 9는 274 항체로 수동 면역화한 후 α-시누클레인 tg 마우스에서 행동 개선 및 감소된 α-시누클레인 축적을 나타냄. A, pole 테스트를 내려오는데 여행한 전체 시간. B,Open field 전체 자발적인 활성(광전지의 크로스 수). C, Open field 자발적인 활성 테스팅 5분 후 여행한 전체 거리.D, Open field 자발적인 활성 테스팅 5분 후 왕복(rearings)의 전체 수.A-D, *non-tg 및 α-시누클레인 tg와 비교시 post hoc Dunnet's를 가지는 일방 ANOVA에 의한 p〈0.05 ; # α-시누클레인 tg에서 IgG 및 274 항체와 비교시 post hoc를 가지는 일방 ANOVA에 의한 p〈0.05 . E, 피질 및 해마에서 α-시누클레인 면역반응성의 저배율 뷰(x20). 스퀘어는 F 및 G에서 확대된 부위를 나타냄.스케일 바, 250μm. F, G, 고 배율(x630) 뷰 (F, 성상세포; G, 뉴런). 스케일 바, 20μm. H-ㅓ, 각각 신피질, 해마, 선조체에서 α-시누클레인을 나타내는 아교세포 및 뉴런 세포의 추정 수(해부 방법)의 이미지 분석.* Student's t test에 의한 p〈0.001. K, 해마에서 α-시누클레인(적색) 및 뉴런 마커(NeuN, 녹색)에 대한 항체로 이중 표지. 화살표 머리는 동시존재를 나타냄.L, α-시누클레인과 동시존재하는 NeuN-포지티브 세포의 비율을 평가하기 위한 이중 표지를 위한 이미지 분석.M, 해마에서 α-시누클레인(적색) 및 GFAP (녹색)에 대한 항체로 이중 표지. 화살표 머리는 성상아교와 동시존재하는 α-시누클레인-포지티브 세포를 나타냄. N, α-시누클레인에 대한 항체와 동시존재하는 GFAP포지티브 세포의 비율의 평가를 위한 이중 표지를 위한 이미지 분석. O, 해마에서 α-시누클레인(적색) 및 Iba-1 (녹색)에 대한 항체로 이중 표지. 화살표 머리는 미세아교와 동시존재하는 α-시누클레인-포지티브 세포를 나타냄. P, α-시누클레인에 대한 항체와 동시존재하는 Iba-1-포지티브 세포의 비율의 평가를 위한 이중 표지를 위한 이미지 분석.Q, 해마에서 α-시누클레인(적색) 및 cathepsin D (녹색)에 대한 항체로 이중 표지. 화살표 머리는 동시존재를 나타냄. R, α-시누클레인에 대한 항체와 동시존재하는 cathepsin-D-포지티브 세포의 비율의 평가를 위한 이중 표지를 위한 이미지 분석. 스케일 바, 10 μm. * 양방 Student's t 테스트에 의한 p〈 0.01 unpaired, .n=8-에서 10-월령 마우스.9 shows improved behavior and reduced α-synuclein accumulation in α-synuclein tg mice after passive immunization with 274 antibody. A , the total time traveled down the pole test. B , open field total spontaneous activity (number of crosses of photocell). C , Open field Total distance traveled 5 minutes after voluntary active testing. D , Open field Total number of roundings after 5 minutes of spontaneous active testing. P <0.05 by unilateral ANOVA with post hoc Dunnet's compared to AD , * non-tg and α-synuclein tg; # p <0.05 by unilateral ANOVA with post hoc compared to IgG and 274 antibodies in α-synuclein tg. E , low magnification view (x20) of α-synuclein immunoreactivity in cortex and hippocampus. Squares show magnified sites in F and G. Scale bar, 250 μm. F , G , high magnification (x630) views ( F , astrocytes; G , neurons). Scale bar, 20 μm. H- sh, respectively neocortex, hippocampus, image analysis of glial and neuronal cells can be estimated (dissection method) representing the α- synuclein in the striatum. * Student's t by test p <0.001. K , double labeled with antibody against α-synuclein (red) and neuronal marker (NeuN, green) in hippocampus. Arrow heads indicate coexistence. L , Image analysis for double labeling to assess the proportion of NeuN-positive cells co-existing with α-synuclein. M , double labeled with antibodies against α-synuclein (red) and GFAP (green) in the hippocampus. Arrowheads indicate α-synuclein-positive cells coexisting with astrocytes. N , Image analysis for double labeling for evaluation of the proportion of GFAP positive cells co-existing with antibodies to α-synuclein. O , double labeled with antibodies against α-synuclein (red) and Iba-1 (green) in hippocampus. Arrowheads indicate α-synuclein-positive cells coexist with microglia. P , Image analysis for double labeling for evaluation of the proportion of Iba-1-positive cells co-existing with antibodies to α-synuclein. Q , double labeled with antibodies against α-synuclein (red) and cathepsin D (green) in the hippocampus. Arrow heads indicate coexistence. R , Image analysis for double labeling for evaluation of the proportion of cathepsin-D-positive cells co-existing with antibodies to α-synuclein. Scale bar, 10 μιη. * 10-month-old mice at p <0.01 unpaired, .n = 8- by both Student's t tests.
도 10은 274 항체로 수동 면역화 후 감소된 신경퇴행성 및 전 염증성 사이토카인을 나타냄. A, IgG 또는 274 항체로 처리된 α-시누클레인 tg 및 non-tg 마우스에서 NeuN에 대한 항체로 면역염색된 절편의 저배율 뷰(x20). IgG로 처리된 α-시누클레인 tg 마우스는 해마에서 뉴런 손실을 나타냄(화살표). 스케일 바, 250μm. B, 해마에서 NeuN의 입체학 분석(해부자 방법). C, 각각 IgG 또는 274 항체로 처리된 α-시누클레인 tg 및 non-tg 마우스에서 synaptophysin에 대한 항체로 면역염색된 절편의 고배율 뷰(x630), 스케일 바, 20μm. D, 해마에서 synaptophysin의 입체학 분석(해부자 방법).E, IgG 또는 274 항체로 처리된 α-시누클레인 tg 및 non-tg 마우스에서 GFAP에 대한 항체로 면역염색된 절편의 저배율 뷰(x20). IgG로 처리된 α-시누클레인 tg 마우스는 신피질(inset) 및 해마에서 증가된 성상교세포증을 나타냄. 스케일 바, 250μm. F, 흡광도에서 발현된 GFAP 면역반응서의 이미지 분석. 그 측정은 전체 해마에서 수행됨.G, 각각 IgG 또는 274 항체로 처리된 α-시누클레인 tg 및 non-tg 마우스에서 Iba-1에 대한 항체로 면역염색된 해마 절편의 고배율 뷰(x630), 스케일 바, 20μm. H, 해마에서 미세아교세포의 평가된 수의 이미지 분석. *non-tg 및 α-시누클레인 tg와 비교시 post hoc Dunnet s를 가지는 일방 ANOVA에 의한 p〈0.05 ; #α-시누클레인 tg에서 IgG 및 274 항체와 비교 시 post hoc Fisher를 가지는 일방 ANOVA에 의한 p〈0.05 . I-L, 절편을 TNFα 및 IL-6에 대한 항체로 면역표지하고 해마에서 미세농도(microdensitometric) 분석을 수행하였다. non-tg 대조군과 비교하여, IgG로 처리된 α-시누클레인 tg 마우스에서, TNFα(I, J) 및 IL-6 (K, L)의 레벨은 증가하였지만, 274 항체로 처리는 α-시누클레인 tg 마우스에서 이들 사이코카인의 레벨을 감소시킴. * 처리 그룹을 비교할 때 Fisher 그리고 non-tg 및 α-시누클레인 tg을 비교할 때 post hoc Dunnet's인 일방 ANOVA에 의한 p〈0.05.10 shows reduced neurodegenerative and pre-inflammatory cytokines after passive immunization with 274 antibodies. Low magnification view (x20) of sections immunostained with antibodies against NeuN in α-synuclein tg and non-tg mice treated with A , IgG or 274 antibodies. Α-synuclein tg mice treated with IgG show neuronal loss in the hippocampus (arrow). Scale bar, 250 μm. B , Stereoscopic Analysis of NeuN in Hippocampus (Anatomy Method). C , high magnification views (x630), scale bars, 20 μm of sections immunostained with antibodies to synaptophysin in α-synuclein tg and non-tg mice treated with IgG or 274 antibodies, respectively. D , stereoscopic analysis of synaptophysin in the hippocampus (anatomist method). Low magnification views (x20) of sections immunostained with antibodies against GFAP in α-synuclein tg and non-tg mice treated with E , IgG or 274 antibodies. Α-synuclein tg mice treated with IgG show increased astrocytes in the neocortex and hippocampus. Scale bar, 250 μm. F , Image analysis of GFAP immune response expressed at absorbance. The measurement is performed on the entire hippocampus. G , high magnification view (x630), scale bar, 20 μm of hippocampal sections immunostained with antibodies against Iba-1 in α-synuclein tg and non-tg mice treated with IgG or 274 antibodies, respectively. H , Image analysis of the estimated number of microglia in the hippocampus. * p <0.05 by unilateral ANOVA with post hoc Dunnet s compared to non-tg and α-synuclein tg; p <0.05 by unilateral ANOVA with post hoc Fisher compared to IgG and 274 antibodies at # α-synuclein tg. IL , sections were immunolabeled with antibodies against TNFα and IL-6 and microdensitometric analysis was performed in the hippocampus. In α-synuclein tg mice treated with IgG compared to non-tg controls, the levels of TNFα ( I , J ) and IL-6 ( K , L ) were increased, but treatment with 274 antibody was not affected by α-synuclein. reduced levels of these psychocaine in tg mice. * P <0.05 by unilateral ANOVA, Fisher and post hoc Dunnet's when comparing treatment groups and comparing non-tg and α-synuclein tg.
이하 비한정적인 실시예를 통하여 본 발명을 더욱 상세하게 설명한다.단 하기 실시예는 본 발명을 예시하기 위한 의도로 기재된 것으로서 본 발명의 범위는 하기 실시예에 의하여 제한되는 것으로 해석되지 아니한다.The present invention is described in more detail with reference to the following non-limiting examples. However, the following examples are intended to illustrate the present invention and the scope of the present invention is not to be construed as being limited by the following examples.
본 발명에 사용된 항체는 : α-시누클레인 단클론 항체(BD Biosciences, #610787), α-시누클레인 다클론 항체 (Cell Signaling Technology, #2642), myc 다클론 항체 (Abcam, #ab9106), CD32 다클론 항체 (United States Biological, #c2384-0B), 및 CD16/CD32 단클론 항체(Abcam,#ab25235), GM130 단클론 항체(BD Biosciences, #G65120),cathepsin D 단클론 항체(Abcam, #ab6313), 및 caveolin-1 단클론 항체(BD Biosciences, #C13620). FITC-표지된 콜레라 톡신 B 소단위체(CTB)는 시그마로부터 구입. Bodipy-표지된 GM1,Bodipy-FL LDL, 및 Alexa Fluor 568-부착된 Dextran은 Invitrogen로부터 구입. UltraLink 고정화 단백질 A/G, Gentle Ag/Ab 결합 버퍼 pH 8.0, 및 IgG 정제용 용출 버퍼는 Piercef부터 구입.Antibodies used in the present invention include: α-synuclein monoclonal antibody (BD Biosciences, # 610787), α-synuclein polyclonal antibody (Cell Signaling Technology, # 2642), myc polyclonal antibody (Abcam, # ab9106), CD32 Polyclonal antibodies (United States Biological, # c2384-0B), and CD16 / CD32 monoclonal antibodies (Abcam, # ab25235), GM130 monoclonal antibodies (BD Biosciences, # G65120), cathepsin D monoclonal antibodies (Abcam, # ab6313), and caveolin-1 monoclonal antibody (BD Biosciences, # C13620). FITC-labeled cholera toxin B subunit (CTB) was purchased from Sigma. Bodipy-labeled GM1, Bodipy-FL LDL, and Alexa Fluor 568-attached Dextran were purchased from Invitrogen. UltraLink immobilized protein A / G, Gentle Ag / Ab binding buffer pH 8.0, and IgG prep elution buffer from Piercef.
실시예 1 항체 생산Example 1 Antibody Production
α-시누클레인에 대한 단클론 항체의 생산 및 특성은 Lee et al., 2011b에 기재. α-시누클레인에 대한 모든 항체는 IgG2a isotype의 단클론 항체이다. 62 및 274 항체의 에피토프는 α-시누클레인의 C말단 끝에 존재하지만 (120-140), 169 및 171 항체의 에피토프는 C-말단 부위(120-140) 및 중간 부위(61-95) 모두를 필요로한다. 종 반응성 테스트는 169, 274, 및 171 항체는 인간 α-시누클레인에 대해서만 반응하지만, 62 항체는 인간 및 마우스 α-시누클레인 모두에 반응성이 있다는 것을 나타낸다. 본 발명에서 사용된 항체들은 다른 형태를 구분하지 못한다. 특히 274 항체는 α-시누클레인의 세포외 및 원형질내 형태 모두와 모노머 및 응집체 형태 모두에 면역반응성을 나타낸다. 대조군 IgG, 마우스 IgG isotypes의 혼합물은 프로틴 A/G 컬럼을 사용하여 모아진 정상 마우스 혈청으로부터 제조하였다.The production and properties of monoclonal antibodies against α-synuclein are described in Lee et al., 2011b. All antibodies against α-synuclein are monoclonal antibodies of the IgG2a isotype. Epitopes of 62 and 274 antibodies are present at the C-terminus of α-synuclein (120-140), but epitopes of 169 and 171 antibodies require both C-terminal regions (120-140) and intermediate regions (61-95) Shall. Species reactivity tests indicate that 169, 274, and 171 antibodies respond only to human α-synuclein, while 62 antibodies are reactive to both human and mouse α-synuclein. The antibodies used in the present invention do not distinguish between different forms. In particular 274 antibodies show immunoreactivity to both the extracellular and intraplasma forms of α-synuclein and to the monomer and aggregate forms. Mixtures of control IgG, mouse IgG isotypes were prepared from normal mouse serum collected using protein A / G columns.
실시예 2: α-시누클레인의 정제 및 피브릴 및 올리고머의 생성 Example 2: Purification of α-synuclein and production of fibrils and oligomers
야생형 인간 α-시누클레인은 Lee HJ, Bae EJ, Jang A, Ho DH, Cho ED, Suk JE, Yun YM, Lee SJ (2011b)J Neurosci Methods 199:249-257의 기재와 같이 정제하였다. 피브릴화를 위하여, α-시누클레인 (3 mg/ml in PBS)를 250 rpm에서 계속 교반하면서 37℃에서 2주간 배양하였다. 짧은 초음파처리 후, α-시누클레인을 1주일 더 배양하였다. 배양 후 그 단백질을 100,000xg에서 1시간 원심분리하고 그 펠렛을 PBS에 재부유하였다. 일부 실험에서, 초음파처리에 의하여 제조된 작은 피브릴 절편을 사용하였다. α-시누클레인 올리고머는 Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, Kretzschmar H, Hengerer B, Kostka M (2007) J Neurosci 27:9220-9232에 기재된 방법으로 제조하였다. 동결건조된 α-시누클레인을 20% ethanol를 가지는 50mM sodium phosphate,pH7.0에 녹여 최종 농도 0.1 mg/ml로 하고 상온에서 4시간 동안 250 rpm으로 교반하였다. α-시누클레인을 다시 동결건조하고 1/2 시작 부피의 10% ethanol을 가지는 50mM sodium phosphate, pH 7.0에 재부유하였다. 그 단백질을 잔류 에탄올의 증발을 위하여 뚜껑을 열어 놓은채로 상온에서 24시간 동안 배양하였다.Wild-type human α-synuclein was purified as described by Lee HJ, Bae EJ, Jang A, Ho DH, Cho ED, Suk JE, Yun YM, Lee SJ (2011b) J Neurosci Methods 199: 249-257. For fibrillation, α-synuclein (3 mg / ml in PBS) was incubated at 37 ° C. for 2 weeks with continued stirring at 250 rpm. After short sonication, α-synuclein was incubated for another week. After incubation, the protein was centrifuged at 100,000 × g for 1 hour and the pellet was resuspended in PBS. In some experiments, small fibril sections prepared by sonication were used. α-synuclein oligomers were prepared by the methods described in Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M, Giese A, Kretzschmar H, Hengerer B, Kostka M (2007) J Neurosci 27: 9220-9232. Lyophilized α-synuclein was dissolved in 50 mM sodium phosphate, pH 7.0 with 20% ethanol to a final concentration of 0.1 mg / ml and stirred at 250 rpm for 4 hours at room temperature. α-synuclein was lyophilized again and resuspended in 50 mM sodium phosphate, pH 7.0 with 10% ethanol of 1/2 starting volume. The protein was incubated at room temperature for 24 hours with the lid open for evaporation of residual ethanol.
실시예 3: 세포 배양Example 3: Cell Culture
SH-SY5Y 세포를 Lee HJ, Khoshaghideh F, Patel S, Lee SJ (2004) J Neurosci 24:1888-1896에 기재된 것과 같이 유지하고 분화하였다. BV-2 미세아교 세포주를 5% fetal bovine serum (FBS) 및 penicillin 및 streptomycin를 가지는 DMEM에서 유지하였다. 하이브리도마 세포를 10% FBS 및 100 μM hypoxanthine 보충(Invitrogen)을 가지는 DMEM에서 배양하였다. 랫트 1차 아교세포 배양은 Lee HJ, Suk JE, Bae EJ, Lee SJ (2008) Biochem Biophys Res Commun 372:423-428에 기재된 것과 같이 준비하였다.SH-SY5Y cells were maintained and differentiated as described in Lee HJ, Khoshaghideh F, Patel S, Lee SJ (2004) J Neurosci 24: 1888-1896. BV-2 microglia cell lines were maintained in DMEM with 5% fetal bovine serum (FBS) and penicillin and streptomycin. Hybridoma cells were cultured in DMEM with 10% FBS and 100 μM hypoxanthine supplement (Invitrogen). Rat primary glial cultures were prepared as described in Lee HJ, Suk JE, Bae EJ, Lee SJ (2008) Biochem Biophys Res Commun 372: 423-428.
1차 피질 뉴런은 Lee HJ, Patel S, Lee SJ (2005) J Neurosci 25:6016-6024에 기재된 방법에 따라서 Sprague Dawley 랫트의 15-16(E15-16)일째 배아로부터 얻었다.Primary cortical neurons were obtained from embryos on day 15-16 (E15-16) of Sprague Dawley rats according to the method described in Lee HJ, Patel S, Lee SJ (2005) J Neurosci 25: 6016-6024.
실시예 4: 조건화된 배지의 제조Example 4: Preparation of Conditioned Media
분화된 SH-SY5Y 세포를 adeno/α-syn로 주사하였다. 주사 2일 후, 세포를 DMEM로 3회 세척하고 혈청 없는 DMEM에서 배양하였다. 37℃에서 18시간 배양 후, 조건화된 배지를 모아서 4℃, 250xg에서 10 분간 원심분리한 후, 세포 찌꺼기를 제거하기 위하여 그 상등액을 4℃, 10,000 xg에서 10 분간 원심분리하였다.그 상등액을 Amicon 10K MWCO 필터(Millipore)를 사용하여 농축하였다.Differentiated SH-SY5Y cells were injected with adeno / α-syn. Two days after injection, cells were washed three times with DMEM and incubated in serum-free DMEM. After 18 hours of incubation at 37 ° C, the conditioned medium was collected and 4 ° C, 250xgin After centrifugation for 10 minutes, the supernatant was removed at 10,000 x 4 ° C to remove cellular debris.gin Centrifuge for 10 minutes. The supernatant was concentrated using an Amicon 10K MWCO filter (Millipore).
실시예 5: 세포에서 α-시누클레인 응집체 및 모노머의 업테이크 및 분해 Example 5: Uptake and Degradation of α-Synuclein Aggregates and Monomers in Cells
BV-2, 미세아교 및 성상세포를 실험 전날 배양 접시로 나누었다. 냉장 PBS로 세포를 2회 세척 후, 상온에서 5분간 5 μg/ml 정상 마우스 IgG 또는 α-시누클레인항체로 미리 배양된 0.2 μM α-시누클레인 피브릴 또는 올리고머를 배양 배지와 세포에 첨가하였다. 모노머를 1 μM로 첨가하였다. 조건화된 배지(5x 농축)를 5분 동안 항체로 미리배양하였다. 그 후 세포를 37℃에서 배양하고 기재된 시간에서 수집하였다. 분해 속도를 측정하기 위하여, 상기와 같이 응집체를 흡수하고 세포를 냉장 PBS로 2회 세척하였다. 신선한 배양 배지를 첨가하고, 기재된 시간 동안 37℃에서 세포의 배양을 수행하였다.BV-2, microglia and astrocytes were divided into petri dishes the day before the experiment. After washing the cells twice with cold PBS, 0.2 μM α-synuclein fibrils or oligomers previously incubated with 5 μg / ml normal mouse IgG or α-synuclein antibodies at room temperature were added to the culture medium and cells. Monomer was added at 1 μM. Conditioned medium (5 × concentrated) was preincubated with antibody for 5 minutes. Cells were then incubated at 37 ° C. and collected at the time indicated. To determine the rate of degradation, aggregates were taken up as above and cells were washed twice with cold PBS. Fresh culture medium was added and culture of cells was performed at 37 ° C. for the time stated.
실시예 6:α-시누클레인 항체의 흡수Example 6 Uptake of α-Synuclein Antibodies
5μg/ml의 274 항체 또는 대조군 IgG으로 처리된 BV-2 세포를 여러 양의 α-시누클레인 피브릴(0, 2.66, 13.3, 66.5, 133, 및 330 nM)으로 미리배양하였다. 274 항체의 비특이적인 흡착의 가능성을 배제하기 위하여, 여러 양의 α-시누클레인 모노머(0, 66.5, 및 330 nM)로 미리배양된 5 μg/ml의 274항체를 세포에 첨가하였다. 흡수된 항체의 레벨을 항-마우스 IgG 항체로 검출하였다.BV-2 cells treated with 5 μg / ml of 274 antibody or control IgG were preincubated with various amounts of α-synuclein fibrils (0, 2.66, 13.3, 66.5, 133, and 330 nM). To rule out the possibility of nonspecific adsorption of the 274 antibody, 5 μg / ml of 274 antibody preincubated with various amounts of α-synuclein monomer (0, 66.5, and 330 nM) was added to the cells. Levels of absorbed antibodies were detected with anti-mouse IgG antibodies.
실시예 7: 세포 추출물의 제조Example 7: Preparation of Cell Extracts
세포를 PBS로 세척하고 추출 버퍼[PBS, 1% Triton X-100, 1% (v/v) 프로테이즈 저해제 혼합물(Sigma)]에서 파쇄하였다. 10분간 얼음에서 배양한 후, 세포 파쇄액을 10분간 16,000 xg에서 원심분리하였다. Triton X-100 불용성 펠렛을 Laemmli 샘플 버퍼에서 재부유하고 잠시 초음파처리하였다.Cells were washed with PBS and disrupted in extraction buffer [PBS, 1% Triton X-100, 1% (v / v) Protease Inhibitor Mixture (Sigma)]. After incubation on ice for 10 minutes, the cell lysate was centrifuged at 16,000 x g for 10 minutes. Triton X-100 insoluble pellets were resuspended in Laemmli sample buffer and sonicated briefly.
실시예 8: 웨스턴 블럿팅Example 8: Western Blotting
웨스턴 블럿팅을 Lee HJ, Lee SJ (2002)J Biol Chem 277:48976-48983에 기재된 것과 같이 수행하였다. 이미지를 얻고 Fujifilm Luminescent Image Analyzer LAS-3000 및 Multi Gauge (v3.0) 소프트웨어(Fujifilm)를 사용하여 정량화하였다.Western blotting was performed as described in Lee HJ, Lee SJ (2002) J Biol Chem 277: 48976-48983. Images were obtained and quantified using Fujifilm Luminescent Image Analyzer LAS-3000 and Multi Gauge (v3.0) software (Fujifilm).
정량화를 위하여, 런닝 젤의 시작으로부터 α-시누클레인 모노머 밴드까지 모든 레인을 분석하였다.For quantification, all lanes were analyzed from the start of the running gel to the α-synuclein monomer band.
실시예 9: 배양된 세포의 면역형광 염색Example 9: Immunofluorescence Staining of Cultured Cells
세포 염색에 대한 과정을 Lee HJ, Lee SJ (2002)J Biol Chem 277:48976-48983에 기재된 것과 같이 수행하였다.The procedure for cell staining was performed as described in Lee HJ, Lee SJ (2002) J Biol Chem 277: 48976-48983.
요약하면, poly-L-lysine-코팅된 커버슬립 상에 배양된 세포를 PBS 내의 4% paraformaldehyde에서 고정화하고 0.1% Triton X-100 내에서 흡수하였다. 블럭킹 용액에서 희석된 1차 항체의 첨가 전에 세포를 블럭킹 용액(5% bovine serum albumin/3% goat serum in PBS)에서 배양하였다. 세척 후, 세포를 형광 염료 부착된 2차 항체로 배양하였다. 핵을 TOPRO-3 iodide (Invitrogen)으로 염색하고, 커버슬립을 antifade reagent (Invitrogen)를 사용하여 슬라이드 글래스 상에 마운트하였다. Olympus FV1000 공초점 레이져 스캐닝 현미경을 세포의 관찰에 사용하였다.In summary, cells cultured on poly-L-lysine-coated coverslips were immobilized in 4% paraformaldehyde in PBS and taken up in 0.1% Triton X-100. Cells were incubated in blocking solution (5% bovine serum albumin / 3% goat serum in PBS) prior to addition of the primary antibody diluted in blocking solution. After washing, the cells were incubated with fluorescent dye attached secondary antibody. Nuclei were stained with TOPRO-3 iodide (Invitrogen) and coverslips mounted on slide glass using antifade reagent (Invitrogen). Olympus FV1000 confocal laser scanning microscope was used for the observation of the cells.
실시예 10: Tg 마우스 주 및 항체의 뇌내(intracerebral) 주사Example 10 Intracerebral Injection of Tg Mouse Main and Antibodies
본 발명을 위하여 platelet-derived growth factor-베타 (PDGF-베타) 프로모터로부터 유래한 α-시누클레인을 과발현하는 마우스(line M)를 사용하였다 (Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A,Sagara Y, Sisk A, Mucke L (2000) Science 287:1265-1269). 이 모델은 이들 라인으로부터 유래한 마우스는 루이소체 질환에서 기재된 것과 유사한 신피질, 해마, 및 선조체를 통하여 분포된 신경 및 아교세포 모두에서 α-시누클레인 응집체를 발생하기 때문에 선택하였다. 이들 마우스에서,α-시누클레인은 신피질 및 해마의 깊은 층에서 신경망 및 뉴런 세포체에 축적한다.α-시누클레인은 3개월 령에서 뉴런에 축적되기 시작하여 12개월 령에서 최대 축적을 가진다. 따라서 이 tg 마우스 모델에서, α-시누클레인 병리는 매우 일정하고 시공적인(spatiotemporal) 순서로 진행한다. 모든 동물 실험 과정은 실험 동물윤리 위원회의 승인을 받았다. 모든 실험은 암컷 마우스에서 수행되었다. 각 실험을 위하여 그룹 당 8마리 마우스를 사용하였다. 전체 32 마리 마우스를 이 실험에 사용하였다;이중 16 마리 마우스는 α-시누클레인 tg 마우스이고 다른 16 마리는 non-tg 동물 대조군(9 월령). 각 그룹으로부터 마우스를 3μl의 비면역 IgG 대조군(n=8 non-tg 및 n=8 α-시누클레인 tg) 또는 α-시누클레인 (clone 274; 1 mg/ml)에 대한 항체(n = 8 non-tg 및 n=8 α-시누클레인 tg)로 해마(5 μl Hamilton 주사기를 사용, 0.25 μl/분)에 주사하였다. 요약하면, 마취하에서 Koft 정위 장치 및 코디네이트(해마:AP: 2.0 mm, 외측 1.5 mm, 깊이 1.3 mm) 상에 위치한 마우스를 Franklin 및 Paxinos (Franklin KBJ, Paxinos G, (2008) The mouse brain in stereotaxic coordinates,Ed 3. Burlington, MA: Elsevier) 도해서에 의해서 결정하였다. 용액의 주사를 위하여 수압 시스템에 연결된 Hamilton 주사기를 항체 전달에 사용하였다. 뇌 조직으로 용액의 확산을 위하여, 바늘을 주사 완료 후 5분간 유지하였다.For the present invention, mouse (line M) overexpressing α-synuclein derived from the platelet-derived growth factor-beta (PDGF-beta) promoter was used (Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M Takeda A, Sagara Y, Sisk A, Mucke L (2000) Science 287: 1265-1269). This model was chosen because mice derived from these lines generate α-synuclein aggregates in both neuronal and glial cells distributed throughout the renal cortex, hippocampus, and striatum, similar to those described in Lewy body disease. In these mice, α-synuclein accumulates in neural networks and neuronal cell bodies in the deep layers of the neocortex and hippocampus. Α-synuclein begins to accumulate in neurons at 3 months of age and has maximum accumulation at 12 months of age. Thus, in this tg mouse model, α-synuclein pathology proceeds in a very constant and spatiotemporal order. All animal testing procedures were approved by the Experimental Animal Ethics Committee. All experiments were performed in female mice. Eight mice per group were used for each experiment. A total of 32 mice were used for this experiment; 16 mice were α-synuclein tg mice and the other 16 non-tg animal controls (9 months of age). Mice from each group were treated with 3 μl of non-immune IgG controls ( n = 8 non-tg and n = 8 α-synuclein tg) or antibodies against α-synuclein (clone 274; 1 mg / ml) ( n = 8 non -tg and n = 8 α-synuclein tg) were injected into hippocampus (0.25 μl / min using a 5 μl Hamilton syringe). In summary, mice placed on Koft stereotactic devices and coordinates (hippocampus: AP: 2.0 mm, outer 1.5 mm, depth 1.3 mm) under anesthesia were placed in Franklin and Paxinos (Franklin KBJ, Paxinos G, (2008) The mouse brain in stereotaxic coordinates , Ed 3. Burlington, MA: Elsevier). Hamilton syringes connected to a hydraulic system were used for antibody delivery for injection of the solution. For diffusion of the solution into brain tissue, the needles were held for 5 minutes after completion of injection.
대측면에 대하여 비교를 위하여 마우스를 일방 주사(우측)를 하였다. 마우스를 항체 주사 후 4 주간 생존시켰다. 12 마리 α-시누클레인 tg 마우스 (3 월령) 추가 그룹은 α-시누클레인 (clone 274; 1 mg/ml)에 대한 항체로 해마에 일방주사를 받았고 주사 후 1, 7, 14, 및 28일에 분석을 위하여 희생시켰다. 동물의 인간적인 처리를 위한 국립보건원 가이드라인에 따라서 마우스를 chloral hydrate로 마취하고 0.9% 식염수로 심장으로 관류하였다. 뇌를 제거하고 신경병리 분석을 위하여 인산-버퍼 4% paraformaldehyde,pH7.4에서 4℃에서 48시간 고정하였다.Mice were unilaterally injected (right) for comparison with the contralateral side. Mice were survived 4 weeks after antibody injection. An additional group of twelve α-synuclein tg mice (3 months old) were unilaterally injected into the hippocampus with antibodies against α-synuclein (clone 274; 1 mg / ml) and at 1, 7, 14, and 28 days after injection Sacrificed for analysis. Mice were anesthetized with chloral hydrate and perfused to the heart with 0.9% saline according to the National Institutes of Health guidelines for human treatment of animals. The brain was removed and fixed for 48 hours at 4 ℃ in phosphate-buffer 4% paraformaldehyde, pH 7.4 for neuropathology analysis.
실시예 11: 수동 면역화 및 행동 연구Example 11: Passive Immunization and Behavioral Studies
전체 32 마리 마우스를 본 실험에 사용하였고, 이중 16 마리는 α-시누클레인 tg 마우스 (line M, 10 월령)이고 나머지 16 마리는 non-tg 동물 대조군 (10 월령)이다. 각 그룹으로부터 마우스에 비면역 IgG 대조군(n=8 non-tg 및 n=8 α-시누클레인 tg) 또는α-시누클레인 (clone 274; 1 mg/ml)에 대한 마우스 단클론 항체(n=8 non-tg 및 n= 8 α-시누클레인 tg)를 복강내로 1 mg/ml의 농도(100 μl/주, 4 동안)로 주었다. 그 첫번째 주사 후 1달 동안 마우스를 생존시키고 Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A,Sagara Y, Sisk A, Mucke L (2000) Science 287:1265-1269에 기재된 것과 같은 폴 테스트에서 모터 기능에 대한 테스트 및 오픈 필드에서 자발적인 활성에 대한 테스트를 수행하였다. 행동 테스트를 수행하여 마우스를 신경 병리 분석을 수행하였다.A total of 32 mice were used in this experiment, of which 16 were α-synuclein tg mice (line M, 10 months old) and the remaining 16 were non-tg animal controls (10 months old). Mouse monoclonal antibodies ( n = 8 non) against non-immune IgG controls ( n = 8 non-tg and n = 8 α-synuclein tg) or α-synuclein (clone 274; 1 mg / ml) in each group -tg and n = 8 α-synuclein tg) were given intraperitoneally at a concentration of 1 mg / ml (100 μl / week, for 4). Mice were survived for 1 month after the first injection and as described in Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L (2000) Science 287: 1265-1269. In the pole test a test for motor function and a spontaneous activity in the open field were performed. Behavioral tests were performed to perform mouse neuropathology analysis.
실시예 12: 신경병리 및 α-시누클레인 축적의 면역조직화학 분석Example 12 Immunohistochemical Analysis of Neuropathology and α-Synuclein Accumulation
α-시누클레인 축적의 분석은 IgG 또는 α-시누클레인 (clone 274)에 대한 항체로 처리된 non-tg 및 α-시누클레인 tg 마우스로부터 유래한 프리 플로팅, 일련의 절편에서 수행하였다. 절편들을 4℃에서 항-α-시누클레인 항체(1:500, affinity purified rabbit polyclonal;Millipore Bioscience Research Reagents)로 오버나잇 배양한 후, 바이오틴화된 염소 항-토기 IgG (1:100; Vector Laboratories), Avidin D-horseradish peroxidase (1:200, ABC Elite; Vector Laboratories)로 처리한 후, diaminobenzidine (DAB)로 검출하였다. 절편을 Olympus 명시야 디지털 현미경으로 이미지화하였다. 각 경우에 대해서, 세 절편을 Stereo-Investigator System (MBF Bioscience)을 사용하여 해부자 분석에 의하여 분석하였고 그 결과를 평균화하고 수/mm3로 표현하였다. 신경퇴행 및 신경 염증성 반응에 대한 항체의 효과를 분석하기 위하여, 절편들을 NeuN (Millipore), glial fibrillary acidic protein (GFAP) (Millipore), Iba1 (Wako), IL6 (Novus Biologicals),및 종양괴사인자(Abcam)에 대한 마우스 단클론 항체로 면역표지하고 DAB로 검출하였다. 항체 GFAP 및 Iba-1로 면역염색된 절편들을 Olympus 명시야 디지털 현미경으로 이미지화하고 Image Pro-Plus 시스템으로 흡광도 및 세포 카운트를 분석하였다. NeuN에 대한 항체로 면역염색된 절편을 Stereo-Investigator System (MBF Bioscience)을 사용한 해부자 분석에 의하여 분석하고 그 결과를 평균화하고 수/mm3로 표현하였다.Analysis of α-synuclein accumulation was performed on a series of sections, pre-floating, derived from non-tg and α-synuclein tg mice treated with antibodies to IgG or α-synuclein (clone 274). Sections were overnight incubated with anti-α-synuclein antibody (1: 500, affinity purified rabbit polyclonal; Millipore Bioscience Research Reagents) at 4 ° C., followed by biotinylated goat anti-earth IgG (1: 100; Vector Laboratories) After treatment with Avidin D-horseradish peroxidase (1: 200, ABC Elite; Vector Laboratories), it was detected with diaminobenzidine (DAB). Sections were imaged with an Olympus brightfield digital microscope. For each case, three sections were analyzed by dissection analysis using the Stereo-Investigator System (MBF Bioscience) and the results were averaged and expressed in number / mm 3 . To analyze the effect of antibodies on neurodegenerative and neuroinflammatory responses, sections were analyzed for NeuN (Millipore), glial fibrillary acidic protein (GFAP) (Millipore), Iba1 (Wako), IL6 (Novus Biologicals), and tumor necrosis factor Mouse monoclonal antibody against Abcam) and detected with DAB. Sections immunostained with antibody GFAP and Iba-1 were imaged with an Olympus brightfield digital microscope and absorbance and cell counts were analyzed with an Image Pro-Plus system. Sections immunostained with antibodies to NeuN were analyzed by dissection analysis using Stereo-Investigator System (MBF Bioscience) and the results were averaged and expressed in number / mm 3 .
실시예 13: 이중 면역조직화학 및 공초점 분석Example 13: Dual Immunohistochemistry and Confocal Analysis
동시위치화를 결정하기 위하여, 40-μm-두께 바이브라톰 절편을 α-시누클레인 (Millipore Bioscience Research Reagents, affinity purified polyclonal, 1:500), 마우스 IgG(274 항체 분포를 검출하기 위함), 및 Iba-1 (미세아교세포 마커)에 대한 항체의 조합으로 이중 표지하였다. α-시누클레인 면역반응성 구조는 Tyramide Signal Amplification-Direct (Red) 시스템(1:100; NEN Life Sciences)으로 검출한 반면 마우스 IgG 또는 Iba-1은 fluorescein isothiocyanate (FITC)-부착된 horse 항-마우스 항체(1:75; Vector Laboratories)로 검출하였다.To determine co-location, 40-μm-thick vibratomic fragments were prepared using α-synuclein (Millipore Bioscience Research Reagents, affinity purified polyclonal, 1: 500), mouse IgG (to detect 274 antibody distribution), and Double labeling with a combination of antibodies to Iba-1 (microglia marker). α-synuclein immunoreactive structures were detected by the Tyramide Signal Amplification-Direct (Red) system (1: 100; NEN Life Sciences) while mouse IgG or Iba-1 were fluorescein isothiocyanate (FITC) -attached horse anti-mouse antibodies (1:75; Vector Laboratories).
모든 절편들을 동일한 조건에서 동시에 진행하였고, 결과의 재생성을 평가하기 위하여 실험을 2회 수행하였다. 절편들을 MRC1024 LSCM 시스템(Bio-Rad)이 부착된 Axiovert 35 현미경(Zeiss) 상에서 Zeiss 63 x (NA 1.4) 대물로 이미지화하였다. 각 절편에 대해서, 10μm의 플레인에서 일련의 광학 절편을 얻어서 α-시누클레인-면역반응성 물질을 나타내는 Iba-1-포지티브 세포의 퍼센트의 분석을 위하여 사용하였다. 각 케이스로부터, 평균 120 Iba-1-포지티브 세포를 분석하였다(절편 당 30 개). 면역화 후 뉴런 또는 성상아교세포에 α-시누클레인의 동시존재를 결정하기 위한 추가적인 연구가 NeuN (뉴런 마커; Millipore, 마우스 단클론, 1:1000) 또는 GFAP (성상아교세포 마커; Millipore, 마우스 단클론, 1:2000) 및 α-시누클레인 (토끼 다클론; Millipore, 1:5000)에 대한 항체로 이중 표지된 절편에 의하여 수행되었다. α-시누클레인을 Tyramide Signal Amplification-Direct (Red) 시스템(1:100; NEN Life Sciences)으로 검출한 반면, NeuN 또는 GFAP는 FITC로 검출하였다.All sections were run simultaneously under the same conditions and two experiments were performed to evaluate the reproducibility of the results. Sections were imaged with a Zeiss 63 x (NA 1.4) objective on an Axiovert 35 microscope (Zeiss) attached with an MRC1024 LSCM system (Bio-Rad). For each section, a series of optical sections were obtained in a 10 μm plane and used for the analysis of the percentage of Iba-1-positive cells representing α-synuclein-immunoreactive material. From each case, an average of 120 Iba-1-positive cells was analyzed (30 per segment). Further studies to determine the co-existence of α-synuclein in neurons or astroglia after immunization include NeuN (neuron marker; Millipore, mouse monoclonal, 1: 1000) or GFAP (astroglia marker; Millipore, mouse monoclonal, 1). : 2000) and fragments double-labeled with antibodies to α-synuclein (rabbit polyclonal; Millipore, 1: 5000). α-synuclein was detected by the Tyramide Signal Amplification-Direct (Red) system (1: 100; NEN Life Sciences), while NeuN or GFAP was detected by FITC.
실시예 14: 통계 분석Example 14 Statistical Analysis
도면에 나타낸 값들은 평균±SEM으로 나타낸다. 편차의 통계적 유의성의 결정을 위한 P 값을 GraphPad InStat 버젼 3.05 소프트웨어를 사용한 paired 또는 unpaired, 양방 Student's t test, 일방 ANOVA with a post hoc Dunnet 또는 일방 ANOVA with Tukey 포스트 테스트를 통하여 계산하였다. 흡수 카이네틱 데이터의 통계적 분석을 위하여, 독립적인 실험들을 통하여 얻은 α-시누클레인의 베스트-fit 상대적인 값들을 Graph-Pad InStat 버젼 3.05 소프트웨어를 사용한 엑스트라 추가 제곱합 F 테스트에 의하여 분석하였다.Values shown in the figures are expressed as mean ± SEM. P value for determining the statistical significance of the deviation Paired or unpaired, both Student's using GraphPad InStat version 3.05 softwarettest, one-way ANOVA with apost hocCalculated via Dunnet or one-way ANOVA with Tukey post test. For statistical analysis of the absorption kinetics data, the best-fit relative values of α-synuclein obtained through independent experiments were analyzed by an extra addition sum sum F test using Graph-Pad InStat version 3.05 software.

Claims (6)

  1. 서열번호 1에 기재된 α-시누클레인 펩타이드에 특이적으로 결합하는 항체.An antibody that specifically binds to the α-synuclein peptide set forth in SEQ ID NO: 1.
  2. 제 1항의 항체를 유효성분으로 포함하는 파킨슨 질환 예방 또는 치료용 조성물.Parkinson's disease prevention or treatment composition comprising the antibody of claim 1 as an active ingredient.
  3. 제 1항의 항체를 유효성분으로 포함하는 루이소체 치매(DLB) 예방 또는 치료용 조성물.A composition for preventing or treating Lewy Body Dementia (DLB) comprising the antibody of claim 1 as an active ingredient.
  4. 후보 물질과 세포외 α-시누클레인 단백질을 이 두 성분이 상호작용하기에 적합한 환경 하에서 미세아교세포에 접촉시키고, 상기 미세아교세포에서 상기 α-시누클레인 단백질의 업테이크 및 분해 속도가 증가한 경우에 해당 후보물질을 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질로 판정하는 것을 특징으로 하는 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질 동정 방법.When a candidate substance and extracellular α-synuclein protein are contacted with microglia in an environment suitable for the two components to interact, and the rate of uptake and degradation of the α-synuclein protein in the microglia is increased. A method for identifying candidates for Parkinson's disease or Lewy body dementia (DLB) treatment, characterized in that the candidate substance is determined as a candidate for Parkinson's disease or Lewy body dementia (DLB) treatment.
  5. 제 4항에 있어서, 상기 후보물질은 항체인 것을 특징으로 하는 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질 동정 방법.5. The method of claim 4, wherein the candidate is an antibody. The method of identifying candidates for Parkinson's disease or Lewy Body Dementia (DLB).
  6. 제 4항에 있어서, 상기 미세아교세포는 Fcγ 수용체를 발현하는 것을 특징으로 하는 파킨슨 질환 또는 루이소체 치매(DLB) 치료제 후보물질 동정 방법.5. The method of claim 4, wherein the microglia express Fcγ receptors. 6.
PCT/KR2012/011024 2012-12-14 2012-12-17 Composition for treatment of diseases related to removal of extracellular alpha-synuclein and method for screening therapeutic agent for said diseases WO2014092227A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020120146518A KR20140077567A (en) 2012-12-14 2012-12-14 Composition for treating diseases related with clearance of extracellular alpah-Synuclein and method for screening the candidate thereof
KR10-2012-0146518 2012-12-14

Publications (1)

Publication Number Publication Date
WO2014092227A1 true WO2014092227A1 (en) 2014-06-19

Family

ID=50934503

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2012/011024 WO2014092227A1 (en) 2012-12-14 2012-12-17 Composition for treatment of diseases related to removal of extracellular alpha-synuclein and method for screening therapeutic agent for said diseases

Country Status (2)

Country Link
KR (1) KR20140077567A (en)
WO (1) WO2014092227A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101956768B1 (en) * 2016-12-30 2019-03-11 서울대학교산학협력단 Method for Large Scale Amplification of Alpha-synuclein Aggregates
KR101896182B1 (en) * 2017-06-15 2018-09-07 재단법인대구경북과학기술원 A method for suppressing alpha-Synuclein aggregates-mediated cellular toxicity

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006020581A2 (en) * 2004-08-09 2006-02-23 Elan Pharmaceuticals, Inc. Prevention and treatment of synucleinopathic and amyloidogenic disease
US20070232556A1 (en) * 2006-03-31 2007-10-04 Montine Thomas J Methods and compositions for the treatment of neurological diseases and disorders
KR20120090672A (en) * 2011-02-08 2012-08-17 건국대학교 산학협력단 A NOVEL ANTI-&alpha;-CYNUCLEIN MONOCLONAL ANTIBADY AND ELISA SYSTEM USING THE SAME

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006020581A2 (en) * 2004-08-09 2006-02-23 Elan Pharmaceuticals, Inc. Prevention and treatment of synucleinopathic and amyloidogenic disease
US20070232556A1 (en) * 2006-03-31 2007-10-04 Montine Thomas J Methods and compositions for the treatment of neurological diseases and disorders
KR20120090672A (en) * 2011-02-08 2012-08-17 건국대학교 산학협력단 A NOVEL ANTI-&alpha;-CYNUCLEIN MONOCLONAL ANTIBADY AND ELISA SYSTEM USING THE SAME

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FELLNER, LISA ET AL.: "Toll-like receptor 4 is required for a-synuclein dependent activation ofmicroglia and astroglia", GLIA, vol. 61, no. 3, 25 October 2012 (2012-10-25), pages 349 - 360 *
LEE, HE-JIN ET AL.: "Enzyme-linked immunosorbent assays for a-synuclein with species and multimeric state specificities", J. NEUROSCI. METHODS, vol. 199, no. 2, 31 May 2011 (2011-05-31), pages 249 - 257, XP028238899, DOI: doi:10.1016/j.jneumeth.2011.05.020 *

Also Published As

Publication number Publication date
KR20140077567A (en) 2014-06-24

Similar Documents

Publication Publication Date Title
US11542323B2 (en) Agents, uses and methods for the treatment of synucleinopathy
Bae et al. Antibody-aided clearance of extracellular α-synuclein prevents cell-to-cell aggregate transmission
US20200215209A1 (en) Antibodies and Vaccines for Use in Therapeutic and Diagnostic Methods for Alpha-Synuclein-Related Disorders
US20100260758A1 (en) In vivo modulation of neuronal transport
US11919947B2 (en) Antibody binding active α-synuclein
EP1735008A2 (en) Tetanus toxin fusion protein for visualization of active synapses
EA020979B1 (en) PEGYLATED Aβ Fab ANTIBODY FRAGMENTS
Huang et al. Naturally occurring autoantibodies against α-synuclein rescues memory and motor deficits and attenuates α-synuclein pathology in mouse model of Parkinson's disease
WO2016053610A1 (en) Antibodies against pathological forms of tdp-43 and uses thereof
WO2014092227A1 (en) Composition for treatment of diseases related to removal of extracellular alpha-synuclein and method for screening therapeutic agent for said diseases
US20200385461A1 (en) Fusion constructs and uses thereof
US6313089B1 (en) Complexes of apolipoprotein E and ciliary neurotrophic factor (CNTF) and methods of use
US9796778B1 (en) Antibodies against pathological forms of TDP-43 and uses thereof
US20230073000A1 (en) Improved Cell-Permeable Modified Parkin Recombinant Protein for Treatment of Neurodegenerative Diseases and Use Thereof
Clarke et al. TrkB agonist antibody delivery to the brain using a TfR1 specific BBB shuttle provides neuroprotection in a mouse model of Parkinson’s disease
US20070092449A1 (en) Methods for direct visualization of active synapses
EP3630828A1 (en) Ligands binding to prion protein for use in the treatment of synucleinopathies
US20240101981A1 (en) A recombinant harsa that crosses the blood brain barrier
WO2023068850A1 (en) Composition for preventing or treating ischemic stroke comprising inhibitor of apoptosis proteins
US20100081197A1 (en) In vivo modulation of neuronal transport
TW201632549A (en) Antibodies against pathological forms of TDP-43 and uses thereof
CN115531543A (en) Alpha-synuclein C-terminal as important target of anti-Parkinson&#39;s disease medicine
KR20180001608A (en) Lactoferrin-low molecular weight heparin nanocomplex and use thereof

Legal Events

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

Ref document number: 12890098

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: 12890098

Country of ref document: EP

Kind code of ref document: A1